1
|
Liao Y, Zhang Q, Shi Q, Liu P, Zhong P, Guo L, Huang Z, Peng Y, Liu W, Zhang S, Adorján I, Fukuzaki Y, Kawashita E, Zhang XQ, Ma N, Zhang X, Molnár Z, Shi L. Neuroserpin alleviates cerebral ischemia-reperfusion injury by suppressing ischemia-induced endoplasmic reticulum stress. Neural Regen Res 2026; 21:333-345. [PMID: 40489346 DOI: 10.4103/nrr.nrr-d-24-00044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202601000-00037/figure1/v/2025-06-09T151831Z/r/image-tiff Neuroserpin, a secreted protein that belongs to the serpin superfamily of serine protease inhibitors, is highly expressed in the central nervous system and plays multiple roles in brain development and pathology. As a natural inhibitor of recombinant tissue plasminogen activator, neuroserpin inhibits the increased activity of tissue plasminogen activator in ischemic conditions and extends the therapeutic windows of tissue plasminogen activator for brain ischemia. However, the neuroprotective mechanism of neuroserpin against ischemic stroke remains unclear. In this study, we used a mouse model of middle cerebral artery occlusion and oxygen-glucose deprivation/reperfusion-injured cortical neurons as in vivo and in vitro ischemia-reperfusion models, respectively. The models were used to investigate the neuroprotective effects of neuroserpin. Our findings revealed that endoplasmic reticulum stress was promptly triggered following ischemia, initially manifesting as the acute activation of endoplasmic reticulum stress transmembrane sensors and the suppression of protein synthesis, which was followed by a later apoptotic response. Notably, ischemic stroke markedly downregulated the expression of neuroserpin in cortical neurons. Exogenous neuroserpin reversed the activation of multiple endoplasmic reticulum stress signaling molecules, the reduction in protein synthesis, and the upregulation of apoptotic transcription factors. This led to a reduction in neuronal death induced by oxygen/glucose deprivation and reperfusion, as well as decreased cerebral infarction and neurological dysfunction in mice with middle cerebral artery occlusion. However, the neuroprotective effects of neuroserpin were markedly inhibited by endoplasmic reticulum stress activators thapsigargin and tunicamycin. Our findings demonstrate that neuroserpin exerts neuroprotective effects on ischemic stroke by suppressing endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Yumei Liao
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Qinghua Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Qiaoyun Shi
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Peng Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Peiyun Zhong
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Lingling Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Zijian Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Yinghui Peng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Wei Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute; Institute of Geriatric Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Shiqing Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - István Adorján
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Yumi Fukuzaki
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Eri Kawashita
- Department of Pathological Biochemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| | - Xiao-Qi Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Nan Ma
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
| | - Xiaoshen Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
- School of Nursing, Jinan University, Guangzhou, Guangdong Province, China
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Lei Shi
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong Province, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Thibaut MM, Roumain M, Piron E, Gillard J, Loriot A, Neyrinck AM, Rodriguez J, Massart I, Thissen JP, Huot JR, Pin F, Bonetto A, Delzenne NM, Muccioli GG, Bindels LB. The microbiota-derived bile acid taurodeoxycholic acid improves hepatic cholesterol levels in mice with cancer cachexia. Gut Microbes 2025; 17:2449586. [PMID: 39780051 PMCID: PMC11730681 DOI: 10.1080/19490976.2025.2449586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Alterations in bile acid profile and pathways contribute to hepatic inflammation in cancer cachexia, a syndrome worsening the prognosis of cancer patients. As the gut microbiota impinges on host metabolism through bile acids, the current study aimed to explore the functional contribution of gut microbial dysbiosis to bile acid dysmetabolism and associated disorders in cancer cachexia. Using three mouse models of cancer cachexia (the C26, MC38 and HCT116 models), we evidenced a reduction in the hepatic levels of several secondary bile acids, mainly taurodeoxycholic (TDCA). This reduction in hepatic TDCA occurred before the appearance of cachexia. Longitudinal analysis of the gut microbiota pinpointed an ASV, identified as Xylanibacter rodentium, as a bacterium potentially involved in the reduced production of TDCA. Coherently, stable isotope-based experiments highlighted a robust decrease in the microbial 7α-dehydroxylation (7α-DH) activity with no changes in the bile salt hydrolase (BSH) activity in cachectic mice. This approach also highlighted a reduced microbial 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 12α-hydroxysteroid dehydrogenase (12α-HSDH) activities in these mice. The contribution of the lower production of TDCA to cancer cachexia was explored in vitro and in vivo. In vitro, TDCA prevented myotube atrophy, whereas in vivo hepatic whole transcriptome analysis revealed that TDCA administration to cachectic mice improved the unfolded protein response and cholesterol homeostasis pathways. Coherently, TDCA administration reversed hepatic cholesterol accumulation in these mice. Altogether, this work highlights the contribution of the gut microbiota to bile acid dysmetabolism and the therapeutic interest of the secondary bile acid TDCA for hepatic cholesterol homeostasis in the context of cancer cachexia. Such discovery may prove instrumental in the understanding of other metabolic diseases characterized by microbial dysbiosis. More broadly, our work demonstrates the interest and relevance of microbial activity measurements using stable isotopes, an approach currently underused in the microbiome field.
Collapse
Affiliation(s)
- Morgane M. Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Martin Roumain
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Edwige Piron
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Justine Gillard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Axelle Loriot
- Computational Biology and Bioinformatics Unit (CBIO), de Duve Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle Massart
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Joshua R. Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Welbio Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
3
|
Kumar R, Gandham S, Bhaskar V, Praharaj MR, Maity HK, Sarkar U, Dey B. Transcriptomic insights into Mycobacterium orygis infection-associated pulmonary granulomas reveal multicellular immune networks and tuberculosis biomarkers in cattle. Vet Q 2025; 45:1-19. [PMID: 40432328 PMCID: PMC12120866 DOI: 10.1080/01652176.2025.2509503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/11/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Mycobacterium orygis, a member of the Mycobacterium tuberculosis complex (MTBC), has emerged as a significant contributor to tuberculosis (TB) in cattle, wildlife, and humans. However, understanding about its pathogenesis and severity is limited, compounded by the lack of reliable TB biomarkers in cattle. This study delves into the comparative pathology and transcriptomic landscape of pulmonary granulomas in cattle naturally infected with M. orygis, using high-throughput RNA sequencing. Histopathological analysis revealed extensive, multistage granulomatous, necrotic, and cavitary lesions, indicative of severe lung pathology induced by M. orygis. Transcriptomic profiling highlighted numerous differentially expressed genes and dysregulated pathways related to immune response modulation and extracellular matrix remodelling. Additionally, cell type enrichment analysis provided insights into the multicellularity of the granulomatous niche, emphasising complex cell-cell interactions within TB granulomas. Via comparative transcriptomics leveraging publicly available bovine and human TB omics datasets, 14 key immunomodulators (SOD2, IL1α/β, IL15, IL18, CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β, CCL8/MCP-2, CCL20/MIP-3α, CXCL2/MIP-2, CXCL10/IP-10, CXCL11, and IFN-γ) were identified as potential biomarkers for active TB in cattle. These findings significantly advance our understanding of M. orygis pathogenesis in bovine TB and highlight potential targets for the development of diagnostic tools for managing and controlling the disease.
Collapse
Affiliation(s)
- Rishi Kumar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Sripratyusha Gandham
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Vinay Bhaskar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
| | - Manas Ranjan Praharaj
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Hemanta Kumar Maity
- Department of Avian Sciences, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Uttam Sarkar
- Department of Animal Genetics and Breeding, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Bappaditya Dey
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| |
Collapse
|
4
|
Chen C, Ma Y, Gao Y, Ge H, Zhang X. Prognostic significance of neutrophil extracellular trap-related genes in childhood acute lymphoblastic leukemia: insights from multi-omics and in vitro experiment. Hematology 2025; 30:2452701. [PMID: 39829399 DOI: 10.1080/16078454.2025.2452701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND This study aimed to develop a prognostic model based on extracellular trap-related genes (NETRGs) for patients with cALL. METHODS Data from the TARGET-ALL-P2 and TARGET-ALL-P3 cohorts in the Genomic Data Commons database, the transcriptome dataset GSE26713, the single-cell transcriptome dataset GSE130116 from the Gene Expression Omnibus database and 306 NETRGs identified were analysed. Differentially expressed genes (DEGs) were identified from GSE26713 and differentially expressed NETRGs (DE-NETRGs) were obtained by overlapping DEGs with NETRGs. Functional analyses were conducted. Key feature genes were identified through univariate and least absolute shrinkage and selection operator (LASSO) regression. Prognostic genes were determined via multivariate Cox regression analysis, followed by the construction and validation of a risk model and nomogram. Additional analyses included immune profiling, drug sensitivity, functional differences, cell-type-specific expression, enrichment analysis and RT-qPCR. RESULTS A total of 1,270 DEGs were identified in GSE26713, of which 74 overlapped with NETRGs. Seven prognostic genes were identified using univariate, LASSO and multivariate Cox regression analyses. Survival analysis revealed lower survival rates in the high-risk group. Independent prognostic analysis identified risk scores and primary diagnosis as independent predictors of prognosis. Immune cell profiling showed significant differences in cell populations such as aDCs, eosinophils and Th2 cells between risk groups. Six cell subtypes were annotated, with prognostic genes predominantly expressed in myeloid cells. RT-qPCR revealed that PTAFR, FCGR2A, RETN and CAT were significantly downregulated, while TLR2 and S100A12 were upregulated in cALL. CONCLUSION TLR2, PTAFR, FCGR2A, RETN, S100A12 and CAT may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| | - Yu Ma
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| | - Yadai Gao
- Department of Pediatrics, Yinchuan Women and Children Healthcare Hospital, Yinchuan, People's Republic of China
| | - Huiqing Ge
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| | - Xiaochun Zhang
- Department of Pediatrics, Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, People's Republic of China
| |
Collapse
|
5
|
Wu IW, Liao YC, Tsai TH, Lin CH, Shen ZQ, Chan YH, Tu CW, Chou YJ, Lo CJ, Yeh CH, Chen CY, Pan HC, Hsu HJ, Lee CC, Cheng ML, Sheu WHH, Lai CC, Sytwu HK, Tsai TF. Machine-learning assisted discovery unveils novel interplay between gut microbiota and host metabolic disturbance in diabetic kidney disease. Gut Microbes 2025; 17:2473506. [PMID: 40050256 PMCID: PMC11901534 DOI: 10.1080/19490976.2025.2473506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/24/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Diabetic kidney disease (DKD) is a serious healthcare dilemma. Nonetheless, the interplay between the functional capacity of gut microbiota and their host remains elusive for DKD. This study aims to elucidate the functional capability of gut microbiota to affect kidney function of DKD patients. A total of 990 subjects were enrolled consisting of a control group (n = 455), a type 2 diabetes mellitus group (DM, n = 204), a DKD group (n = 182) and a chronic kidney disease group (CKD, n = 149). Full-length sequencing of 16S rRNA genes from stool DNA was conducted. Three findings are pinpointed. Firstly, new types of microbiota biomarkers have been created using a machine-learning (ML) method, namely relative abundance of a microbe, presence or absence of a microbe, and the hierarchy ratio between two different taxonomies. Four different panels of features were selected to be analyzed: (i) DM vs. Control, (ii) DKD vs. DM, (iii) DKD vs. CKD, and (iv) CKD vs. Control. These had accuracy rates between 0.72 and 0.78 and areas under curve between 0.79 and 0.86. Secondly, 13 gut microbiota biomarkers, which are strongly correlated with anthropometric, metabolic and/or renal indexes, concomitantly identified by the ML algorithm and the differential abundance method were highly discriminatory. Finally, the predicted functional capability of a DKD-specific biomarker, Gemmiger spp. is enriched in carbohydrate metabolism and branched-chain amino acid (BCAA) biosynthesis. Coincidentally, the circulating levels of various BCAAs (L-valine, L-leucine and L-isoleucine) and their precursor, L-glutamate, are significantly increased in DM and DKD patients, which suggests that, when hyperglycemia is present, there has been alterations in various interconnected pathways associated with glycolysis, pyruvate fermentation and BCAA biosynthesis. Our findings demonstrate that there is a link involving the gut-kidney axis in DKD patients. Furthermore, our findings highlight specific gut bacteria that can acts as useful biomarkers; these could have mechanistic and diagnostic implications.
Collapse
Affiliation(s)
- I-Wen Wu
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yu-Chieh Liao
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | | | - Chieh-Hua Lin
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Zhao-Qing Shen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Chih-Wei Tu
- Advanced Tech BU, Acer Inc, New Taipei City, Taiwan
| | - Yi-Ju Chou
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Hsiao Yeh
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Yu Chen
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Heng-Chih Pan
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Heng-Jung Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chin-Chan Lee
- Department of Nephrology, Chang Gung Memorial Hospital, Keelung, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Mei-Ling Cheng
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University (MLC), Taoyuan, Taiwan
| | - Wayne Huey-Herng Sheu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Chun Lai
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Department & Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
6
|
Sall I, Foxall R, Felth L, Maret S, Rosa Z, Gaur A, Calawa J, Pavlik N, Whistler JL, Whistler CA. Gut dysbiosis was inevitable, but tolerance was not: temporal responses of the murine microbiota that maintain its capacity for butyrate production correlate with sustained antinociception to chronic morphine. Gut Microbes 2025; 17:2446423. [PMID: 39800714 PMCID: PMC11730370 DOI: 10.1080/19490976.2024.2446423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/24/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
The therapeutic benefits of opioids are compromised by the development of analgesic tolerance, which necessitates higher dosing for pain management thereby increasing the liability for drug dependence and addiction. Rodent models indicate opposing roles of the gut microbiota in tolerance: morphine-induced gut dysbiosis exacerbates tolerance, whereas probiotics ameliorate tolerance. Not all individuals develop tolerance, which could be influenced by differences in microbiota, and yet no study design has capitalized upon this natural variation. We leveraged natural behavioral variation in a murine model of voluntary oral morphine self-administration to elucidate the mechanisms by which microbiota influences tolerance. Although all mice shared similar morphine-driven microbiota changes that largely masked informative associations with variability in tolerance, our high-resolution temporal analyses revealed a divergence in the progression of dysbiosis that best explained sustained antinociception. Mice that did not develop tolerance maintained a higher capacity for production of the short-chain fatty acid (SCFA) butyrate known to bolster intestinal barriers and promote neuronal homeostasis. Both fecal microbial transplantation (FMT) from donor mice that did not develop tolerance and dietary butyrate supplementation significantly reduced the development of tolerance independently of suppression of systemic inflammation. These findings could inform immediate therapies to extend the analgesic efficacy of opioids.
Collapse
Affiliation(s)
- Izabella Sall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Graduate program in Molecular and Evolutionary Systems Biology, University of New Hampshire, Durham, NH, USA
| | - Randi Foxall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Lindsey Felth
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Soren Maret
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Zachary Rosa
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Anirudh Gaur
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Jennifer Calawa
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Microbiology Graduate Program, University of New Hampshire, Durham, NH, USA
| | - Nadia Pavlik
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Jennifer L. Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| | - Cheryl A. Whistler
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
7
|
Chen B, Zhen L, Yang Z, Liu T, Yang S, Mu W, Xiao X, Chen J. miRNA-mRNA integrated analysis reveals candidate genes associated with salt stress response in Halophytic Sonneratia apetala. RNA Biol 2025; 22:1-13. [PMID: 40296366 PMCID: PMC12045576 DOI: 10.1080/15476286.2025.2496097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/20/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Sonneratia apetala is a pioneering species of mangrove plants, which has evolved various mechanisms to tolerate salt-stress due to their long-term exposure to a salinized environment as compared to the of terrestrial freshwater plants. However, limited attempt has been made to uncover the underlying molecular mechanism of their saline adaptation. Here, we integrated mRNA and microRNA (miRNA) sequencing to identify the genes and pathways that may be involved in salt stress-response in the roots of S. apetala. A comprehensive full‑length transcriptome containing 295,501 high‑quality unigenes was obtained by PacBio sequencing technology. Of these, 6,686 genes exhibited significantly differential accumulation after salt stress treatment (p < 0.001, Q < 0.01). They were mainly implicated in plant signal transduction and diverse metabolic pathways, such as those involving phenylpropanoid biosynthesis, plant-pathogen interaction and protein processing. Also, our results identified the regulatory interaction between miRNA-target counterparts during salt stress. Taken together, we present the first global overview of the transcriptome of S. apetala roots, and identify potentially important genes and pathways associated with salt tolerance for further investigation. This study is expected to deliver novel insights in understanding the regulatory mechanism in S. apetala response to salt stress.
Collapse
Affiliation(s)
- Beibei Chen
- Guangdong Engineering Technology Research Center of Tropical Crops High Efficient Production, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, PR, China
| | - Lishan Zhen
- Guangdong Engineering Technology Research Center of Tropical Crops High Efficient Production, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, PR, China
| | - Zhuanying Yang
- Guangdong Engineering Technology Research Center of Tropical Crops High Efficient Production, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, PR, China
| | - Tingting Liu
- Guangdong Engineering Technology Research Center of Tropical Crops High Efficient Production, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, PR, China
| | - Shaoxia Yang
- Guangdong Engineering Technology Research Center of Tropical Crops High Efficient Production, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, PR, China
| | - Wei Mu
- Guangdong Engineering Technology Research Center of Tropical Crops High Efficient Production, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, PR, China
| | - Xiao Xiao
- School of Chemistry and Environment, Guangdong Ocean University, Zhanjiang, PR, China
| | - Jinhui Chen
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University, Sanya, PR, China
| |
Collapse
|
8
|
Wang Z, Liu T, He K, Wang L, Ma X, Yang Z, Zhang Y, Zhao L. Knockdown of HGH1 in breast cancer cell lines can inhibit the viability, invasion and migration of tumor cells. Cell Adh Migr 2025; 19:1-14. [PMID: 39691959 DOI: 10.1080/19336918.2024.2442349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/19/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Research on the function of HGH1 in breast cancer remains lacking. METHODS TCGAand GEO (GSE45827) datasets investigated discrepancies in HGH1 expression in BC. An aggregate of 106 clinical samples were gathered through immunohistochemistry, KM curves were drawn for prognostic analysis, and the function of HGH1 of BC was predicted. Finally, the effects of HGH1 knockdown on MDA-MB-231 and MCF-7 BC cells were verified via CCK8, invasion, wound healing and colony formation assays. RESULTS HGH1 is highly expressed in BC and is linked to unfavorable prognosis. HGH1 overexpression is connected to keratinization and the cell cycle and is closely related to ER and PR expression and tumor stage in BC patients. Knocking down HGH1 in BC cells inhibited the viability, invasion and migration. CONCLUSION Knockdown of HGH1 in breast cancer cell lines can inhibit the viability, invasion and migration of tumor cells.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Taiyuan Liu
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, China
| | - Kang He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Longyun Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xiaoxuan Ma
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Zhaoyun Yang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yingchao Zhang
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
9
|
Oppenheimer M, Tao J, Moidunny S, Roy S. Anxiety-like behavior during protracted morphine withdrawal is driven by gut microbial dysbiosis and attenuated with probiotic treatment. Gut Microbes 2025; 17:2517838. [PMID: 40518557 PMCID: PMC12169037 DOI: 10.1080/19490976.2025.2517838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 05/12/2025] [Accepted: 06/03/2025] [Indexed: 06/18/2025] Open
Abstract
The development of anxiety during protracted opioid withdrawal heightens the risk of relapse into the cycle of addiction. Understanding the mechanisms driving anxiety during opioid withdrawal could facilitate the development of therapeutics to prevent negative affect and promote continued abstinence. Our lab has previously established the gut microbiome as a driver of various side effects of opioid use, including analgesic tolerance and somatic withdrawal symptoms. We therefore hypothesized that the gut microbiome contributes to the development of anxiety-like behavior during protracted opioid withdrawal. In this study, we first established a mouse model of protracted morphine withdrawal, characterized by anxiety-like behavior and gut microbial dysbiosis. Next, we used fecal microbiota transplantation (FMT) to show that gut dysbiosis alone is sufficient to induce anxiety-like behavior. We further demonstrated that probiotic therapy during morphine withdrawal attenuated the onset of anxiety-like behavior, highlighting its therapeutic potential. Lastly, we examined transcriptional changes in the amygdala of morphine-withdrawn mice treated with probiotics to explore mechanisms by which the gut-brain axis mediates anxiety-like behavior. Our results support the use of probiotics as a promising therapeutic strategy to prevent gut dysbiosis and associated anxiety during opioid withdrawal, with potential implications for improving treatment outcomes in opioid recovery programs.
Collapse
Affiliation(s)
- Mark Oppenheimer
- Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, USA
| | - Junyi Tao
- Department of Surgery, University of Miami Miller School of Medicine, Miami, USA
| | - Shamsudheen Moidunny
- Department of Surgery, University of Miami Miller School of Medicine, Miami, USA
| | - Sabita Roy
- Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
10
|
Hwang HG, Park JW, Lee HJ, Ko MY, Ka M, Lee YK, Choi J, In SA, Lee YE, Lee S, Kim MS, Kim JY. Akkermansia muciniphila reverses neuronal atrophy in Negr1 knockout mice with depression-like phenotypes. Gut Microbes 2025; 17:2508424. [PMID: 40388597 PMCID: PMC12091914 DOI: 10.1080/19490976.2025.2508424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/07/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025] Open
Abstract
Genetic predispositions can shape the gut microbiome, which in turn modulates host gene expression and impacts host physiology. The complex interplay between host genetics and the gut microbiome likely contributes to the development of neuropsychiatric disorders, yet the mechanisms behind these interactions remain largely unexplored. In this study, we investigated the gut microbiota in Negr1 knockout (KO) mice, which exhibit anxiety- and depression-like behaviors, as NEGR1 (neuronal growth regulator 1) is a cell adhesion molecule linked to neuronal development and neuropsychiatric disorders. Our findings show significant early-life alterations in the gut microbiota composition of Negr1 KO mice, most notably a marked reduction in Akkermansia spp. along with reduced dendritic arborization and spine density in the nucleus accumbens (NAc) and the dentate gyrus (DG) of the hippocampus. Remarkably, daily administration of an Akkermansia strain isolated from wild-type mice reversed the neuronal structural abnormalities and ameliorated anxiety- and depression-like behaviors in Negr1 KO mice. Transcriptomic profiling revealed upregulation of mitochondrial genome-encoded genes in the NAc and hippocampus of Negr1 KO mice, along with a predisposition toward a pro-inflammatory state in the colon of Negr1 KO mice. The Akkermansia supplementation downregulated these mitochondrial genes in the NAc and hippocampus and upregulated genes involved in T cell activation and immune homeostasis in the colon. These findings demonstrate a novel gene-microbiome interaction in the pathophysiology of Negr1 KO mice, positioning Akkermansia spp. as a key mediator that improves neuronal atrophy and modulates anxiety- and depression-like behaviors. Our study provides compelling evidence for bidirectional interactions between host genetics and the gut microbiome in modulating neuropsychiatric phenotypes, offering new insights for addressing genetically influenced mental disorders.
Collapse
Affiliation(s)
- Hee-Gon Hwang
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ji-Woo Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyo-Jin Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Moon Yi Ko
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Minhan Ka
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Jaeyoon Choi
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Su-A In
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ye-Eun Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Soojin Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Min-Soo Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Yoon Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
11
|
Barbaro MR, Bianco F, Cremon C, Marasco G, Bonomini F, Palombo M, Delprete C, Perez M, Espadaler-Mazo J, Stanghellini V, Guglielmetti S, Barbara G. Lactiplantibacillus plantarum (CECT7484 and CECT7485) and Pedioccoccus acidilactici (CECT7483) enhance actin cytoskeleton and CYP1A1 expression restoring epithelial permeability alterations induced by irritable bowel syndrome mediators. Gut Microbes 2025; 17:2452235. [PMID: 39817446 PMCID: PMC11740675 DOI: 10.1080/19490976.2025.2452235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a multifactorial condition with heterogeneous pathophysiology, including intestinal permeability alterations. The aim of the present study was to assess the ability of a probiotic blend (PB) consisting of two Lactiplantibacillus plantarum strains (CECT7484 and CECT7485) and one strain of Pediococcus acidilactici (CECT7483) to recover the permeability increase induced by mediators from IBS mucosal biopsies and to highlight the underlying molecular mechanisms. Twenty-one IBS patients diagnosed according to ROME IV criteria (11 IBS-D and 10 IBS-M) and 7 healthy controls were enrolled. Mucosal mediators spontaneously released by IBS and HC biopsies were collected and incubated with/without the PB (104 and 106 CFU/ml). Paracellular permeability was assessed by evaluating the amount of sulfonic-acid-conjugated to fluorescein passing through the Caco-2 monolayer. RNA was extracted from Caco-2 cells and used to perform qPCR analyses, to evaluate the expression of ZO-1 and β-actin, and RNAseq to evaluate the transcriptomic profile. Untargeted metabolomics was used to characterize metabolites produced by the PB. The PB significantly reduced paracellular permeability after 3 h of incubation. Both doses of the PB significantly recovered the increase in paracellular permeability induced by IBS mediators. qPCR analyses showed that both doses of the PB co-incubated with IBS mediators induced a significant increase in beta-actin expression compared to IBS mediators alone. Concerning IBS subtypes, the high dose of the PB recovered the increase of permeability induced by IBS-D mediators. Transcriptomic analyses, confirmed by qPCR, showed that the high dose of the PB significantly increased CYP1A1 compared to IBS mediators alone. The PB produced a high amount of indole-3-lactic acid. The PB recovers the permeability increase induced by IBS mediators inducing the up-regulation of β-actin. In addition, the PB up-regulates the expression of CYP1A1, known to be involved in the metabolism of xenobiotics, possibly through the production of the indole-3-lactic acid.
Collapse
Affiliation(s)
- Maria Raffaella Barbaro
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesca Bianco
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Cesare Cremon
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Giovanni Marasco
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesca Bonomini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Marta Palombo
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Cecilia Delprete
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Marta Perez
- AB-Biotics S.A (KANEKA Group), Barcelona, Spain
| | | | - Vincenzo Stanghellini
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Simone Guglielmetti
- Department of Biotechnology and Biosciences (BtBs), University of Milan Biococca, Milan, Italy
| | - Giovanni Barbara
- Gastroenterology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
12
|
Roesel R, Strati F, Basso C, Epistolio S, Spina P, Djordjevic J, Sorrenti E, Villa M, Cianfarani A, Mongelli F, Galafassi J, Popeskou SG, Facciotti F, Caprera C, Melle F, Majno-Hurst PE, Franzetti-Pellanda A, De Dosso S, Bonfiglio F, Frattini M, Christoforidis D, Iezzi G. Combined tumor-associated microbiome and immune gene expression profiling predict response to neoadjuvant chemo-radiotherapy in locally advanced rectal cancer. Oncoimmunology 2025; 14:2465015. [PMID: 39992705 PMCID: PMC11853554 DOI: 10.1080/2162402x.2025.2465015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Locally advanced rectal cancer (LARC) is treated with neoadjuvant chemo-radiotherapy (nCRT) followed by surgery. A minority of patients show complete response (CR) to nCRT and may avoid surgery and its functional consequences. Instead, most patients show non-complete response (non-CR) and may benefit from additional treatments to increase CR rates. Reliable predictive markers are lacking. Aim of this study was to identify novel signatures predicting nCRT responsiveness. We performed a combined analysis of tumor-associated microbiome and immune gene expression profiling of diagnostic biopsies from 70 patients undergoing nCRT followed by rectal resection, including 16 with CR and 54 with non-CR. Findings were validated by an independent cohort of 49 patients, including 7 with CR and 42 with non-CR. Intratumoral microbiota significantly differed between CR and non-CR groups at genus and species level. Colonization by bacterial species of Ruminococcus genera was consistently associated with CR, whereas abundance of Fusobacterium, Porhpyromonas, and Oscillibacter species predicted non-CR. Immune gene profiling revealed a panel of 59 differentially expressed genes and significant upregulation of IFN-gamma and -alpha response in patients with CR. Integrated microbiome and immune gene profiling analysis unraveled clustering of microbial taxa with each other and with immune cell-related genes and allowed the identification of a combined signature correctly identifying non-CRS in both cohorts. Thus, combined intratumoral microbiome-immune profiling improves the prediction of response to nCRT. Correct identification of unresponsive patients and of bacteria promoting responsiveness might lead to innovative therapeutic approaches based on gut microbiota pre-conditioning to increase nCRT effectiveness in LARC.
Collapse
Affiliation(s)
- Raffaello Roesel
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Francesco Strati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Camilla Basso
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Samantha Epistolio
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Paolo Spina
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Julija Djordjevic
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Elisa Sorrenti
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Martina Villa
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Agnese Cianfarani
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Francesco Mongelli
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Jacopo Galafassi
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Sotirios G. Popeskou
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Federica Facciotti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Cecilia Caprera
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Federica Melle
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Pietro Edoardo Majno-Hurst
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | | | - Sara De Dosso
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Ferdinando Bonfiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, Naples, Italy
- CEINGE Advanced Biotechnology Franco Salvatore, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Milo Frattini
- Laboratory of Molecular Pathology, Institute of Pathology, Locarno, Switzerland
| | - Dimitrios Christoforidis
- Department of Surgery, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Visceral Surgery, CHUV, University of Lausanne, Lausanne, Switzerland
| | - Giandomenica Iezzi
- Laboratory for Translational Surgical Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
13
|
Moore M, Whittington HD, Knickmeyer R, Azcarate-Peril MA, Bruno-Bárcena JM. Non-stochastic reassembly of a metabolically cohesive gut consortium shaped by N-acetyl-lactosamine-enriched fibers. Gut Microbes 2025; 17:2440120. [PMID: 39695352 DOI: 10.1080/19490976.2024.2440120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Diet is one of the main factors shaping the human microbiome, yet our understanding of how specific dietary components influence microbial consortia assembly and subsequent stability in response to press disturbances - such as increasing resource availability (feeding rate) - is still incomplete. This study explores the reproducible re-assembly, metabolic interplay, and compositional stability within microbial consortia derived from pooled stool samples of three healthy infants. Using a single-step packed-bed reactor (PBR) system, we assessed the reassembly and metabolic output of consortia exposed to lactose, glucose, galacto-oligosaccharides (GOS), and humanized GOS (hGOS). Our findings reveal that complex carbohydrates, especially those containing low inclusion (~1.25 gL-1) components present in human milk, such as N-acetyl-lactosamine (LacNAc), promote taxonomic, and metabolic stability under varying feeding rates, as shown by diversity metrics and network analysis. Targeted metabolomics highlighted distinct metabolic responses to different carbohydrates: GOS was linked to increased lactate, lactose to propionate, sucrose to butyrate, and CO2, and the introduction of bile salts with GOS or hGOS resulted in butyrate reduction and increased hydrogen production. This study validates the use of single-step PBRs for reliably studying microbial consortium stability and functionality in response to nutritional press disturbances, offering insights into the dietary modulation of microbial consortia and their ecological dynamics.
Collapse
Affiliation(s)
- Madison Moore
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Hunter D Whittington
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Rebecca Knickmeyer
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jose M Bruno-Bárcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
14
|
Spiegelhauer MR, Offersen SM, Mao X, Gambino M, Sandris Nielsen D, Nguyen DN, Brunse A. Protection against experimental necrotizing enterocolitis by fecal filtrate transfer requires an active donor virome. Gut Microbes 2025; 17:2486517. [PMID: 40207909 PMCID: PMC11988273 DOI: 10.1080/19490976.2025.2486517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Necrotizing enterocolitis (NEC) remains a frequent catastrophic disease in preterm infants, and fecal filtrate transfer (FFT) has emerged as a promising prophylactic therapy. This study explored the role of virome viability for the protective effect of FFT. Using ultraviolet (UV) irradiation, we established a viral inactivation protocol and administered FFT, UV-inactivated FFT (iFFT) or sterile saline orally to preterm piglets at risk for experimental NEC. The gut pathology and barrier properties were assessed, while the microbiome was explored by 16S rRNA amplicon and metavirome sequencing. Like in prior studies, FFT reduced NEC severity and intestinal inflammation, while these effects were absent in the iFFT group. Unexpectedly, piglets receiving FFT exhibited mild side effects in the form of early-onset diarrhea. The FFT also converged the gut colonization by increased viral heterogeneity and a reduced abundance of pathobionts like Clostridium perfringens and Escherichia. In contrast, the gut microbiome of iFFT recipients diverged from both FFT and the controls. These findings highlight the clear distinction between the ability of active and inactivate viromes to modulate gut microbiota and decrease pathology. The efficacy of FFT may be driven by active bacteriophages, and loss of virome activity could have consequences for the treatment efficacy.
Collapse
Affiliation(s)
- Malene Roed Spiegelhauer
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simone Margaard Offersen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiaotian Mao
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Michela Gambino
- Institute for Conservation, Royal Danish Academy, Copenhagen, Denmark
| | - Dennis Sandris Nielsen
- Section for Food Microbiology, Gut Health and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Brunse
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Ma L, Lin X, Xu M, Ke X, Liu D, Chen Q. Exploring the biological mechanisms of severe COVID-19 in the elderly: Insights from an aged mouse model. Virulence 2025; 16:2487671. [PMID: 40228062 PMCID: PMC12005417 DOI: 10.1080/21505594.2025.2487671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/04/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
The elderly population, who have increased susceptibility to severe outcomes, have been particularly impacted by the coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), leading to a global health crisis. However, definitive parameters or mechanisms underlying the severity of COVID-19 in elderly people remain unclear. Thus, this study seeks to elucidate the mechanism behind the increased vulnerability of elderly individuals to severe COVID-19. We employed an aged mouse model with a mouse-adapted SARS-CoV-2 strain to mimic the severe symptoms observed in elderly patients with COVID-19. Comprehensive analyses of the whole lung were performed using transcriptome and proteome sequencing, comparing data from aged and young mice. For transcriptome analysis, bulk RNA sequencing was conducted using an Illumina sequencing platform. Proteomic analysis was performed using mass spectrometry following protein extraction, digestion, and peptide labelling. We analysed the transcriptome and proteome profiles of young and aged mice and discovered that aged mice exhibited elevated baseline levels of inflammation and tissue damage repair. After SARS-CoV-2 infection, aged mice showed increased antiviral and inflammatory responses; however, these responses were weaker than those in young mice, with significant complement and coagulation cascade responses. In summary, our study demonstrates that the increased vulnerability of the elderly to severe COVID-19 may be attributed to an attenuated antiviral response and the overactivation of complement and coagulation cascades. Future research on antiviral and inflammatory responses is likely to yield treatments that reduce the severity of viral respiratory diseases in the elderly.
Collapse
Affiliation(s)
- Li Ma
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xian Lin
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Meng Xu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Xianliang Ke
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Di Liu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Quanjiao Chen
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
16
|
Kempf F, Drumo R, Chaussé AM, Menanteau P, Kubasova T, Roche S, Lalmanach AC, Guabiraba R, Chaumeil T, Larivière-Gauthier G, Caballero-Posadas I, Laroche B, Rychlík I, Virlogeux-Payant I, Velge P. The immune response modulated by inoculation of commensal bacteria at birth impacts the gut microbiota and prevents Salmonella colonization. Gut Microbes 2025; 17:2474151. [PMID: 40079593 PMCID: PMC11913379 DOI: 10.1080/19490976.2025.2474151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/17/2025] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
Super- and low-shedding phenomena have been observed in genetically homogeneous hosts infected by a single bacterial strain. To decipher the mechanisms underlying these phenotypes, we conducted an experiment with chicks infected with Salmonella Enteritidis in a non-sterile isolator, which prevents bacterial transmission between animals while allowing the development of the gut microbiota. We investigated the impact of four commensal bacteria called Mix4, inoculated at hatching, on chicken systemic immune response and intestinal microbiota composition and functions, before and after Salmonella infection. Our results revealed that these phenotypes were not linked to changes in cell invasion capacity of bacteria during infection. Mix4 inoculation had both short- and long-term effects on immune response and microbiota and promoted the low-shedder phenotype. Kinetic analysis revealed that Mix4 activated immune response from day 4, which modified the microbiota on day 6. This change promotes a more fermentative microbiota, using the aromatic compounds degradation pathway, which inhibited Salmonella colonization by day 11 and beyond. In contrast, control animals exhibited a delayed TNF-driven pro-inflammatory response and developed a microbiota using anaerobic respiration, which facilitates Salmonella colonization and growth. This strategy offers promising opportunities to strengthen the barrier effect against Salmonella and possibly other pathogens.
Collapse
Affiliation(s)
- Florent Kempf
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| | - Rosanna Drumo
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| | | | | | - Tereza Kubasova
- Department of Microbiology and antimicrobial resistance, Veterinary Research Institute, Brno, Czech Republic
| | - Sylvie Roche
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| | | | | | - Thierry Chaumeil
- Plate-Forme d’Infectiologie Expérimentale, INRAE, Nouzilly, France
| | | | | | | | - Ivan Rychlík
- Department of Microbiology and antimicrobial resistance, Veterinary Research Institute, Brno, Czech Republic
| | | | - Philippe Velge
- ISP, INRAE, Université François Rabelais de Tours, Nouzilly, France
| |
Collapse
|
17
|
Kong G, Li R, Huang W, Yang Y, Guan T, Liu J, Li W, Hsiang T, Xi P, Li M, Jiang Z. A RACK1 family protein regulates pathogenicity of Peronophythora litchii by acting as a scaffold for MAPK signal modules. Virulence 2025; 16:2503429. [PMID: 40356437 PMCID: PMC12077431 DOI: 10.1080/21505594.2025.2503429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 03/03/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Litchi downy blight caused by Peronophythora litchii is the most destructive disease of litchi (Litchi chinensis). RACK1 (Receptor for activated C kinase 1) is a group of scaffold proteins, mainly involved in the regulation of various signaling pathways by interacting with signal transduction proteins and affecting the activity of these proteins. In this study, a RACK1 homologue identified in P. litchii, and named PlRACK1. The protein was found to interact with the mitogen-activated protein kinases, PlMAPK1 and PlMAPK2. CRISPR/Cas9-mediated genome editing technology was used to knock out PlRACK1, and we found that it was involved in mycelial growth, cell wall integrity, ROS metabolism, laccase activity, and pathogenicity of P. litchii. PlMAPK1 interacted with RACK1, and they jointly regulated sporangiophore branching of P. litchii. Transcriptome analysis showed that P. litchii MAPK Phosphatase 1 (PlMKP1) and beta-glucoside (PlBglX) were regulated by PlRACK1, both of which were also required for the pathogenicity of P. litchii. As well, PlMKP1 also interacted with PlMAPK1 and PlMAPK2. These results provide insights into the direct interactions between RACK1, MAPKs, and MKP, and their functions in growth, development, and pathogenesis in a plant pathogenic oomycete.
Collapse
Affiliation(s)
- Guanghui Kong
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Rui Li
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Weixiong Huang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Yaowen Yang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Tianfang Guan
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Jinghan Liu
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Wen Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Agriculture, Guangxi University, Nanning, China
| | - Tom Hsiang
- School of Environmental Sciences, University of Guelph, Guelph, ON, Canada
| | - Pinggen Xi
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Minhui Li
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| | - Zide Jiang
- Guangdong Province Key Laboratory of Microbial Signals and Disease Control/National Key Laboratory of Green Pesticide, South China Agricultural University, Guangzhou, China
| |
Collapse
|
18
|
Pinto C, Widawski J, Zahalka S, Thaler B, Schuster LC, Lukowski SW, Ramírez F, Tirapu I. Cross-disease integration of single-cell RNA sequencing data from lung myeloid cells reveals TAM signature in in vitro model. Oncoimmunology 2025; 14:2502278. [PMID: 40448976 DOI: 10.1080/2162402x.2025.2502278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 04/22/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Advancements in single-cell RNA sequencing (scRNA-seq) have revealed the phenotypic and functional diversity of tumor-associated macrophages (TAMs), identifying specific populations that directly impact the antitumor response. However, despite the recognition of TAMs as promising therapeutic targets for cancer treatment, research is hindered by the lack of validated human preclinical models. Here, we applied scRNA-seq to a 3D human cell-based model comprising tumor cell line-derived spheroids, cancer-associated fibroblasts and primary monocytes, a setup widely used in immuno-oncology research. Integration of our in vitro data with publicly available patient-derived datasets showed that the macrophages in this model share phenotypic characteristics with the pro-angiogenic and pro-fibrotic SPP1+ TAM population recently found across multiple cancer types and inflammatory lung diseases. This population was linked to aspects of disease progression and associated with poor prognosis in several tumor indications, highlighting the need for relevant models enabling its study as an immunotherapy target. Our research validates the use of a 3D human cell-based culture as a more in vivo-relevant model and enables the preclinical testing of novel macrophage-targeting drugs in a human disease-relevant setup.
Collapse
Affiliation(s)
- Catarina Pinto
- Oncology Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Jakub Widawski
- Computational Innovation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Sophie Zahalka
- Oncology Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Barbara Thaler
- Oncology Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Linda C Schuster
- Oncology Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Samuel W Lukowski
- Oncology Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Fidel Ramírez
- Computational Innovation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Iñigo Tirapu
- Oncology Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|
19
|
Xu B, Liu Y, Chen G, Jiang P, Qu Y, Wang M, Kao X. Genome-wide analysis of abnormal splicing regulators and alternative splicing involved in immune regulation in systemic lupus erythematosus. Autoimmunity 2025; 58:2448463. [PMID: 39743791 DOI: 10.1080/08916934.2024.2448463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with complex clinical manifestations and no current cure. Alternative splicing (AS) plays a key role in SLE by regulating immune-related genes, but its genome-wide regulatory mechanisms remain unclear. To investigate the involvement of abnormal splicing regulators and AS events in the immune regulation of SLE. Transcriptome data from the SLE dataset GSE162828 were analyzed for differential gene expression and AS events using bioinformatics tools. Immune infiltration analysis was conducted with CIBERSORT, and co-expression of key splicing factors (SFs) and AS events was assessed using SUVA software. A total of 5144 differentially expressed genes and 73 SFs were identified. Significant immune cell differences were observed between SLE and controls, highlighting SFs such as HNRNPDL, RBM47, TIA1, SSB, and DHX15. Eighty-three AS events were identified, with IRF9 and PTPRC emerging as key regulatory events linked to SLE. Dysregulated SFs influence AS in immune-related genes, affecting immune cell composition and SLE progression. These findings offer potential new therapeutic targets for modulating the immune microenvironment in SLE.
Collapse
Affiliation(s)
- Bing Xu
- Department of Rheumatology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Liu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guangfeng Chen
- Department of Geriatric Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Qu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengjie Wang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiliang Kao
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
20
|
Kaartinen L, Jääskeläinen T, Sliz E, Yazgeldi Gunaydin G, Wedenoja S, Katayama S, Kajantie E, Rinne V, Heinonen S, Kere J, Merikallio H, Hannele Laivuori submitted on behalf of FINNPEC group, Sliz E, submitted on behalf of FinnGen group, Laivuori H, Hukkanen J. Role of oxysterol 4β-hydroxycholesterol and liver X receptor alleles in pre-eclampsia. Ann Med 2025; 57:2495763. [PMID: 40298034 PMCID: PMC12042236 DOI: 10.1080/07853890.2025.2495763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/25/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Liver X receptors (LXRs) are expressed in placenta and may be associated with pre-eclampsia (PE). Oxysterols act as agonists for LXRs. We recently proposed a new blood pressure-regulating circuit with oxysterol 4β-hydroxycholesterol (4βHC) acting as a hypotensive factor via LXRs. MATERIALS AND METHODS This study investigated the association between maternal plasma 4βHC, blood pressure (BP) indices, placental expression of LXR target genes, and patient characteristics using data from the Finnish Genetics of Pre-Eclampsia Consortium (FINNPEC) cohort. Plasma samples of 144 women with PE and 38 healthy pregnant controls as well as 44 PE and 40 control placental samples were available. In addition, genetic data from the FinnGen project was utilized to explore the associations of LXR alleles with PE and pregnancy hypertension. RESULTS There were no significant associations between 4βHC and BP or maternal and perinatal characteristics in FINNPEC cohort. However, plasma 4βHC was inversely correlated with the maternal body mass index. There were no associations with the genetic variants of LXRs with PE in FinnGen. LXR target genes APOD, SCARB1, TGM2, and LPCAT3 were expressed differently between PE and normal pregnancies in placental samples of FINNPEC. CONCLUSIONS Our results demonstrate that plasma 4βHC and genetic LXR variants do not play a major role in PE and BP regulation during pregnancy. However, key LXR target genes involved in lipid metabolism were expressed differently in normal and PE pregnancies. Further research is needed to understand the complexities of oxysterols, LXRs, and their potential contributions to placental function and pregnancy outcomes.
Collapse
Affiliation(s)
- Lassi Kaartinen
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tiina Jääskeläinen
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
| | - Eeva Sliz
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Gamze Yazgeldi Gunaydin
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Satu Wedenoja
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Shintaro Katayama
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Eero Kajantie
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Research unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Seppo Heinonen
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Juha Kere
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Heta Merikallio
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Hannele Laivuori submitted on behalf of FINNPEC group
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
- Research Unit of Population Health, University of Oulu, Oulu, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Admescope (Symeres Finland Ltd), Oulu, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, The Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Eeva Sliz
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
- Research Unit of Population Health, University of Oulu, Oulu, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Admescope (Symeres Finland Ltd), Oulu, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, The Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - submitted on behalf of FinnGen group
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland
- Research Unit of Population Health, University of Oulu, Oulu, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki and Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Admescope (Symeres Finland Ltd), Oulu, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, The Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, The Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Janne Hukkanen
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
21
|
Chowdhury SR, Shilpi A, Felsenfeld G. RNA Pol-II transcripts in nucleolar associated domains of cancer cell nucleoli. Nucleus 2025; 16:2468597. [PMID: 39987497 PMCID: PMC11849958 DOI: 10.1080/19491034.2025.2468597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 01/03/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
We performed a comparative study of the non-ribosomal gene content of nucleoli from seven cancer cell lines, using identical methods of purification and analysis. We identified unique chromosomal domains associated with the nucleolus (NADs) and genes within these domains (NAGs). Four cell lines have relatively few NAGs, which appears mostly transcriptionally inactive, consistent with literature. The remaining three lines formed a separate group with nucleoli with unique features and NADS. They constitute larger number of common NAGs, marked by ATAC-seq and having accessible promoters, with histone markers for transcriptional activity and detectable RNA Pol II bound at their promoters. The transcripts of these genes are almost entirely exported from the nucleolus. These results indicate that RNA Pol II dependent transcription in NADs can vary widely in different cell types, presumably dependent on the cell's developmental stage. Nucleolus-associated genes are likely to be distinguished marks reflecting the cell's metabolism.
Collapse
Affiliation(s)
- Soumya Roy Chowdhury
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases
| | - Arunima Shilpi
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases
| | | |
Collapse
|
22
|
Khan W, Kanwar S, Mannan MM, Kabir F, Iqbal N, Nadeem Rajab Ali M, Zia SR, Mian S, Aziz F, Muneer S, Kalam A, Hussain A, Javed I, Qazi MF, Khalid J, Nisar MI, Jehan F. Identification of differentially expressed non-coding RNAs in the plasma of women with preterm birth. RNA Biol 2025; 22:1-8. [PMID: 39804675 PMCID: PMC11730358 DOI: 10.1080/15476286.2024.2449278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
This study aimed to identify differentially expressed non-coding RNAs (ncRNAs) associated with preterm birth (PTB) and determine biological pathways being influenced in the context of PTB. We processed cell-free RNA sequencing data and identified seventeen differentially expressed (DE) ncRNAs that could be involved in the onset of PTB. Per the validation via customized RT-qPCR, the recorded variations in expressions of eleven ncRNAs were concordant with the in-silico analyses. The results of this study provide insights into the role of DE ncRNAs and their impact on pregnancy-related biological pathways that could lead to PTB. Further studies are required to elucidate the precise mechanisms by which these DE ncRNAs contribute to adverse pregnancy outcomes (APOs) and their potential as diagnostic biomarkers.
Collapse
Affiliation(s)
- Waqasuddin Khan
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Samiah Kanwar
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Mohammad Mohsin Mannan
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Furqan Kabir
- Infectious Diseases Research Lab (IDRL), Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Naveed Iqbal
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Mehdia Nadeem Rajab Ali
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Syeda Rehana Zia
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Sharmeen Mian
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Fatima Aziz
- Infectious Diseases Research Lab (IDRL), Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Sahrish Muneer
- Infectious Diseases Research Lab (IDRL), Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Adil Kalam
- Infectious Diseases Research Lab (IDRL), Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Akram Hussain
- Infectious Diseases Research Lab (IDRL), Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Iqra Javed
- Infectious Diseases Research Lab (IDRL), Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Muhammad Farrukh Qazi
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Javairia Khalid
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Muhammad Imran Nisar
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Fyezah Jehan
- Biorepository and Omics Research Group, Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| |
Collapse
|
23
|
Danev N, Harman RM, Sipka AS, Oliveira L, Huntimer L, Van de Walle GR. The secretomes of bovine mammary epithelial cell subpopulations differentially modulate macrophage function. Vet Q 2025; 45:1-14. [PMID: 39921381 PMCID: PMC11809179 DOI: 10.1080/01652176.2025.2463338] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Bovine mammosphere-derived epithelial cell (MDEC) cultures are heterogeneous and enriched for stem and progenitor cells. We previously reported that the bovine MDEC secretome, comprised of all bioactive factors secreted by the cells, displays regenerative properties, exerts antimicrobial effects, and modulates neutrophil activity, positioning it as a promising non-antibiotic biologic therapy for infectious diseases important to the dairy industry, like mastitis. Mastitis is defined as inflammation of the udder, and it is typically caused by bacterial infection. The effect of the MDEC secretome on macrophages, a first line of defense against bacterial infections in the udder, is unknown and could impact the utility of the secretome as a therapy for mastitis. To address this, we isolated bovine monocytes from peripheral blood and maintained them as an unpolarized (M0) population or polarized them into M1 or M2 phenotypes. Macrophages cultured with the secretome of bovine MDECs were assessed for their ability to phagocytose labeled bacterial particles and accumulate reactive oxygen species (ROS). We used single-cell RNA sequencing (scRNA-seq) and fluorescence-activated cell sorting (FACS) to isolate a subpopulation of MDECs that exert enhanced effects on macrophages. We found that the secretome of MDECs that do not express cluster of differentiation (CD) 73, a cell surface enzyme used as a marker for mesenchymal stromal cells, most strongly increased macrophage phagocytosis and ROS accumulation. These findings will help optimize the generation of the bovine MDEC secretome as a suitable treatment option for mastitis.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Rebecca M. Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Anja S. Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- The Royal (Dick) School of Veterinary Studies and the Roslin Institute, University of Edinburgh, Midlothian, Scotland
| |
Collapse
|
24
|
You H, Yang B, Liu H, Wu W, Yu F, Lin N, Yang W, Hu B, Liu Y, Zou H, Hao S, Xiao Y, Xu T, Jiang Y. Unravelling distinct patterns of metagenomic surveillance and respiratory microbiota between two P1 genotypes of Mycoplasma pneumoniae. Emerg Microbes Infect 2025; 14:2449087. [PMID: 39760260 PMCID: PMC11730683 DOI: 10.1080/22221751.2024.2449087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 12/04/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
To unravel distinct patterns of metagenomic surveillance and respiratory microbiota between Mycoplasma pneumoniae (M. pneumoniae) P1-1 and P1-2 and to explore the impact of the COVID-19 pandemic on epidemiological features, we conducted a multicentre retrospective study which spanned 90,886 pneumonia patients, among which 3164 cases M. pneumoniae were identified. Our findings revealed a concurrent outbreak of M. pneumoniae, with the positivity rate rising sharply to 9.62% from July 2023, compared to the 0.16% to 4.06% positivity rate observed during the 2020-2022 COVID-19 pandemic. P1-1 had a higher odds ratio of co-detecting opportunistic pathogens. However, no significant differences were observed in the co-detection odds ratio between children and other age groups in P1-2. This study is the first to demonstrate differences in relative abundance, diversity of respiratory microbiota and co-detection rate of opportunistic pathogen between M. pneumoniae P1-1 and P1-2. Through bronchoalveolar lavage (BAL) metagenomic and host transcriptomic analyses, we identified variations in co-detection rates of M. pneumoniae P1-1 genotype with opportunistic pathogens like S. pneumoniae, alterations in respiratory microbiota composition, lung inflammation, and disruption of ciliary function. Consistent with the results of host transcriptome, we found that P1-1 infections were associated with significantly higher rates of requiring respiratory support and mechanical ventilation compared to P1-2 infections (Fisher's exact test, p-value = 0.035/0.004). Our study provides preliminary evidence of clinical severity between M. pneumoniae strains, underscoring the need for ongoing research and development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Hailong You
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Bin Yang
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong, People’s Republic of China
| | - Huifang Liu
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong, People’s Republic of China
| | - Wencai Wu
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong, People’s Republic of China
| | - Fei Yu
- Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Nan Lin
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - WenJiao Yang
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong, People’s Republic of China
| | - Bingxue Hu
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong, People’s Republic of China
| | - Yong Liu
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Hongyan Zou
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Sijia Hao
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yunping Xiao
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Teng Xu
- Center for Infectious Diseases, Vision Medicals Co., Ltd, Guangzhou, Guangdong, People’s Republic of China
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
25
|
Zhang Y, Castro-Mejía JL, Deng L, Shah SA, Thorsen J, Leal Rodríguez C, Jessen LE, Dion MB, Chawes B, Bønnelykke K, Sørensen SJ, Bisgaard H, Moineau S, Petit MA, Stokholm J, Nielsen DS. The influence of early life exposures on the infant gut virome. Gut Microbes 2025; 17:2501194. [PMID: 40396485 PMCID: PMC12101590 DOI: 10.1080/19490976.2025.2501194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/21/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025] Open
Abstract
The factors influencing the establishment of the gut bacterial community in early life are fairly well studied. However, the factors shaping the infant gut virome remain elusive. Interestingly, early life gut virome imbalances have recently been linked with increased risk of developing diseases like type 1 diabetes and asthma. We utilized the deeply phenotyped COPSAC2010 cohort to investigate how environmental factors influence the gut virome at one year age. We demonstrate that the presence of older siblings as well as residential location (urban or rural) had the strongest impact on gut virome composition at 1 year of age. A total of 16,118 species-level clustered viral representative contigs (here termed viral Operational Taxonomic Units - vOTUs) were identified and of these 2105 vOTUs varied in abundance with environmental exposures. Of these vOTUs 94.1% were phages mainly predicted to infect Bacteroidaceae, Prevotellaceae, and Ruminococcaceae. Strong co-abundance of phages and their bacterial hosts was confirmed underlining the predicted phage-host connections. Furthermore, we found some gut viruses affected by environmental factors encode enzymes involved in the utilization and degradation of major dietary components, potentially affecting infant health by influencing the bacterial host metabolic capacity. These findings provide a valuable insights for understanding the early life factors that predispose to autoimmune and metabolic disorders.
Collapse
Affiliation(s)
- Yichang Zhang
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | | | - Ling Deng
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Shiraz A. Shah
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cristina Leal Rodríguez
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Leon E. Jessen
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, DTU, Lyngby, Denmark
| | - Moïra B. Dion
- Département de Biochimie, de Microbiologie, et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, QC, Canada
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC, Canada
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Søren J. Sørensen
- Department of Biology, University of Copenhagen, Frederiksberg, Denmark
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Sylvain Moineau
- Département de Biochimie, de Microbiologie, et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, QC, Canada
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC, Canada
- Félix d’Hérelle Reference Center for Bacterial Viruses, Faculté de médecine dentaire, Université Laval, Québec, QC, Canada
| | - Marie-Agnès Petit
- Université Paris-Saclay, INRAE, AgroParis Tech, Micalis Institute, Jouy-en-Josas, France
| | - Jakob Stokholm
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Dennis S. Nielsen
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
26
|
Osborne N, Rupani A, Makarov V, Chan TA, Srivastava RM. Avelumab induces greater Fc-Fc receptor-dependent natural killer cell activation and dendritic cell crosstalk compared to durvalumab. Oncoimmunology 2025; 14:2494995. [PMID: 40311014 PMCID: PMC12051578 DOI: 10.1080/2162402x.2025.2494995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Several FDA-approved anti-PD-L1 (programmed cell death ligand-1) monoclonal antibodies (mAbs) are used to treat cancer. While these mAbs primarily target and intercept PD-L1:PD-1 inhibitory signaling in T-cells, the Fc-domains of these mAbs are distinct, and the unique cellular cascades triggered by differing Fc-domains of PD-L1 mAbs have not been directly investigated. In this study, we compared the innate immune effects of two widely used anti-PD-L1 IgG1 mAbs which bear distinct Fc-domains, avelumab (native-Fc) and durvalumab (mutated-Fc), using two-cell and three-cell co-culture systems containing Natural Killer cells (NK-cells), dendritic cells (DCs) and various tumor cell lines of multiple cancer origins. We show a robust enhancement in NK-cell effector function, DC maturation, reciprocal NK:DC crosstalk and DC editing that is unique to avelumab treatment using multiple functional immune assays. By transcriptomic analysis, we show for the first time pivotal differences in gene sets involved in NK-cell effector function, DC maturation, immunoregulatory interactions, and cytokine production between innate immune cells treated with avelumab versus durvalumab. Furthermore, we report several previously unknown Fc-receptor-associated biological pathways uniquely triggered by avelumab. Our findings elucidate novel mechanisms of Fc-dependent actions of PD-L1 mAbs which may inform their use in future clinical trials.
Collapse
MESH Headings
- Humans
- Dendritic Cells/immunology
- Dendritic Cells/drug effects
- Dendritic Cells/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/metabolism
- Antibodies, Monoclonal, Humanized/pharmacology
- Receptors, Fc/metabolism
- Receptors, Fc/immunology
- Cell Line, Tumor
- Coculture Techniques
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Antibodies, Monoclonal/pharmacology
- Cell Communication/drug effects
- Cell Communication/immunology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- Neoplasms/immunology
- Neoplasms/drug therapy
- Antineoplastic Agents, Immunological/pharmacology
Collapse
Affiliation(s)
- Nicole Osborne
- Discovery Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Amit Rupani
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Timothy A. Chan
- Discovery Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Raghvendra M. Srivastava
- Discovery Laboratory, Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
27
|
Chen H, Cao S, Zhou Y, Wang T, Jiao Y, Tan Y, Wu Y, Ren Y, Song Y, Zhang JR, Du Z, Yang R. Molecular turn in Yersinia pestis pathogenesis: implications of the gppA frameshift for bacterial survival in human macrophage. Emerg Microbes Infect 2025; 14:2467778. [PMID: 39945756 PMCID: PMC11878169 DOI: 10.1080/22221751.2025.2467778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/04/2025]
Abstract
Yersinia pestis, the etiological agent of the devastating plague, has caused three pandemics in human history. While known for its fatality, it has long been intriguing that biovar microtus strains are highly attenuated to humans. The survival and replication within macrophages are critical in the early stages of the Y. pestis lifestyle within warm-blooded hosts. Here, we demonstrate that a frameshift truncation of gppA, a gene encoding the phosphohydrolase GppA that responsible for the conversion of stringent response alarmone pppGpp to ppGpp, significantly promotes Y. pestis to survive inside human macrophages. This frameshift mutation of gppA is present in all the evolutionary branches formed by the modern Y. pestis strains responsible for the plague pandemics, while the relative ancient microtus strains express a functional GppA showing high activity in catalyzing pppGpp to ppGpp conversion. This adaptive evolution potentially explains why microtus Y. pestis strains exhibit attenuated virulence in humans in contrast to the lethal pathogenicity of non-microtus strains. Transcriptome analysis suggests that the disturbed balance of the ratio of ppGpp to pppGpp caused by GppA inactivation results in an upregulation of genes involved in the synthesis of branched-chain amino acids, which are essential for bacterial growth. This enhanced survival ability within macrophages could be a key factor for the virulence of Y. pestis towards humans. Our work sheds light on the molecular mechanisms behind Y. pestis host-specific pathogenicity, offering significant implications for enhancing our ability to predict and counteract the emergence of new infectious diseases.
Collapse
Affiliation(s)
- Hongyan Chen
- Center for Infection Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, People’s Republic of China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Shiyang Cao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Yazhou Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Tong Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Yang Jiao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Yafang Tan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Yarong Wu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Yifan Ren
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Yajun Song
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Jing-Ren Zhang
- Center for Infection Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, People’s Republic of China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, People’s Republic of China
| | - Zongmin Du
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
28
|
Gutiérrez-Larruscain D, Krüger M, Abeyawardana OAJ, Belz C, Dobrev PI, Vaňková R, Eliášová K, Vondráková Z, Juříček M, Štorchová H. Contrasting gene expression patterns during floral induction in two Chenopodium ficifolium genotypes reveal putative flowering regulators. PLANT SIGNALING & BEHAVIOR 2025; 20:2486083. [PMID: 40184219 PMCID: PMC11980483 DOI: 10.1080/15592324.2025.2486083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 04/06/2025]
Abstract
Chenopodium ficifolium is a close diploid relative of the tetraploid crop Chenopodium quinoa. Owing to its reproducible germination and seedling development, it becomes a promising model for studying floral induction, providing a basis for the comparison with C. quinoa. Two C. ficifolium genotypes differ in photoperiodic requirement: C. ficifolium 283 accelerates flowering under long days, whereas C. ficifolium 459 flowers earlier under short days. This study conducted a comprehensive transcriptomic and hormonomic analysis of floral induction in the long-day C. ficifolium 283 and compared the findings to previous experiments with the short-day C. ficifolium. Phytohormone concentrations and gene expression profiles during floral induction were largely similar between the two genotypes. However, a subset of genes exhibited contrasting expression patterns, aligning with the genotypes' differing photoperiodic requirements. These genes, predominantly homologs of flowering-related genes in Arabidopsis thaliana, were activated under long days in C. ficifolium 283 and under short days in C. ficifolium 459. Notably, the contrasting expression of the FLOWERING LOCUS T-LIKE 2-1 gene, which was previously shown to induce precocious flowering in A. thaliana, confirmed its role as a floral activator, despite its low expression levels.
Collapse
Affiliation(s)
| | - Manuela Krüger
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | | | - Claudia Belz
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Petre I. Dobrev
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Radomíra Vaňková
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Kateřina Eliášová
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Vondráková
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Miloslav Juříček
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Helena Štorchová
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
29
|
Li H, Liu H, Wu H, Guo C, Zuo W, Zheng Y, Deng X, Xu J, Wang Y, Wang Z, Lu B, Hou B, Cao B. Reading of human acute immune dynamics in omicron SARS-CoV-2 breakthrough infection. Emerg Microbes Infect 2025; 14:2494705. [PMID: 40231451 PMCID: PMC12064115 DOI: 10.1080/22221751.2025.2494705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 04/16/2025]
Abstract
The dynamics of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infections remain unclear, particularly when compared to responses in naive individuals. In this longitudinal prospective cohort study, 13 participants were recruited. Peripheral blood samples were collected every other day until day 7 after symptom onset. Transcriptome sequencing, single-cell sequencing, T-cell receptor (TCR) sequencing, B-cell receptor (BCR) sequencing, Olink proteomics, and antigen-antibody binding experiments were then performed. During the incubation periods of breakthrough infections, peripheral blood exhibited type 2 cytokine response, which shifted to type 1 cytokine response upon symptom onset. Plasma cytokine levels of C-X-C motif chemokine ligand 10, monocyte chemoattractant protein-1, interferon-γ, and interleukin-6 show larger changes in breakthrough infections than naïve infections. The inflammatory response in breakthrough infections rapidly subsided, returning to homeostasis by day 5 after symptom onset. Notably, the levels of monocyte-derived S100A8/A9, previously considered a marker of severe disease, physiologically significantly increased in the early stages of mild cases and persisted until day 7, suggesting a specific biological function. Longitudinal tracking also revealed that antibodies anti-Receptor Binding Domain (anti-RBD) in breakthrough infections significantly increased by day 7 after symptom onset, whereas cytotoxic T lymphocytes appeared by day 5. This study presents a reference for interpreting the immunological response to breakthrough infectious disease in humans.
Collapse
Affiliation(s)
- Haibo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
| | - Hongyu Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Hongping Wu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Chang Guo
- Changping National Laboratory (CPNL), Beijing, People’s Republic of China
| | - Wenting Zuo
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Ying Zheng
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyan Deng
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, People’s Republic of China
| | - Jiuyang Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yeming Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Binghuai Lu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Baidong Hou
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Bin Cao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| |
Collapse
|
30
|
Roberti MP, Charoentong P, Lyu Y, Meyer M, Eichmüller SB, Schmidt P, Momburg F, Cetin M, Hartmann F, Valous NA, Stenzinger A, Michel L, Lichter P, Schneeweiss A, Thewes V, Fremd C, Zörnig I, Jäger D. Isolation of a tumor neoantigen specific CD8+ TCR from a skin biopsy of a vaccination site. Oncoimmunology 2025; 14:2457793. [PMID: 39902862 PMCID: PMC11796541 DOI: 10.1080/2162402x.2025.2457793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
T cells that recognize tumor-specific mutations are crucial for cancer immunosurveillance and in adoptive transfer of TILs or transgenic-TCR T cell products. However, their challenging identification and isolation limits their use in clinical practice. Therefore, novel approaches to isolate tumor-specific T cells are needed. Here, we report the isolation of neoantigen-specific CD8+ T cells from a vaccination site of a metastatic breast cancer patient who received a personalized vaccine. Based on the somatic mutations, potential MHC binding epitopes were predicted, of which 17 were selected to generate a peptide vaccine. Cutaneous biopsies were processed after the fifth vaccination cycle to obtain infiltrating lymphocytes from the vaccination site (VILs). IFNγ ELISpot revealed reactivity to four peptides used in the vaccine. Reactive T cells from VILs were non-overlapping with those detected in the blood and the tumor-microenvironment. ScTCR Seq analysis revealed the presence of a clonotype in VILs that further expanded after a round of in vitro stimulation and validated to be specific against a private mutation, namely NCOR1L1475R, presented in the context of HLA-B * 07:02, with no reactivity to the wild-type peptide. Our study shows, for the first time, that tumor mutation - specific T cells are generated at high frequencies in the vaccination site and can be isolated with standard methods for TCR screening. The easy and safe accessibility of skin biopsies overcomes the major hurdles of current TCR screening approaches and present exciting opportunities for the development of innovative immunotherapeutic strategies.
Collapse
Affiliation(s)
- Maria Paula Roberti
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
| | - Pornpimol Charoentong
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | - Yanhong Lyu
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | - Marten Meyer
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B. Eichmüller
- GMP and T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Schmidt
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- GMP and T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Momburg
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miray Cetin
- Systems Immunology and Single Cell Biology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Felix Hartmann
- Systems Immunology and Single Cell Biology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Core Center Heidelberg, Heidelberg, Germany
| | - Nektarios A. Valous
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | | | - Laura Michel
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Schneeweiss
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Thewes
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carlo Fremd
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Inka Zörnig
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
| | - Dirk Jäger
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
| |
Collapse
|
31
|
Enssle S, Sax A, May P, El Khawanky N, Soliman N, Perl M, Enssle JC, Krey K, Ruland J, Pichlmair A, Bassermann F, Poeck H, Heidegger S. Gasdermin E links tumor cell-intrinsic nucleic acid signaling to proinflammatory cell death for successful checkpoint inhibitor cancer immunotherapy. Oncoimmunology 2025; 14:2504244. [PMID: 40366863 PMCID: PMC12080277 DOI: 10.1080/2162402x.2025.2504244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
Durable clinical responses to immune checkpoint inhibitors (ICI) are limited to a minority of patients, and molecular pathways that modulate their efficacy remain incompletely defined. We have recently shown that activation of the innate RNA-sensing receptor RIG-I and associated apoptotic tumor cell death can facilitate tumor immunosurveillance and -therapy, but the mechanism that drives its immunogenicity remained unclear. We here show that intratumoral activity of the pore-forming protein gasdermin E (GSDME) links active RIG-I signaling and apoptotic cell death in tumor cells to inflammatory pyroptosis. Activation of tumor-intrinsic RIG‑I triggered cleavage of GSDME, pore formation, loss of cell membrane integrity and leakage of cytosolic components from dying tumor cells. Tumor antigen cross-presentation by dendritic cells and subsequent expansion of cytotoxic T cells strongly relied on tumor-intrinsic GSDME activity. In preclinical murine cancer models, defective GSDME signaling rendered tumors resistant to ICI therapy. Epigenetic reprogramming with upregulation of Gdsme enhanced the susceptibility of tumor cells to inflammatory cell death and immunotherapy. In humans, transcriptome analysis of melanoma samples showed strong correlation between genetic activity of the RIG-I and pyroptosis pathways. In melanoma patients, high transcriptional activity of a pyroptosis gene set was associated with prolonged survival and beneficial response to ICI therapy. In summary, our data show that GSDME links RIG-I and apoptotic signaling to inflammatory cell death, thereby driving its immunogenicity and responsiveness to ICI. A deeper understanding of these pathways may allow for the development of novel combined modality approaches to improve ICI treatment responses in cancer patients.
Collapse
Affiliation(s)
- Stefan Enssle
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anna Sax
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Peter May
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nadia El Khawanky
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nardine Soliman
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Markus Perl
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Julius C. Enssle
- Department of Medicine II, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Karsten Krey
- Institute of Virology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jürgen Ruland
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Clinical Chemistry and Pathobiochemistry, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Pichlmair
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Virology, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Florian Bassermann
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner-site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hendrik Poeck
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
- Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
- Center for immunomedicine intransplantation and oncology (CITO), Regensburg, Germany
| | - Simon Heidegger
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Centerfor Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
32
|
Zhao L, Huang J, Li Y, Wu S. LncRNA transcriptome analysis of rainbow trout ( Oncorhynchus mykiss) skin infected with IHNV reveals that lncRNA SARL/miR-205-z/ SOCS3 axis negatively regulates antiviral immunity mechanisms. Virulence 2025; 16:2486990. [PMID: 40287819 PMCID: PMC12036486 DOI: 10.1080/21505594.2025.2486990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/06/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are new gene regulators involved in various biological processes. However, the regulatory effect of lncRNA on the rainbow trout (Oncorhynchus mykiss) antiviral immune response has not been reported. Here, we measured lncRNA profiles at 48 hpi compared to the control group, expression levels of lncRNA, miRNA, and gene, and lncRNA SARL/miR-205-z/SOCS3 functions after rainbow trout skin infected with infectious haematopoietic necrosis virus (IHNV) by RNA-seq, qRT-PCR, and overexpression and inhibition assays. Transcriptome analysis identified twelve upregulated and four downregulated DElncRNAs. Twelve key immune-related competing endogenous RNA (ceRNA) networks were identified, and the target genes were enriched in the TLR, RLR, NLR, and p53 signalling pathways. Expression patterns suggested that changes in lncRNA SARL, miR-205-z, and SOCS3 expression presented a ceRNA regulatory relationship. Further studies demonstrated that the lncRNA SARL was a ceRNA of SOCS3 by sponging miR-205-z in vitro, thereby playing a negative regulatory role in the antiviral immune response of rainbow trout. We also found that miR-205-z was a positive regulator of rainbow trout liver cell proliferation, and this effect could be reversed by SOCS3. In vivo, SOCS3 expression significantly increased after antagomiR-205-z injection. Furthermore, SOCS3 overexpression significantly promoted the replication of IHNV. This study provides fundamental data for disease resistance breeding and targeted drug therapy in rainbow trout.
Collapse
Affiliation(s)
- Lu Zhao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jinqiang Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yongjuan Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- College of Science, Gansu Agricultural University, Lanzhou, China
| | - Shenji Wu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
33
|
Ferraris S, Scalia AC, Nascimben M, Perero S, Rimondini L, Spriano S, Cochis A. Bacteriostatic, silver-doped, zirconia-based thin coatings for temporary fixation devices tuning stem cells' expression of adhesion-relevant genes and proteins. BIOMATERIALS ADVANCES 2025; 176:214360. [PMID: 40449285 DOI: 10.1016/j.bioadv.2025.214360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/22/2025] [Accepted: 05/26/2025] [Indexed: 06/03/2025]
Abstract
Temporary fixation devices must support bone healing, be easily removed without bone tissue overgrowth, and reduce the risk of infection. To match these needs, mechanically and chemically stable thin coatings, based on a zirconia matrix doped with silver (ZrO2-Ag), were sputtered on Ti6Al4V. Coatings with two silver concentrations were produced: a low (0.2 % at Ag) concentration (AL) for bacteriostatic effect and a high (0.5 % at Ag) concentration (AH) for antibacterial properties. Surfaces were characterized for silver content and release, mechanical adhesion, morphology, roughness, wettability, and surface zeta potential, reporting high stability and a continuous Ag release over 28 days. Direct cytocompatibility was shown for human mesenchymal stem cells (hMSC), while antibacterial properties were verified towards Staphylococcus aureus. Results revealed non-toxic and anti-adhesion effects of AL that were deeply investigated towards hMSC by a multi-omics approach. Transcriptomics revealed a down-regulation of cadherins- and integrins-related genes involved in the cell-to-cell and cell-to-substrate adhesion, whereas proteomics confirmed a reduced expression of adhesion proteins (Talin and Ras homolog family member A - RhoA). The OMICS profiles were matched by bioinformatics analysis, confirming a cluster of preserved biological functions strongly related to the cells' adhesion but not to apoptosis. Therefore, AL is a good candidate for bone temporary fixation devices, not interfering with bone healing (cytocompatible), avoiding bone adhesion on the implant surface, and being bacteriostatic.
Collapse
Affiliation(s)
- Sara Ferraris
- Department of Applied Science and Technology, Politecnico di Torino, Italy
| | - Alessandro C Scalia
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases CAAD, Università del Piemonte Orientale UPO, Italy
| | - Mauro Nascimben
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases CAAD, Università del Piemonte Orientale UPO, Italy
| | - Sergio Perero
- Department of Applied Science and Technology, Politecnico di Torino, Italy
| | - Lia Rimondini
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases CAAD, Università del Piemonte Orientale UPO, Italy
| | - Silvia Spriano
- Department of Applied Science and Technology, Politecnico di Torino, Italy.
| | - Andrea Cochis
- Department of Health Sciences, Center for Translational Research on Autoimmune and Allergic Diseases CAAD, Università del Piemonte Orientale UPO, Italy
| |
Collapse
|
34
|
Yu Y, Zhang W, Ding Q, Cheng X, Wang K, Zhang G, Jiang B, Yu X, Li YT, Zhang GJ. Dual-antibody functionalized transistor biosensor for specific diagnosis of liver cancer. Talanta 2025; 293:128095. [PMID: 40203597 DOI: 10.1016/j.talanta.2025.128095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/29/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Selectively and sensitively detecting specific exosomal markers is critical for early diagnosis of liver cancer. However, identifying specific exosomal biomarkers and establishing accurate, convenient detection methods remain challenging. In this study, we used bioinformatics to identify the higher levels of EpCAM and GPC-3 proteins on liver cancer exosomes. These markers were used to create a dual-antibody functionalized transistor biosensor for precise detection of liver cancer exosomes. The techniques exhibited outstanding specificity and sensitivity. Detection thresholds in PBS and simulated plasma were established at 20 particles/μL and 47 particles/μL, respectively, facilitating the distinction of liver cancer cell-derived exosomes from those originating from various other cancer cells. Furthermore, in clinical samples testing, this approach not only distinguished clinical samples among liver cancer patients and healthy individuals, but also demonstrated the ability to differentiate liver cancer from other types of tumors, achieving a precision and accuracy rate of 100 %. The developed biosensor demonstrates excellent potential for clinical application and this work offers a promising and effective approach for cancer diagnosis.
Collapse
Affiliation(s)
- Yi Yu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, PR China
| | - Wenhao Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Qiyue Ding
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Xiaolu Cheng
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Kaiwei Wang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Guangxin Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Boan Jiang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China
| | - Xionghua Yu
- Xiantao Hospital of Traditional Chinese Medicine, Xiantao, Hubei, 433000, PR China.
| | - Yu-Tao Li
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, PR China.
| | - Guo-Jun Zhang
- School of Laboratory Medicine, Hubei University of Chinese Medicine, 16 Huangjia Lake West Road, Wuhan, 430065, PR China; Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, PR China.
| |
Collapse
|
35
|
Kuhn KD, Cho UH, Hetzer MW. PSME3 regulates migration and differentiation of myoblasts. Life Sci Alliance 2025; 8:e202503208. [PMID: 40537284 DOI: 10.26508/lsa.202503208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 06/03/2025] [Accepted: 06/04/2025] [Indexed: 06/22/2025] Open
Abstract
The acquisition of cellular identity requires large-scale alterations in cellular state. The noncanonical proteasome activator PSME3 is known to regulate diverse cellular processes, but its importance for differentiation remains unclear. Here, we demonstrate that PSME3 binds dynamically to highly active promoters over the course of differentiation. However, loss of PSME3 does not globally affect mRNA transcription. We find instead that PSME3 influences the levels of several adhesion-related proteins and acts upstream of the HSP90 co-chaperone NUDC to regulate cell motility and myoblast differentiation in a proteasome-independent manner. Our findings reveal several new facets of PSME3 functionality and highlight its importance for the differentiation of myogenic cells.
Collapse
Affiliation(s)
- Kenneth D Kuhn
- Institute of Science and Technology (ISTA), Klosterneuburg, Austria
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- The Neurosciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Ukrae H Cho
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Martin W Hetzer
- Institute of Science and Technology (ISTA), Klosterneuburg, Austria
| |
Collapse
|
36
|
Comertpay B, Gov E. Multiomics Analysis and Machine Learning-based Identification of Molecular Signatures for Diagnostic Classification in Liver Disease Types Along the Microbiota-gut-liver Axis. J Clin Exp Hepatol 2025; 15:102552. [PMID: 40292334 PMCID: PMC12019836 DOI: 10.1016/j.jceh.2025.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Background Liver disease, responsible for around two million deaths annually, remains a pressing global health challenge. Microbial interactions within the microbiota-gut-liver axis play a substantial role in the pathogenesis of various liver conditions, including early chronic liver disease (eCLD), chronic liver disease (CLD), acute liver failure (ALF), acute-on-chronic liver failure (ACLF), non-alcoholic fatty liver disease (NAFLD), steatohepatitis, and cirrhosis. This study aimed to identify key molecular signatures involved in liver disease progression by analyzing transcriptomic and gut microbiome data, and to evaluate their diagnostic utility using machine learning models. Methods Transcriptomic analysis identified differentially expressed genes (DEGs) that, when integrated with regulatory elements microRNAs, transcription factors, receptors, and the gut microbiome highlight disease-specific molecular interactions. To assess the diagnostic potential of these molecular signatures, a two-step analysis involving principal component analysis (PCA) and Random Forest classification was conducted, achieving accuracies of 75% for ALF and 89% for NAFLD. Additionally, machine learning algorithms, including K-neighbors, multi-layer perceptron (MLP), decision tree, Random Forest, logistic regression, gradient boosting, CatBoost, Extreme Gradient Boosting (XGB), and Light Gradient Boosting Machine (LGBM), were applied to gene expression data for ALF and NAFLD. Results Key genes including CLDN14, EGFR, GSK3B, MYC, and TJP2, alongside regulatory miRNAs let-7a-5p, miR-124-3p, and miR-195-5p and transcription factors NFKB1 and SP1 may be suggested as critical to liver disease progression. Additionally, gut microbiota members, Dictyostelium discoideum and Eikenella might be novel candidates associated with liver disease, highlighting the importance of the gut-liver axis. The Random Forest model reached 75% accuracy and 83% area under the curve for ALF, while NAFLD classification achieved 100% accuracy, precision, recall, and area under the curve underscoring robust diagnostic potential. Conclusion This study establishes a solid foundation for further research and therapeutic advancement by identifying key biomolecules and pathways critical to liver disease. Additional experimental validation is needed to confirm clinical applicability.
Collapse
Affiliation(s)
- Betul Comertpay
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| | - Esra Gov
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| |
Collapse
|
37
|
Dong J, Mao Z, Li H, Wang R, Wang Y, Jia H, Li J, Liu Q, Zhang C, Liao X, Liu D, Ma H, Tian C. MTD: A cloud-based omics database and interactive platform for Myceliophthora thermophila. Synth Syst Biotechnol 2025; 10:783-793. [PMID: 40276250 PMCID: PMC12018684 DOI: 10.1016/j.synbio.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/26/2025] Open
Abstract
Nowadays, biological databases are playing an increasingly critical role in biological research. Myceliophthora thermophila is an excellent thermophilic fungal chassis for industrial enzyme production and plant biomass-based chemical synthesis. The lack of a dedicated public database has made access to and reanalysis of M. thermophila data difficult. To bridge this gap, we developed MTD (https://mtd.biodesign.ac.cn/), a cloud-based omics database and interactive platform for M. thermophila. MTD integrates comprehensive genome annotations, sequence-based predictions, transcriptome data, curated experimental descriptions, and bioinformatics analysis tools, offering a comprehensive, one-stop solution with a 'top-down' search strategy to streamline M. thermophila research. The platform supports data reproduction, rapid querying, and in-depth mining of existing transcriptome datasets. Based on analyses using data and tools in MTD, we identified shifts in metabolic allocation in a glucoamylase hyperproduction strain of M. thermophila, highlighting changes in fatty acid biosynthesis and amino acids biosynthesis pathways, which provide new insights into the underlying phenotypic alterations. As a pioneering resource, MTD marks a key advancement in M. thermophila research and sets the model for developing similar databases for other species.
Collapse
Affiliation(s)
- Jiacheng Dong
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Zhitao Mao
- Biodesign Center, State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Haoran Li
- Biodesign Center, State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Ruoyu Wang
- Biodesign Center, State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Yutao Wang
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Haokai Jia
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Jingen Li
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Qian Liu
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Chenglin Zhang
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Xiaoping Liao
- Biodesign Center, State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Defei Liu
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Hongwu Ma
- Biodesign Center, State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| | - Chaoguang Tian
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, 300308, China
| |
Collapse
|
38
|
Kulkarni MM, Popovic B, Nolfi AL, Skillen CD, Brown BN. Distinct impacts of aging on the immune responses to extracellular matrix-based versus synthetic biomaterials. Biomaterials 2025; 320:123204. [PMID: 40056612 DOI: 10.1016/j.biomaterials.2025.123204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 03/10/2025]
Abstract
All implanted materials inevitably trigger an acute inflammatory response. The long-term outcome, however, is dependent on the trajectory of this response. This study investigates the effects of aging on the immune response to two commercially available biomaterials. Extracellular matrix-based urinary bladder matrix (UBM) and synthetic polypropylene mesh (PPM) were implanted in young (4 months) and aged (18 months) C57BL/6J mice. Overall, PPM led to a sustained inflammatory response regardless of the age of the mice. In contrast, UBM induced an initial inflammatory response that matured into a pro-regenerative/remodeling response with time, though aged mice exhibited a delayed resolution of inflammation. The PPM-induced response was predominantly pro-inflammatory with consistently higher M1-like macrophage phenotype, whereas the response to UBM was characterized by an anti-inflammatory M2-like phenotype, especially in young mice. RNA sequencing revealed marked age-related differences in gene transcription. At day 7 post-implantation, the young mice with UBM showed a robust upregulation of both pro- and anti-inflammatory pathways as compared to young mice implanted with PPM, however, by day 14, the gene expression profile transitioned into an anti-inflammatory profile. Intriguingly, in aged mice, the response to UBM was distinct with consistent downregulation of inflammatory genes compared to PPM, while the response to PPM in both young and aged animals was largely consistent. Upstream analysis identified cytokines as key drivers of the host response, with IL-4 and IL-13 in young mice, and TNF-α and IL-1β driving chronic inflammation in aged mice. These findings highlight the importance of host age in biomaterial outcome, and the potential of ECM-based materials to mount a favorable response even in the presence of age-related immune dysregulation.
Collapse
Affiliation(s)
- Mangesh M Kulkarni
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Branimir Popovic
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alexis L Nolfi
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Clint D Skillen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Bryan N Brown
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
39
|
Zhu Z, Zhao J, Ji X, Hu W, Leng W, Xu C, Li X, Yang K, Li X, Zheng Y, Lin J. Bacterial cellulose-based scaffold with in-situ cationic micelle modification for urethral stricture disease: Sustained drug components release, cytokines recruitment, and bacterial microenvironment regulation. Bioact Mater 2025; 51:306-317. [PMID: 40491686 PMCID: PMC12145995 DOI: 10.1016/j.bioactmat.2025.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/10/2025] [Accepted: 04/25/2025] [Indexed: 06/11/2025] Open
Abstract
The treatment of urethral stricture disease and the prevention of restenosis present considerable challenges in the field of urology. Tissue-engineered materials, particularly bacterial cellulose scaffolds, have emerged as promising solutions due to their abundant sources, excellent mechanical properties, and biocompatibility. However, for attaining superior treatment for patients with USD, further modification of bacterial cellulose is necessary. We have fabricated a dual-network scaffold with enhanced antibacterial properties and cytokines absorption ability through in-situ polymerization of cationic polyurethane micelles and cyclodextrin on oxidized bacterial cellulose. This scaffold also enables long-term sustained release of loaded drug components. Animal model studies have confirmed that this scaffold can achieve urethral repair outcomes comparable to those of normal urethral tissue. This innovative material provides a robust foundation for advancing new concepts and methodologies in the treatment of urethral stricture disease, potentially transforming clinical approaches to this challenging condition.
Collapse
Affiliation(s)
- Zhenpeng Zhu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Jianming Zhao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Xing Ji
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Weimin Hu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Wenyuan Leng
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Chunru Xu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xiaoyu Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Kunlin Yang
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Yudong Zheng
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| |
Collapse
|
40
|
King JS, Wan M, Kim A, Prabhu S, Novak S, Kalajzic I, Delany AM, Sanjay A. Effects of aging on the immune and periosteal response to fracture injury. Bone 2025; 198:117524. [PMID: 40381878 PMCID: PMC12168153 DOI: 10.1016/j.bone.2025.117524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/14/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Aging predisposes individuals to reduced bone mass and fragility fractures, which are costly and linked to high mortality. Understanding how aging affects fracture healing is essential for developing therapies to enhance bone regeneration in older adults. During the inflammatory phase of fracture healing, immune cells are recruited to the injury site as periosteal skeletal stem/progenitor cells (pSSPCs) rapidly proliferate and differentiate into osteochondral lineages, allowing for fibrocartilaginous callus formation and, subsequently, complete bone healing. Irrespective of age, how periosteal mesenchymal and immune cells interact during early fracture healing is incompletely understood, limiting our ability to modulate this process. To address this, we directly analyzed, in parallel, at a single-cell level, isolated murine CD45(+) and CD45(-) periosteal cells dissected from intact and fractured bones, collected three days after injury. Comprehensive analysis, corroborated by bulk RNA-sequencing, flow cytometry, and histology, demonstrated that aging decreased pSSPC proliferation, markedly reduced expression of genes required for callus formation, and increased senescence signature. During the regeneration phase, at 14 days post injury, aged mice demonstrated reduced mineralization of the callus, accompanied by elevated Sox9 expression and increased cartilage content, suggesting delayed repair. We also found that the chemokine Cxcl9 was highly upregulated in aged intact Prrx1+ pSSPCs, which has the potential to directly regulate other pSSPCs, and was associated with increased recruitment of CD8+ T cells at the fracture site. Cell-to-cell communication analysis provided further appreciation of the complex interactions among the many mesenchymal and hematopoietic cell types regulating fracture healing and highlighted the impact of aging on these interactions. Together, these results provide insight into age-induced alterations in early fracture healing, which could facilitate the development of improved therapeutic approaches for fracture repair in the elderly.
Collapse
Affiliation(s)
- Justin S King
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America
| | - Matthew Wan
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America
| | - Adam Kim
- Department of Medicine, United States of America
| | - Shagun Prabhu
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America
| | - Sanja Novak
- UConn Musculoskeletal Institute, United States of America; Center for Regenerative Medicine and Skeletal Development, United States of America
| | - Ivo Kalajzic
- UConn Musculoskeletal Institute, United States of America; Center for Regenerative Medicine and Skeletal Development, United States of America
| | - Anne M Delany
- Department of Medicine, United States of America; Center for Molecular Oncology, UConn Health, Farmington, CT 06032, United States of America
| | - Archana Sanjay
- Department of Orthopedic Surgery, United States of America; UConn Musculoskeletal Institute, United States of America.
| |
Collapse
|
41
|
Uusi-Mäkelä M, Harjula SKE, Junno M, Sillanpää A, Nätkin R, Niskanen MT, Saralahti AK, Nykter M, Rämet M. The inflammasome adaptor pycard is essential for immunity against Mycobacterium marinum infection in adult zebrafish. Dis Model Mech 2025; 18:dmm052061. [PMID: 39916610 PMCID: PMC11972081 DOI: 10.1242/dmm.052061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/03/2025] [Indexed: 03/25/2025] Open
Abstract
Inflammasomes regulate the host response to intracellular pathogens including mycobacteria. We have previously shown that the course of Mycobacterium marinum infection in adult zebrafish (Danio rerio) mimics the course of tuberculosis in human. To investigate the role of the inflammasome adaptor pycard in zebrafish M. marinum infection, we produced two zebrafish knockout mutant lines for the pycard gene with CRISPR/Cas9 mutagenesis. Although the zebrafish larvae lacking pycard developed normally and had unaltered resistance against M. marinum, the loss of pycard led to impaired survival and increased bacterial burden in the adult zebrafish. Based on histology, immune cell aggregates, granulomas, were larger in pycard-deficient fish than in wild-type controls. Transcriptome analysis with RNA sequencing of a zebrafish haematopoietic tissue, kidney, suggested a role for pycard in neutrophil-mediated defence, haematopoiesis and myelopoiesis during infection. Transcriptome analysis of fluorescently labelled, pycard-deficient kidney neutrophils identified genes that are associated with compromised resistance, supporting the importance of pycard for neutrophil-mediated immunity against M. marinum. Our results indicate that pycard is essential for resistance against mycobacteria in adult zebrafish.
Collapse
Affiliation(s)
- Meri Uusi-Mäkelä
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | | | - Maiju Junno
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Alina Sillanpää
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Reetta Nätkin
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, FI-33521 Tampere, Finland
| | | | | | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, FI-33521 Tampere, Finland
| | - Mika Rämet
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| |
Collapse
|
42
|
Santosa EK, Zhang JM, Sauter JC, Lee ME, Ng BD, Stulz SV, Takizawa M, Grassmann S, Weizman OE, Adams NM, Chaligné R, Oxenius A, Gasteiger G, Lau CM, Sun JC. Defining molecular circuits of CD8+ T cell responses in tissues during latent viral infection. J Exp Med 2025; 222:e20242078. [PMID: 40387857 DOI: 10.1084/jem.20242078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/18/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025] Open
Abstract
Latent viral infections rely on a precise coordination of the immune response to control sporadic viral reactivation. CD8+ T cells play a crucial role in controlling viral latency by generating diverse memory responses in an epitope-specific manner. Among these distinct responses, conventional and inflationary memory responses have been described during herpesvirus infections. Using a newly generated TCR transgenic mouse strain, we investigated the transcriptomic and epigenetic remodeling of distinct epitope-specific CD8+ T cells during CMV infection across tissues at both population and single-cell levels. Our findings reveal that whereas the transcriptomic and epigenetic landscapes of conventional and inflationary memory responses diverge in the spleen and liver, these molecular programs converge in the salivary gland, a site of CMV persistence. Thus, we provide evidence that the dynamics of memory CD8+ T cell responses are distinct between tissues.
Collapse
Affiliation(s)
- Endi K Santosa
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University , New York, NY, USA
| | - Jennifer M Zhang
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - John C Sauter
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Mariah E Lee
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Brandon D Ng
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
- Pharmacology Program, Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University , New York, NY, USA
| | - Sigrun V Stulz
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg , Würzburg, Germany
| | - Meril Takizawa
- Single Cell Analytics Innovation Lab, Memorial Sloan Kettering Cancer Center , New York, NY, USA
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Simon Grassmann
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Orr-El Weizman
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Nicholas M Adams
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ronan Chaligné
- Single Cell Analytics Innovation Lab, Memorial Sloan Kettering Cancer Center , New York, NY, USA
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | | | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg , Würzburg, Germany
| | - Colleen M Lau
- Department of Microbiology and Immunology, College of Veterinary Medicine of Cornell University, Ithaca, NY, USA
| | - Joseph C Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College and Graduate School of Medical Sciences of Cornell University , New York, NY, USA
| |
Collapse
|
43
|
Johansson Å, Venkita Subramani M, Yilmaz B, Nyström EE, Layunta E, Arike L, Sommer F, Rosenstiel P, Vereecke L, Mannerås-Holm L, Wullaert A, Pelaseyed T, Johansson ME, Birchenough GM. Neonatal microbiota colonization primes maturation of goblet cell-mediated protection in the pre-weaning colon. J Exp Med 2025; 222:e20241591. [PMID: 40323318 PMCID: PMC12051479 DOI: 10.1084/jem.20241591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/06/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Regulated host-microbe interactions are a critical aspect of lifelong health. Colonic goblet cells protect from microorganisms via the generation of a mucus barrier structure. Bacteria-sensing sentinel goblet cells provide secondary protection by orchestrating mucus secretion when microbes breach the mucus barrier. Mucus deficiencies in germ-free mice implicate a role for the microbiota in programming barrier generation, but its natural ontogeny remains undefined. We now investigate the mucus barrier and sentinel goblet cell development in relation to postnatal colonization. Combined in vivo and ex vivo analyses demonstrate rapid and sequential microbiota-dependent development of these primary and secondary goblet cell protective functions, with dynamic changes in mucus processing dependent on innate immune signaling via MyD88 and development of functional sentinel goblet cells dependent on the NADPH/dual oxidase family member Duox2. Our findings identify new mechanisms of microbiota-goblet cell regulatory interaction and highlight the critical importance of the pre-weaning period for the normal development of protective systems that are key legislators of host-microbiota interaction.
Collapse
Affiliation(s)
- Åsa Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mahadevan Venkita Subramani
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Bahtiyar Yilmaz
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Elisabeth E.L. Nyström
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Elena Layunta
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Liisa Arike
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Felix Sommer
- Institute of Clinical & Molecular Biology, University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical & Molecular Biology, University of Kiel, Kiel, Germany
| | - Lars Vereecke
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Louise Mannerås-Holm
- Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Andy Wullaert
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Biomedical Sciences, Cell Death Signalling Lab, University of Antwerp, Antwerp, Belgium
| | - Thaher Pelaseyed
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Malin E.V. Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - George M.H. Birchenough
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Xu J, Jia Z, Zhao X, Wang L, Jin G, Li Z, Yin N, Li Y, Peng M. BCOR and ZC3H12A suppress a core stemness program in exhausted CD8+ T cells. J Exp Med 2025; 222:e20241133. [PMID: 40327039 PMCID: PMC12054362 DOI: 10.1084/jem.20241133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
In chronic viral infections, sustained CD8+ T cell response relies on TCF1+ precursor-exhausted T cells (TPEX) exhibiting stem-like properties. TPEX self-renew and respond to PD-1 blockade, underscoring their paramount importance. However, strategies for effectively augmenting TPEX remain limited. Here, we demonstrate that ZC3H12A deficiency initiates a stemness program in TPEX but also increases cell death, whereas BCOR deficiency predominantly promotes TPEX proliferation. Consequently, co-targeting of both BCOR and ZC3H12A imparts exceptional stemness and functionality to TPEX, thereby enhancing viral control. Mechanistically, BCOR and ZC3H12A collaboratively suppress a core stemness program in TPEX characterized by heightened expression of ∼216 factors. While TCF1 plays a role, this core stemness program relies on novel factors, including PDZK1IP1, IFIT3, PIM2, LTB, and POU2F2. Crucially, overexpressing POU2F2 robustly boosts TPEX and enhances antiviral immunity. Thus, a core stemness program exists in exhausted T cells, jointly repressed by BCOR and ZC3H12A, robustly controlling TPEX differentiation and providing new targets for addressing T cell exhaustion.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zeran Jia
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Xiaocui Zhao
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Lixia Wang
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Gang Jin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhuoyang Li
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Na Yin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yinqing Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Min Peng
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
45
|
Kim CW, Kim HJ, Kang I, Ku KB, Kim Y, Park JH, Lim J, Kang BH, Park WH, La J, Chang S, Hwang I, Kim M, Ahn S, Lee HK. Gut dysbiosis from high-salt diet promotes glioma via propionate-mediated TGF-β activation. J Exp Med 2025; 222:e20241135. [PMID: 40402148 PMCID: PMC12097148 DOI: 10.1084/jem.20241135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 03/08/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025] Open
Abstract
The purpose of this study is to investigate the impact of a high-salt diet (HSD), which is commonly found in Western countries, on the progression of glioma. Our research shows that the alterations in gut microbiota caused by an HSD facilitated the development of glioma. Mice fed an HSD have elevated levels of intestinal propionate, which accelerated the growth of glioma cells. We also find that propionate supplementation enhanced the response of glioma cells to low oxygen levels. Moreover, we identify a link between TGF-β signaling, response to low oxygen levels, and invasion-related pathways. Propionate treatment increases the expression of HIF-1α, leading to an increase in TGF-β1 production. Additionally, propionate treatment promotes glioma cell invasion through TGF-β signaling. Our findings suggest that an HSD-induced increase in propionate plays a crucial role in glioma progression by facilitating invasion through the hypoxic response and TGF-β signaling pathways, thereby establishing a significant connection between gut microbiota and the progression of glioma.
Collapse
Affiliation(s)
- Chae Won Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- KAIST Life Science Institute, KAIST, Daejeon, Republic of Korea
| | - Hyun-Jin Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- KAIST Life Science Institute, KAIST, Daejeon, Republic of Korea
| | - In Kang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Keun Bon Ku
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Yumin Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jang Hyun Park
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- KAIST Life Science Institute, KAIST, Daejeon, Republic of Korea
| | - Juhee Lim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Byeong Hoon Kang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Won Hyung Park
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jeongwoo La
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Sungwoo Chang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Inju Hwang
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Minji Kim
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
46
|
Ameen ZS, Mubarak AS, Hamad M, Hamoudi R, Jemimah S, Ozsahin DU, Hamad M. Incorporating time as a third dimension in transcriptomic analysis using machine learning and explainable AI. Comput Biol Chem 2025; 117:108432. [PMID: 40132403 DOI: 10.1016/j.compbiolchem.2025.108432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
Transcriptomic data analysis entails the measurement of RNA transcript (gene expression products) abundance in a cell or a cell population at a single point in time. In other words, transcriptomics as it is currently practiced is two-dimensional (2DTA). Gene expression profiling by 2DTA has proven invaluable in furthering our understanding of numerous biological processes in health and disease. That said, shortcomings including technical variability, small sample size, differential rates of transcript decay, and the lack of linearity between transcript abundance and functionality or the formation of functional proteins limit the interpretive utility and generalizability of transcriptomic data. 2DTA utility may also be constrained by its reliance on RNA extracts obtained at a single time point. In other words, much like judging a movie by a single frame, 2DTA can only provide a snapshot of the transcriptome at time of RNA extraction. Whether this perceived "temporality" problem is real and whether it has any bearing on transcriptomic data interpretation have yet to be addressed. To investigate this problem, 25 publicly available datasets relating to MCF-7 cells, where RNA extracts obtained at 12- or 48-hours post-culture were subjected to transcriptomic analysis. The individual datasets were downloaded and compiled into two separate datasets (MCF-7 U12hr and MCF-7 U48hr). To comparatively analyze the two compiled datasets, three machine learning approaches (decision trees (DT), random forests (RF), and XGBoost (Extreme Gradient Boosting)) were used as classifiers to search for genes with distinct expression patterns between the two groups. Shapley additive explanation (SHAP), an explainable AI method, was used to assess the fundamental principles of the DT, RF, and XGBoost models. Coefficient of Determination (DC), Mean Absolute Error (MAE), and Mean Squared Error (MSE) were used to evaluate the models. The results show that the two datasets exhibited very significant gene expression patterns. The XGBoost model performed better than the DT or RF models with MSE, MAE, and DC values of 0.00028, 0.00028, and 0.95778 respectively. These observations suggest that time, as a third dimension, can impact transcriptomic data interpretation and that machine learning and explainable AI are useful tools in resolving the temporality problem in transcriptomics.
Collapse
Affiliation(s)
- Zubaida Said Ameen
- Operational Research Center in Healthcare, Near East University, Mersin 99138, Turkey
| | - Auwalu Saleh Mubarak
- Operational Research Center in Healthcare, Near East University, Mersin 99138, Turkey
| | - Mohamed Hamad
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, UAE; Research Institute of Medical and Health Sciences, University of Sharjah, UAE
| | - Rifat Hamoudi
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, UAE; BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, UAE; Division of Surgery and Interventional Science, University College London, London NW3 2QG, UK
| | - Sherlyn Jemimah
- Department of Biology, College of Science, American University of Sharjah, UAE
| | - Dilber Uzun Ozsahin
- Operational Research Center in Healthcare, Near East University, Mersin 99138, Turkey; Research Institute of Medical and Health Sciences, University of Sharjah, UAE; Department of Diagnostic Medical Imaging, College of Health Sciences, University of Sharjah, UAE.
| | - Mawieh Hamad
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, UAE; Research Institute of Medical and Health Sciences, University of Sharjah, UAE.
| |
Collapse
|
47
|
Loomis S, Silva DG, Savopoulos R, Cilia J, Li J, Davis MD, Virley D, Foley A, Loro E, McCreary AC. Behavioral and transcriptomic effects of a novel cannabinoid on a rat valproic acid model of autism. Neuropharmacology 2025; 273:110450. [PMID: 40187640 DOI: 10.1016/j.neuropharm.2025.110450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by impaired social communication, restricted interests, repetitive behavior and irritability. Exposure to valproic acid (VPA) during pregnancy has been shown to increase the risk of autism in children and has led to the development of the in-utero VPA rat model that elicits neurodevelopmental autistic-like features. Offspring exhibit behavioral and neurobiological alterations modelling ASD symptoms. We performed a behavioral and molecular assessment in a rat in-utero VPA model treated with a novel botanical cannabinoid, JZP541. Male offspring from dams treated with VPA were tested acutely and sub-chronically with JZP541 (10, 30, or 100 mg/kg, intraperitoneally). A behavioral testing battery was performed, and brain frontal cortex and hippocampus used for RNA sequencing. In utero exposure to VPA resulted in progeny showing behavioral phenotypes characteristic of ASD. JZP541 attenuated these deficits in social, stereotypic, hyperactivity and irritability behavior in a dose-dependent fashion. VPA exposure was associated with a substantial transcriptional dysregulation impacting multiple key biological processes in a tissue-dependent manner. The expression profiles were integrated with publicly available datasets of autism-associated genes to support the validity of the model used and to focus on the effects of treatment on known autism-relevant transcriptional targets. This approach indicated a strong and dose-dependent reduction of the autism-associated gene expression signature in brain samples from animals dosed with JZP541. Our findings demonstrate JZP541 was able to ameliorate ASD associated behavioral deficits, and this was supported by improvements in putative transcriptional biomarkers of ASD.
Collapse
Affiliation(s)
- Sally Loomis
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK.
| | - Diogo G Silva
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | | - Jackie Cilia
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | - Jennifer Li
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | - Mat D Davis
- Jazz Pharmaceuticals Inc., Palo Alto, CA, USA
| | - David Virley
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | | - Emanuele Loro
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | |
Collapse
|
48
|
Clark KL, George JW. Environmentally relevant concentrations of individual per- and polyfluoroalkyl substances (PFAS) and a PFAS mixture impact proliferation, migration, and gene transcription in a human myometrial cell line. Toxicology 2025; 515:154173. [PMID: 40334771 DOI: 10.1016/j.tox.2025.154173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 05/01/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are persistent environmental contaminants linked to adverse health effects. Epidemiological studies have linked PFAS with an increased risk of uterine diseases including fibroids however, the mechanisms involved remain to be elucidated. This study examined the impact of individual PFAS, such as legacy compounds [perfluorooctanoic acid (PFOA), perfluorooctanesulfonic acid (PFOS)] and alternative short-chain compounds [GENX/HFPO-DA and perfluorobutanesulfonic acid (PFBS)], along with a PFAS mixture, on the function and transcriptome of immortalized human myometrial cells (UT-TERT). Exposure to these PFAS resulted in increased cell viability and proliferation. Flow cytometry showed that PFOS and the PFAS mixture altered cell cycle progression, while migration assays indicated significant enhancement of cell migration following PFOS and mixture exposure. Moreover, PFOA, PFBS, and the PFAS mixture impaired gap junction intercellular communication (GJIC), suggesting possible disruptions in cellular communication in the uterine environment. Transcriptomic analysis identified extensive changes in gene expression after exposure to environmentally relevant PFAS levels, revealing common molecular pathways involved in cell signaling, lipid metabolism, and cell survival. These findings provide crucial insights into how PFAS may contribute to reproductive health risks, warranting further investigation into the long-term effects of PFAS on uterine function and overall reproductive health.
Collapse
Affiliation(s)
- Kendra L Clark
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States; Department of Environmental, Agricultural, and Occupational Health, University of Nebraska Medical Center, Omaha, NE, United States; Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States.
| | - Jitu W George
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States; University of Minnesota Genomics Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
49
|
Liu Z, Zhang H, Wang J, Yao Y, Wang X, Liu Y, Fang W, Liu X, Zheng Y. Clca1 deficiency exacerbates colitis susceptibility via impairment of mucus barrier integrity and gut microbiota homeostasis. Microbiol Res 2025; 297:128191. [PMID: 40300372 DOI: 10.1016/j.micres.2025.128191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
The intestinal mucus barrier has emerged as a promising therapeutic target for inflammatory bowel disease. Understanding its regulatory mechanisms is critical for elucidating ulcerative colitis (UC) pathogenesis, improving diagnostics, guiding treatments, and preventing relapse. Chloride Channel Accessory 1 (Clca1), a constituent of the mucus layer, remains understudied in colitis. Here, we investigated Clca1's role in mucosal immunity and intestinal homeostasis using experimental colitis models. Clca1-deficient (Clca1-/-) mice displayed compromised mucus layer integrity, reduced neutrophil infiltration, and gut microbiota dysbiosis. Notably, Clca1-/- mice exhibited exacerbated colitis severity following dextran sulfate sodium (DSS) challenge, accompanied by a diminished goblet cell populations. Fecal microbiota transplantation (FMT) studies revealed that gut microbiota critically modulates divergent phenotypic outcomes between genotypes. Our findings establish Clca1 as a multifunctional regulator of mucus barrier integrity through mechanisms involving goblet cell maintenance, neutrophil-mediated immunity, and host-microbiota crosstalk. These results advance the understanding of UC pathogenesis and identify Clca1-associated pathways as potential targets for barrier restoration therapies.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hong Zhang
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jingjing Wang
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yutong Yao
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyi Wang
- Core Facility Center, The First Afliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yang Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xingyin Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| | - Yi Zheng
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
50
|
Guan L, Wang P, Li Y, Zhang S. FERONIA interacts with NPL4 to regulate immunity gene mRNA nucleocytoplasmic transport in response to plant immunity. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2025; 357:112545. [PMID: 40348341 DOI: 10.1016/j.plantsci.2025.112545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Nucleocytoplasmic transport plays a critical role in the activation of immune mechanisms in plant cells. Fluorescence imaging analysis indicated that a high concentration of rapid alkalinization factor (RALF) suppresses the immune response and can induce nuclear envelope (NE) shape deformation. This phenomenon depends on the receptor kinase FERONIA (FER). Consistently, bacterial infection also affects NE shape. This study presents evidence that FER displays functional interactions with NPL4 and that phosphorylated NPL4 promotes its stability. We reported the identification and characterization of the nuclear localization protein NPL4, which is directly involved in general mRNA nuclear export. FER and NPL4 mutations both affected rhizosphere Pseudomonas colonization levels, suggesting that the interactions between FER and NPL4 are largely indispensable for regulating rhizosphere Pseudomonas colonization levels. In addition, NPL4 altered the mRNA nucleocytoplasmic distribution of immune genes in conjunction with the function of RALF-FER in the suppression of plant immunity. In brief, NPL4, which is downstream of FER is required for innate immunity and mRNA nuclear accumulation of resistance genes in Arabidopsis. Overall, through the analysis of RALF-FER and the nuclear localization protein NPL4, this work provides novel insights into the mRNA nucleocytoplasmic transport of immune genes and plant health.
Collapse
Affiliation(s)
- Li Guan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, Hunan University, Changsha 410082, PR China.
| | - Peilong Wang
- Zhejiang Institute of Subtropical Crops, Zhejiang Academy of Agricultural Sciences, 334 Xueshan Road, Wenzhou 325005, PR China
| | - Yongliang Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Key Laboratory of Plant Functional Genomics and Developmental Regulation, Hunan University, Changsha 410082, PR China
| | - Sufang Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Key Laboratory for Innovative Development and Utilization of Forest Plant Germplasm, College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou 510642, PR China
| |
Collapse
|