1
|
Xia F, Santacruz A, Wu D, Bertho S, Fritz E, Morales-Sosa P, McKinney S, Nowotarski SH, Rohner N. Reproductive Adaptation of Astyanax mexicanus Under Nutrient Limitation. Dev Biol 2025:S0012-1606(25)00101-0. [PMID: 40222642 DOI: 10.1016/j.ydbio.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025]
Abstract
Reproduction is a fundamental biological process for the survival and continuity of species. Examining changes in reproductive strategies offers valuable insights into how animals have adapted their life histories to different environments. Since reproduction is one of the most energy-intensive processes in female animals, nutrient scarcity is expected to interfere with the ability to invest in gametes. Lately, a new model to study adaptation to nutrient limitation has emerged; the Mexican tetra Astyanax mexicanus. This fish species exists as two different morphs, a surface river morph and a cave-dwelling morph. The cave-dwelling morph has adapted to the dark, lower biodiversity, and nutrient-limited cave environment and consequently evolved an impressive starvation resistance. However, how reproductive strategies have adapted to nutrient limitations in this species remains poorly understood. Here, we compared breeding activities and maternal contributions between laboratory-raised surface fish and cavefish. We found that cavefish produce different clutch sizes of eggs with larger yolk compared to surface fish, indicating a greater maternal nutrient deposition in cavefish embryos. To systematically characterize yolk compositions, we used untargeted proteomics and lipidomics approaches to analyze protein and lipid profiles in 2-cell stage embryos and found an increased proportion of sphingolipids in cavefish compared to surface fish. Additionally, we generated transcriptomic profiles of surface fish and cavefish ovaries using a combination of single cell and bulk RNA sequencing to examine differences in maternal contribution. We found that genes essential for hormone regulation were upregulated in cavefish follicular somatic cells compared to surface fish. To evaluate whether these differences contribute to their reproductive abilities under natural-occurring stress, we induced breeding in starved female fish. Remarkably, cavefish maintained their ability to breed under starvation, whereas surface fish largely lost this ability. We identified insulin-like growth factor 1a receptor (igf1ra) as a potential candidate gene mediating the downregulation of ovarian development genes, potentially contributing to the starvation-resistant fertility of cavefish. Taken together, we investigated the female reproductive strategies in Astyanax mexicanus, which will provide fundamental insights into the adaptations of animals to environments with extreme nutrient deficit.
Collapse
Affiliation(s)
- Fanning Xia
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Ana Santacruz
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Di Wu
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Elizabeth Fritz
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Sean McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO, USA.
| |
Collapse
|
2
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
3
|
Park K, Shin KO, Kim YI, Nielsen-Scott AL, Mainzer C, Celli A, Bae Y, Chae S, An H, Choi Y, Park JH, Park SH, Hwang JT, Kang SG, Wakefield JS, Arron ST, Holleran WM, Mauro TM, Elias PM, Uchida Y. Sphingosine-1-Phosphate-Cathelicidin Axis Plays a Pivotal Role in the Development of Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2025; 145:854-863.e6. [PMID: 39218144 DOI: 10.1016/j.jid.2024.08.008] [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: 10/12/2023] [Revised: 07/10/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a common skin cancer caused by mutagenesis resulting from excess UVR or other types of oxidative stress. These stressors also upregulate the production of a cutaneous innate immune element, cathelicidin antimicrobial peptide (CAMP), through endoplasmic reticulum stress-initiated, sphingosine-1-phosphate (S1P) signaling pathway. Although CAMP has beneficial antimicrobial activities, it also can be proinflammatory and procarcinogenic. We addressed whether and how S1P-induced CAMP production leads to cSCC development. Our study demonstrated that (i) CAMP expression is increased in cSCC cells and skin from patients with cSCC; (ii) S1P levels are elevated in cSCC cells, whereas inhibition of S1P production attenuates CAMP-stimulated cSCC growth; (iii) exogenous CAMP stimulates cSCC but not normal human keratinocyte growth; (iv) blockade of FPRL1 protein, a CAMP receptor, attenuates cSCC growth as well as the growth and invasion of cSCC cells mediated by CAMP into an extracellular matrix-containing fibroblast substrate; (v) FOXP3+ regulatory T-cell (which decreases antitumor immunity) levels increase in cSCC skin; and (vi) CAMP induces endoplasmic reticulum stress in cSCC cells. Together, the endoplasmic reticulum stress-S1P-CAMP axis forms a vicious circle, creating a favorable environment for cSCC development, that is, cSCC growth and invasion impede anticancer immunity.
Collapse
Affiliation(s)
- Kyungho Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; LaSS, Chuncheon, Republic of Korea
| | - Young-Il Kim
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Anna L Nielsen-Scott
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Carine Mainzer
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Anna Celli
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Yoojin Bae
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| | - Seungwoo Chae
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| | - Hahyun An
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| | - Yerim Choi
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; LaSS, Chuncheon, Republic of Korea
| | - Jae-Ho Park
- Personalized Diet Research Group, Korea Food Research Institute, Jeonju, Republic of Korea
| | - Soo-Hyun Park
- Personalized Diet Research Group, Korea Food Research Institute, Jeonju, Republic of Korea
| | - Jin-Taek Hwang
- Personalized Diet Research Group, Korea Food Research Institute, Jeonju, Republic of Korea; Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Seung Goo Kang
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Joan S Wakefield
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Sarah T Arron
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Walter M Holleran
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Theodora M Mauro
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Peter M Elias
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Yoshikazu Uchida
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.
| |
Collapse
|
4
|
Reinisch I, Enzenhofer S, Prokesch A. Mechanisms of Lipid-Associated Macrophage Accrual in Metabolically Stressed Adipose Tissue. Bioessays 2025; 47:e202400203. [PMID: 39828607 PMCID: PMC11931678 DOI: 10.1002/bies.202400203] [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: 08/21/2024] [Revised: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Adipose tissue (AT) inflammation, a hallmark of the metabolic syndrome, is triggered by overburdened adipocytes sending out immune cell recruitment signals during obesity development. An AT immune landscape persistent throughout weight loss and regain constitutes an immune-obesogenic memory that hinders long-term weight loss management. Lipid-associated macrophages (LAMs) are emerging as major players in diseased, inflamed metabolic tissues and may be key contributors to an obesogenic memory in AT. Our previous study found that LAM abundance increases with weight loss via intermittent fasting (IF) in obese mice, which is driven by adipocyte p53 signalling. However, the specific signals causing LAM accumulation in AT under IF remain unknown. In this piece, we hypothesise on a range of adipocyte-secreted signals that can harbor immune-attractive features upon fasting/refeeding cycles. We highlight possible mechanisms including cell death signalling, matrikines, and other damage-associated molecular patterns (DAMPs), as well as adipo(-cyto)kines, lipid mediators, metabolites, extracellular vesicles, and epigenetic rewiring. Finally, we consider how advances in mechanisms of AT LAM recruitment gleaned from preclinical models might be translatable to long-term weight management in humans. Thus, we provide vantage points to study signals driving monocyte recruitment, polarisation towards LAMs, and LAM retention, to harness the therapeutic potential of modulating AT LAM levels by impacting the immune-obesogenic memory in metabolic disease.
Collapse
Affiliation(s)
- Isabel Reinisch
- Department of Health Sciences and Technology, Institute of Food Nutrition and HealthEidgenössische Technische Hochschule Zürich (ETH)SchwerzenbachSwitzerland
| | - Sarah Enzenhofer
- Gottfried Schatz Research Center for Cell SignalingMetabolism and Aging, Division of Cell Biology, Histology and EmbryologyMedical University of GrazGrazAustria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell SignalingMetabolism and Aging, Division of Cell Biology, Histology and EmbryologyMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| |
Collapse
|
5
|
Gao X, Shen J, Lv P, Niu X, Huang Y. Mechanism of MWCNTs induced hepatotoxicity in Cyprinus carpio: New perspective from a Lipidomic study. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 283:107339. [PMID: 40179775 DOI: 10.1016/j.aquatox.2025.107339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Carbon nanotubes (CNTs), an extensively applied nanomaterial, can cause damage to the liver in fish. However, the underlying lipotoxic modes of CNTs exposure have not been studied. In the present study, juvenile Cyprinus carpio was exposed via water to various doses of multi-walled CNTs (MWCNTs) (0, 0.25, and 2.5 mg L-1) for a consecutive 28-day period. Our results showed elevated hepatosomatic index (HSI) and aspartate aminotransferase (AST) activities in plasma in all exposure groups, indicating that MWCNTs exposure induced liver dysfunction. Significantly increased low-density lipoprotein (LDL) and total cholesterol (CHO) levels along with significantly decreased triglyceride (TG) levels were observed in fish of HSC (2.5 mg L-1 MWCNTs), indicating abnormal lipid metabolism. Subsequently, the livers of fish in the HSC group were harvested for LC-MS/MS-based targeted lipidomic analysis. Changes in the lipid profiles showed 87 and 92 lipids significant up-regulated and down-regulated, respectively. These significantly altered lipids were classed into 20 lipid classes. Among them, Cer, Hex1Cer, CerG2GNAc1, SM, SPH, and phSM belong to sphingolipid. Taken together with our previous findings, these results suggested that the dysregulation of sphingolipid metabolism may be associated with oxidative stress and apoptosis induced by MWCNTs exposure in C. carpio. This study provides new insights into the potential mechanism of MWCNTs in fish, especially from the aspect of lipid metabolism.
Collapse
Affiliation(s)
- Xiaochan Gao
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, PR China.
| | - Jiaqi Shen
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, PR China.
| | - Pinpin Lv
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, PR China.
| | - Xuehan Niu
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, PR China.
| | - Yong Huang
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471003, PR China.
| |
Collapse
|
6
|
Wang S, Wang P, Ma X, Qiao F, Zhang Z, Li J, Yi H, De Souza C, Zhang L, Lin K. Effects of refrigeration and freezing on milk fat globule membrane lipids in stored breast milk: Insights from non-targeted lipidomics. Food Chem 2025; 468:142478. [PMID: 39700807 DOI: 10.1016/j.foodchem.2024.142478] [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: 09/11/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Breast milk storage provides flexibility for working mothers, though its effects on milk fat globule membrane (MFGM) lipids are not fully understood. This study examined breast milk stored under refrigerated (2 and 4 days) and frozen (7, 14, and 21 days) conditions, finding that these storage durations preserved similar structural characteristics during in vitro gastrointestinal digestion. The analysis focused on MFGM lipid composition under various storage conditions using non-targeted lipidomics. Results revealed that freezing for 14 days reduced phosphatidylserine (PS) levels by 64.55%, while 21 days of freezing significantly decreased both PS (74.59%) and sphingomyelin (SM) (43.85%). Examination of MFGM lipid species indicated that refrigeration for 4 days and freezing for 21 days led to notable reductions in key MFGM lipid species. Thus, breast milk should be refrigerated for up to 4 days to minimize nutrient depletion or frozen for no more than 7 days.
Collapse
Affiliation(s)
- Shaolei Wang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Pengyue Wang
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | - Xia Ma
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | - Fengzhi Qiao
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Zhe Zhang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Jiadong Li
- Innochina Biotech Co., Ltd, Shanghai 201400, China
| | - Huaxi Yi
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Cristabelle De Souza
- Department of Stem Cell Research and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Lanwei Zhang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| | - Kai Lin
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
7
|
Zhang X, Zhao S, Ma Y, Kang W, Zhou W, Zhang C, Abliz Z. Lipidomic profiling of the febrile rat hypothalamus by the intervention of Artemisia japonica extracts. J Pharm Biomed Anal 2025; 255:116588. [PMID: 39644683 DOI: 10.1016/j.jpba.2024.116588] [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: 08/31/2024] [Revised: 11/13/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Artemisia species have been regarded as an important source of ethnic medicinal plants, such as A. annua and A. capillaris, both of which are widely used in clinical treatment. The clinical efficacy of A. japonica is similar to that of A. capillaris, but fewer pharmaceutical studies have been reported. Given that the extracts of A. japonica were observed to reduce the rectal temperature of febrile rats induced by LPS, this study was designed to demonstrate this regulatory effect of the extracts, with a particular focus on the lipidomic profiling of the febrile rat hypothalamus. A total of 72 differential metabolites were filtered out and the association between lipid profiling and potential mechanism was explored. Sphingolipid, glycerophospholipid, arachidonic acid and ether lipid metabolism pathways were significantly enriched. TNF-α, IL-6 and PGE2 cytokines in the hypothalamus were significantly downregulated by the intervention of the extracts of A. japonica. Enzymatic reaction enrichment analysis suggested that PEMT and COX-2 might be potential targets of the efficacy, and which were testified to be downregulated by the ELISA assay under the extracts intervention.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), State Ethnic Affairs Commission, Beijing 100081, China; College of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Suqing Zhao
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), State Ethnic Affairs Commission, Beijing 100081, China; College of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Yuxue Ma
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), State Ethnic Affairs Commission, Beijing 100081, China; College of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Wen Kang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), State Ethnic Affairs Commission, Beijing 100081, China; College of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Wenbin Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), State Ethnic Affairs Commission, Beijing 100081, China; College of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Chen Zhang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), State Ethnic Affairs Commission, Beijing 100081, China; Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China.
| | - Zeper Abliz
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), State Ethnic Affairs Commission, Beijing 100081, China; College of Pharmacy, Minzu University of China, Beijing 100081, China; Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China; Key Laboratory of Ethnomedicine of Ministry of Education, Minzu University of China, Beijing 100081, China.
| |
Collapse
|
8
|
Sima N, Ayllon-Hermida A, Fernández-Becerra C, del Portillo HA. Extracellular vesicles in malaria: proteomics insights, in vitro and in vivo studies indicate the need for transitioning to natural human infections. mBio 2025; 16:e0230424. [PMID: 39868784 PMCID: PMC11898581 DOI: 10.1128/mbio.02304-24] [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] [Indexed: 01/28/2025] Open
Abstract
Globally, an estimated 2.1 billion malaria cases and 11.7 million malaria deaths were averted in the period 2000-2022. Noticeably, despite effective control measurements, in 2022 there were an estimated 249 million malaria cases in 85 malaria-endemic countries and an increase of 5 million cases compared with 2021. Further understanding the biology, epidemiology, and pathogenesis of human malaria is therefore essential for achieving malaria elimination. Extracellular vesicles (EVs) are membrane-enclosed nanoparticles pivotal in intercellular communication and secreted by all cell types. Here, we will review what is currently known about EVs in malaria, from biogenesis and cargo to molecular insights of pathophysiology. Of relevance, a meta-analysis of proteomics cargo, and comparisons between in vitro and in vivo human studies revealed striking differences with those few studies reported from patients. Thus, indicating the need for rigor standardization of methodologies and for transitioning to human infections to elucidate their physiological role. We conclude with a focus on translational aspects in diagnosis and vaccine development and highlight key gaps in the knowledge of EVs in malaria research.
Collapse
Affiliation(s)
- Núria Sima
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
| | - Alberto Ayllon-Hermida
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
- School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Carmen Fernández-Becerra
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Hernando A. del Portillo
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
9
|
Galande S, Ravikanth VV, Tokala RK, Satyanarayana Singh S, Rao GV, Talukdar R, Peddapulla C, Reddy DN, Sasikala M. Transcriptomic analysis of pancreatic tissue from humans and mice identifies potential gene signatures and unexplored pathways during progression from acute to chronic pancreatitis. Gene 2025; 940:149200. [PMID: 39732348 DOI: 10.1016/j.gene.2024.149200] [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: 07/10/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND A comprehensive understanding of the molecular pathogenesis of chronic pancreatitis (CP), a fibroinflammatory disorder of the pancreas, is warranted for the development of targeted therapies. The current study focused on comparing the transcriptomes of pancreatic tissues obtained from patients with CP with those of two rodent models of chemically induced CP to identify dysregulated genes/signaling pathways. METHODS Pancreatitis was induced in mice using cerulein and L-arginine. Pancreatic tissues were obtained from humans and mice. The RNA was isolated, and the transcriptomes were generated using the GeneChip Human Transcriptome Array 2.0 and Clariom D Mouse Array respectively. Differentially expressed genes with log2-fold changes ≥ +2 and ≤ -2 were considered for functional and signaling pathway enrichment analysis. The expression of NUCB2, which plays a role in β-cell function, was validated by ELISA in acute pancreatitis (AP) and immune cell responses in AP and CP using flow cytometry. RESULTS The current study identifies L-arginine-induced CP as a better model for investigating the pathogenesis of human CP, with greater similarity in dysregulated genes (22%), transcription factors (34%) and enriched pathways (58%) compared to cerulein model (2%, 11% and 9%) respectively. Nesfatin-1, encoded by NUCB2, was decreased in patients with AP (12% nondiabetic, 41% post pancreatitis diabetes). The Th1 immune cell response was greater in the patients with AP (44%), whereas Th17 immune response was greater in patients with CP (18%). CONCLUSION Our study highlights potential novel and unexplored pathways involved in inflammation, fibrosis, and pain in CP and paves the way for testing them as putative drug targets using a severe disease model.
Collapse
Affiliation(s)
- Sheethal Galande
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | - V V Ravikanth
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | - Ranjeet K Tokala
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | | | - G V Rao
- Department of Surgical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Rupjyoti Talukdar
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Chandan Peddapulla
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - D Nageshwar Reddy
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Mitnala Sasikala
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India.
| |
Collapse
|
10
|
Yasuda T, Ueura D, Nakagomi M, Hanashima S, Murata M. Relevance of ceramide 1-phosphate domain formation in activation of cytosolic phospholipase A 2. Biophys Chem 2025; 322:107433. [PMID: 40080927 DOI: 10.1016/j.bpc.2025.107433] [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: 01/11/2025] [Revised: 02/23/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Ceramide 1-phosphate (C1P), as a lipid mediator, specifically binds and activates cytosolic phospholipase A2α (cPLA2α). Previous findings revealed that modification of the specific hydrophobic moiety decreases the affinity with cPLA2α. However, the possible biological role of the temporal C1P-enriched domains formed in biomembranes for the molecular recognition of cPLA2α has not been fully elucidated. In this study we elucidated the properties of segregated domains formed by C1P (and its analogs) and the affinity of cPLA2α for C1P in different co-lipid environments by fluorescence spectroscopy using trans-parinaric acid and surface plasmon resonance (SPR). Fluorescence measurements suggested that the formation of C1P ordered domains is strongly influenced by interfacial 3-OH and phosphate groups of C1P, such as hydrogen-bonding and electrostatic interactions, and depends on the co-lipid composition of the host bilayer. SPR indicated that C1P under the lipid environment favorable for the formation of C1P clusters has higher affinity for cPLA2α. Thus, we speculate that C1P clusters formed under certain membrane conditions are important in specific binding with cPLA2α by increasing the interaction between the C1P headgroup and basic residues of cPLA2α. In conclusion, this study revealed that the local formation of lipid mediator-rich clusters in biomembranes likely has a significant effect on the interaction between the mediator and its receptor protein.
Collapse
Affiliation(s)
- Tomokazu Yasuda
- Research Foundation ITSUU Laboratory, C1232, Kanagawa Science Park R&D Building, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan; Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan.
| | - Daiki Ueura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Madoka Nakagomi
- Research Foundation ITSUU Laboratory, C1232, Kanagawa Science Park R&D Building, 3-2-1 Sakado, Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan
| | - Shinya Hanashima
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Graduate School of Engineering, Tottori University, 4-101 Koyamacho-minami, Tottori, Tottori 680-8550, Japan
| | - Michio Murata
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan; Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Hodgkiss R, Acharjee A. Unravelling metabolite-microbiome interactions in inflammatory bowel disease through AI and interaction-based modelling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167618. [PMID: 39662756 DOI: 10.1016/j.bbadis.2024.167618] [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: 09/20/2024] [Revised: 11/20/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
Inflammatory Bowel Diseases (IBDs) are chronic inflammatory disorders of the gastrointestinal tract and colon affecting approximately 7 million individuals worldwide. The knowledge of specific pathology and aetiological mechanisms leading to IBD is limited, however a reduced immune system, antibiotic use and reserved diet may initiate symptoms. Dysbiosis of the gut microbiome, and consequently a varied composition of the metabolome, has been extensively linked to these risk factors and IBD. Metagenomic sequencing and liquid-chromatography mass spectrometry (LC-MS) of N = 220 fecal samples by Fransoza et al., provided abundance data on microbial genera and metabolites for use in this study. Identification of differentially abundant microbes and metabolites was performed using a Wilcoxon test, followed by feature selection of random forest (RF), gradient-boosting (XGBoost) and least absolute shrinkage operator (LASSO) models. The performance of these features was then validated using RF models on the Human Microbiome Project 2 (HMP2) dataset and a microbial community (MICOM) model was utilised to predict and interpret the interactions between key microbes and metabolites. The Flavronifractor genus and microbes of the families Lachnospiraceae and Oscillospiraceae were found differential by all models. Metabolic pathways commonly influenced by such microbes in IBD were CoA biosynthesis, bile acid metabolism and amino acid production and degradation. This study highlights distinct interactive microbiome and metabolome profiles within IBD and the highly potential pathways causing disease pathology. It therefore paves way for future investigation into new therapeutic targets and non-invasive diagnostic tools for IBD.
Collapse
Affiliation(s)
- Rebecca Hodgkiss
- College of Medicine and Health, Cancer and Genomic Sciences, University of Birmingham, B15 2TT Birmingham, UK
| | - Animesh Acharjee
- College of Medicine and Health, Cancer and Genomic Sciences, University of Birmingham, B15 2TT Birmingham, UK; Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, B15 2TT Birmingham, UK; MRC Health Data Research UK (HDR), Midlands Site, UK; Centre for Health Data Research, University of Birmingham, B15 2TT, UK.
| |
Collapse
|
12
|
García-Hermoso A, Huerta-Uribe N, Izquierdo M, González-Ruíz K, Correa-Bautista JE, Ramírez-Vélez R. Comparative lipidomic profiling in adolescents with obesity and adolescents with type 1 diabetes. Curr Probl Cardiol 2025; 50:102991. [PMID: 39828111 DOI: 10.1016/j.cpcardiol.2025.102991] [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: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Both adolescents with obesity and those with type 1 diabetes (T1D) exhibit alterations in lipid profiles, but direct comparisons are limited. Comparing lipidomic profiles between obese individuals and those with T1D is crucial for identifying specific metabolic markers, informing tailored interventions, and advancing precision medicine strategies for these distinct populations. The aim of the study was to compare lipidomic profiles between adolescents with obesity and those with T1D, and to analyze associations between metabolites and clinical parameters. METHODS We included 156 adolescents aged 11-18 years (59.6% girls) from the HEPAFIT (n=114, obesity) and Diactive-1 Cohort (n=42, T1D) studies. Clinical measures included anthropometrics, body composition, lipids, liver enzymes, glucose, and HbA1c. Lipidomic analysis of 277 serum/plasma metabolites used UHPLC-MS. RESULTS Distinct lipid profiles were seen, with higher diglycerides, triglycerides, and certain phosphatidylinositols in the obesity group, while phosphatidylcholines, phosphatidylethanolamines, cholesterol esters, sphingomyelins, and ceramides were elevated in T1D. Triglycerides acyl chain lengths and saturation levels also varied. Multivariate analysis identified seven metabolites -PC(O-18:1/18:1), PC(O-18:1/22:4), PE(O-16:0/18:1), PE(18:2e/22:6), PC(40:1), PC(O-22:1/20:4), and PE(P-18:0/18:1)- significantly associated with clinical parameters. CONCLUSIONS Distinct lipid profiles were observed among adolescents with obesity and T1D in the study, emphasizing the importance of understanding specific metabolite associations with clinical parameters for more precise health management.
Collapse
Affiliation(s)
- Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.
| | - Nidia Huerta-Uribe
- Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Robinson Ramírez-Vélez
- Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
13
|
Xuan X, Huang Z, Kong Z, Li R, Li J, Huang H. GENETIC INSIGHTS INTO SEPSIS: MENDELIAN RANDOMIZATION ANALYSIS OF CEREBROSPINAL FLUID METABOLITES. Shock 2025; 63:379-384. [PMID: 39454631 DOI: 10.1097/shk.0000000000002494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
ABSTRACT Background: Sepsis, a life-threatening response to infection leading to systemic inflammation and organ dysfunction, has been hypothesized to be influenced by metabolic alterations in cerebrospinal fluid (CSF). Despite extensive research, the specific metabolic pathways contributing to sepsis remain unclear. This study aims to elucidate the causal relationships between CSF metabolites and sepsis risk using Mendelian randomization (MR), offering insights that could lead to novel therapeutic strategies. Methods: We conducted a two-sample MR analysis using genetic variants as instrumental variables (IVs) to investigate 338 CSF metabolites identified through a genome-wide association study. Data on sepsis-related outcomes were extracted from the genome-wide association study catalog encompassing 486,484 individuals of European descent. IVs were rigorously selected based on stringent genetic association and linkage disequilibrium criteria. Statistical analyses, including inverse variance weighting (IVW) and weighted median methods, were performed using the "TwoSampleMR" package in R software, supplemented by comprehensive sensitivity analyses to ensure the robustness of our findings. Results: Our analysis identified 19 CSF metabolites causally associated with sepsis risk. Notably, metabolites such as 1-palmitoyl-2-stearoyl-gpc (16:0/18:0) and 2-hydroxyglutarate showed significant negative correlations with sepsis risk. The reverse MR analysis further revealed that sepsis could negatively impact certain CSF metabolite levels, particularly ribonate, suggesting a bidirectional relationship. These relationships were substantiated by rigorous statistical testing and sensitivity analyses confirming the absence of horizontal pleiotropy and the stability of our results across various MR methods. Conclusions: This study demonstrates significant causal associations between specific CSF metabolites and the risk of developing sepsis, highlighting the potential for these metabolites to serve as biomarkers or therapeutic targets. The bidirectional nature of these findings also suggests that sepsis itself may alter metabolic profiles, offering further avenues for intervention.
Collapse
Affiliation(s)
- Xin Xuan
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Zhihao Huang
- Department of Otorhinolaryngology, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Zhiqian Kong
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Ruoyu Li
- Department of Intensive Care Unit, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Jianfeng Li
- Department of Emergency Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| | - Haiyan Huang
- Department of Internal Medicine of Chinese Medicine, Dongguan Hospital of Guangzhou University of Traditional Chinese Medicine, Dongguan, Guangdong, China
| |
Collapse
|
14
|
Felippe TVD, Toro DM, de Carvalho JCS, Nobre-Azevedo P, Rodrigues LFM, Oliveira BTM, da Silva-Neto PV, Vilela AFL, Almeida F, Faccioli LH, Sorgi CA. High-resolution targeted mass spectrometry for comprehensive quantification of sphingolipids: clinical applications and characterization of extracellular vesicles. Anal Biochem 2025; 698:115732. [PMID: 39622401 DOI: 10.1016/j.ab.2024.115732] [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: 10/16/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Sphingolipids (SL), a class of membrane lipids, play important roles in numerous biological processes. Their significant structural diversity poses challenges for accurate quantification. To address this, liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) has emerged as a powerful tool for sphingolipidomics, capable of profiling these lipids comprehensively. In this study, we utilized LC-MS/MS with high-resolution mass spectrometry (MRMHR) to develop a targeted method for the identification and quantification of various SL species. This method, based on validated parameters such as precursor/fragment ions (m/z) and retention time, demonstrated high sensitivity and accuracy, successfully identifying SL species across 12 distinct classes. Its open-panel design also facilitates the analysis of new SL-species targets. Notably, using this approach, we identified 40 SL species in plasma samples from COVID-19 patients, and we determined the influence of matrix metalloproteinase-3 (MMP-3) expression on the positive downstream of SL metabolism. Beyond plasma analysis, this method has potential applications in other biomedical contexts, such as extracellular vesicles (EVs), describing the cargo of sphingosine-1-phosphate (S1P) on macrophage-derived EVs. The establishment of this targeted workflow enabling precise quantification of a wide range of SL species, holds promise for identifying novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Thiago V D Felippe
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil
| | - Diana M Toro
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada - PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas - UFAM, Manaus, 69080-900, AM, Brazil
| | - Jonatan C S de Carvalho
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil
| | - Pedro Nobre-Azevedo
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil
| | - Luiz F M Rodrigues
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil
| | - Bianca T M Oliveira
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil
| | - Pedro V da Silva-Neto
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada - PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas - UFAM, Manaus, 69080-900, AM, Brazil
| | - Adriana F L Vilela
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil
| | - Fausto Almeida
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil
| | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil; Centro de Excelência em Quantificação e Identificação de Lipídios (CEQIL), Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil
| | - Carlos A Sorgi
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto - FFCLRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-901, SP, Brazil; Programa de Pós-Graduação em Imunologia Básica e Aplicada - PPGIBA, Instituto de Ciências Biológicas, Universidade Federal do Amazonas - UFAM, Manaus, 69080-900, AM, Brazil; Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto - FMRP, Universidade de São Paulo-USP, Ribeirão Preto, 14049-900, SP, Brazil; Centro de Excelência em Quantificação e Identificação de Lipídios (CEQIL), Faculdade de Ciências Farmacêuticas de Ribeirão Preto - FCFRP, Universidade de São Paulo-USP, Ribeirão Preto, 14040-903, SP, Brazil.
| |
Collapse
|
15
|
Justa HCD, Baldissera AB, Machado MI, Souza SH, Polli NLC, Boia-Ferreira M, Schluga PHDC, Donatti L, Wille ACM, Minozzo JC, Gremski LH, Veiga SS. Induction of ectosome formation by binding of phospholipases D from Loxosceles venoms to endothelial cell surface: Mechanism of interaction. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159579. [PMID: 39547302 DOI: 10.1016/j.bbalip.2024.159579] [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: 07/05/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Members of the phospholipase D (PLD) superfamily found in Loxosceles spider venoms are potent toxins with inflammatory and necrotizing activities. They degrade phospholipids in cell membranes, generating bioactive molecules that activate skin cells. These skin cells, in turn, activate leukocytes involved in dermonecrosis, characterized by aseptic coagulative necrosis. Although the literature has advanced in understanding the structure-function relationship, the cell biology resulting from the interactions of these molecules with cells remains poorly understood. In this study, we show that different cells exposed to recombinant PLDs bind these molecules to their plasma membrane, leading to the subsequent organization of extracellular microvesicles/ectosomes. The binding occurs as quickly as five minutes or less after exposure, increases over time, and eventually, the PLDs are expelled from the cell surface without generating cytotoxicity. PLDs are not endocytosed, nor do they spatially colocalize with acidic organelles in the intracellular environment. At least two regions of PLDs - the domain involved in magnesium ion coordination and the choline binding site - appear to play a role in cell surface binding and ectosome organization. However, the amino acids involved in catalysis do not participate in these events. The binding of these PLDs to the cell membrane, independent of catalytic activity, is sufficient to trigger intracellular signaling and enhance the expression of the pro-inflammatory IL-8 gene. These results are supported by the observation that isoforms of PLDs lacking catalytic activity induce an inflammatory response in vivo when injected into the skin of rabbits, without causing dermonecrosis. Our data indicate that these PLDs bind to the surface of target cells, promoting the organization of extracellular vesicles/ectosomes. Subsequently, these events activate pro-inflammatory genes and induce an inflammatory response in vivo. The binding to cells is not dependent on amino acids involved in catalysis but rather on amino acids involved in magnesium coordination. The binding of PLDs to the cell surface, formation of ectosomes, and activation of cells appear to initiate signals involved in inflammatory responses that can lead to dermonecrosis in accidents. This correlation is supported by experimental observations indicating that the events of toxin binding to cells, formation of microvesicles, and inflammatory responses observed both in vitro and in vivo are interconnected.
Collapse
Affiliation(s)
- Hanna Câmara da Justa
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | | | - Samira Hajjar Souza
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | - Marianna Boia-Ferreira
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | - Lucelia Donatti
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | - Ana Carolina M Wille
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa (UEPG), Ponta Grossa 84030-900, Brazil
| | - João Carlos Minozzo
- Production and Research Center of Immunobiological Products (CPPI), State Department of Health, Piraquara 83302-200, Brazil
| | - Luiza Helena Gremski
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | - Silvio S Veiga
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil.
| |
Collapse
|
16
|
Xia F, Santacruz A, Wu D, Bertho S, Fritz E, Morales-Sosa P, McKinney S, Nowotarski SH, Rohner N. Reproductive Adaptation of Astyanax mexicanus Under Nutrient Limitation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638191. [PMID: 40027826 PMCID: PMC11870393 DOI: 10.1101/2025.02.13.638191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Reproduction is a fundamental biological process for the survival and continuity of species. Examining changes in reproductive strategies offers valuable insights into how animals have adapted their life histories to different environments. Since reproduction is one of the most energy-intensive processes in female animals, nutrient scarcity is expected to interfere with the ability to invest in gametes. Lately, a new model to study adaptation to nutrient limitation has emerged; the Mexican tetra Astyanax mexicanus . This fish species exists as two different morphs, a surface river morph and a cave-dwelling morph. The cave-dwelling morph has adapted to the dark, biodiversity, and nutrient-limited cave environment and consequently evolved an impressive starvation resistance. However, how reproductive strategies have adapted to nutrient limitations in this species remains poorly understood. Here, we compared breeding activities and maternal contributions between laboratory-raised surface fish and cavefish. We found that cavefish produce different clutch sizes of eggs with larger yolk compared to surface fish, indicating a greater maternal nutrient deposition in cavefish embryos. To systematically characterize yolk compositions, we used untargeted proteomics and lipidomics approaches to analyze protein and lipid profiles in 2-cell stage embryos and found an increased proportion of sphingolipids in cavefish compared to surface fish. Additionally, we generated transcriptomic profiles of surface fish and cavefish ovaries using a combination of single cell and bulk RNA sequencing to examine differences in maternal contribution. We found that genes essential for hormone regulation were upregulated in cavefish follicular somatic cells compared to surface fish. To evaluate whether these differences contribute to their reproductive abilities under natural-occurring stress, we induced breeding in starved female fish. Remarkably, cavefish maintained their ability to breed under starvation, whereas surface fish largely lost this ability. We identified insulin-like growth factor 1a receptor ( igf1ra ) as a potential candidate gene mediating the downregulation of ovarian development genes, potentially contributing to the starvation-resistant fertility of cavefish. Taken together, we investigated the female reproductive strategies in Astyanax mexicanus , which will provide fundamental insights into the adaptations of animals to environments with extreme nutrient deficit.
Collapse
|
17
|
Duan J, Sun J, Ma X, Du P, Dong P, Xue J, Lu Y, Jiang T. Association of escitalopram-induced shifts in gut microbiota and sphingolipid metabolism with depression-like behavior in wistar-kyoto rats. Transl Psychiatry 2025; 15:54. [PMID: 39962083 PMCID: PMC11833111 DOI: 10.1038/s41398-025-03277-8] [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: 07/06/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
The microbiota-gut-brain axis plays a pivotal role in neuropsychiatric disorders, particularly in depression. Escitalopram (ESC) is a first-line antidepressant, however, its regulatory mechanisms on the microbiota-gut-brain axis in the treatment of depression remain unclear. The antidepressant effects of ESC were evaluated using the forced swim test in Wistar-Kyoto (WKY) rats, while damage in the gut and brain regions was assessed through H&E staining and immunohistochemistry. The therapeutic mechanisms in WKY rats with depression-like behavior were investigated through 16S rRNA sequencing of the gut microbiota, serum untargeted metabolomics, and hippocampal proteomics. Results indicated that ESC intervention improved depressive-like behaviors, as evidenced by increased swimming times in WKY rats, and also restored intestinal permeability and brain tissue integrity. Significant changes in the gut microbiota composition, particularly an increase in Bacteroides barnesiae, as well as increases in serum sphingolipid metabolites (Sphinganine 1-phosphate, Sphingosine, and Sphingosine-1-phosphate) and hippocampal proteins (Sptlc1, Enpp5, Enpp2), were strongly correlated. These robust correlations suggest that ESC may exert its antidepressant effects by modulating sphingolipid metabolism through the influence of gut microbiota. Accordingly, this research elucidates novel mechanisms underlying the antidepressant efficacy of ESC and highlights the pivotal importance of the microbiota-gut-brain axis in mediating these effects.
Collapse
Affiliation(s)
- Jiajia Duan
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jiaxing Sun
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiao Ma
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Peipei Du
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Pengfei Dong
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Juan Xue
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yanli Lu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Tao Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
18
|
Cozzolino KA, Sanford L, Hunter S, Molison K, Erickson B, Courvan MCS, Jones T, Ajit D, Galbraith MD, Espinosa JM, Bentley D, Allen MA, Dowell RD, Taatjes DJ. Mediator kinase inhibition suppresses hyperactive interferon signaling in Down syndrome. eLife 2025; 13:RP100197. [PMID: 39928031 PMCID: PMC11810109 DOI: 10.7554/elife.100197] [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] [Indexed: 02/11/2025] Open
Abstract
Hyperactive interferon (IFN) signaling is a hallmark of Down syndrome (DS), a condition caused by Trisomy 21 (T21); strategies that normalize IFN signaling could benefit this population. Mediator-associated kinases CDK8 and CDK19 drive inflammatory responses through incompletely understood mechanisms. Using sibling-matched cell lines with/without T21, we investigated Mediator kinase function in the context of hyperactive IFN in DS over a 75 min to 24 hr timeframe. Activation of IFN-response genes was suppressed in cells treated with the CDK8/CDK19 inhibitor cortistatin A (CA), via rapid suppression of IFN-responsive transcription factor (TF) activity. We also discovered that CDK8/CDK19 affect splicing, a novel means by which Mediator kinases control gene expression. To further probe Mediator kinase function, we completed cytokine screens and metabolomics experiments. Cytokines are master regulators of inflammatory responses; by screening 105 different cytokine proteins, we show that Mediator kinases help drive IFN-dependent cytokine responses at least in part through transcriptional regulation of cytokine genes and receptors. Metabolomics revealed that Mediator kinase inhibition altered core metabolic pathways in cell type-specific ways, and broad upregulation of anti-inflammatory lipid mediators occurred specifically in kinase-inhibited cells during hyperactive IFNγ signaling. A subset of these lipids (e.g. oleamide, desmosterol) serve as ligands for nuclear receptors PPAR and LXR, and activation of these receptors occurred specifically during hyperactive IFN signaling in CA-treated cells, revealing mechanistic links between Mediator kinases, lipid metabolism, and nuclear receptor function. Collectively, our results establish CDK8/CDK19 as context-specific metabolic regulators, and reveal that these kinases control gene expression not only via TFs, but also through metabolic changes and splicing. Moreover, we establish that Mediator kinase inhibition antagonizes IFN signaling through transcriptional, metabolic, and cytokine responses, with implications for DS and other chronic inflammatory conditions.
Collapse
Affiliation(s)
- Kira A Cozzolino
- Department of Biochemistry, University of ColoradoBoulderUnited States
| | - Lynn Sanford
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Samuel Hunter
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Kayla Molison
- Department of Biochemistry, University of ColoradoBoulderUnited States
| | - Benjamin Erickson
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
- UC-Denver RNA Bioscience InitiativeAuroraUnited States
| | - Meaghan CS Courvan
- Department of Biochemistry, University of ColoradoBoulderUnited States
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
- Crnic Institute Boulder BranchBoulderUnited States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Taylor Jones
- Department of Biochemistry, University of ColoradoBoulderUnited States
| | - Deepa Ajit
- Metabolon Inc, DurhamMorrisvilleUnited States
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - David Bentley
- Department of Biochemistry and Molecular Genetics, University of Colorado School of MedicineAuroraUnited States
- UC-Denver RNA Bioscience InitiativeAuroraUnited States
| | - Mary Ann Allen
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Robin D Dowell
- Department of Molecular, Cellular, and Developmental Biology, University of ColoradoBoulderUnited States
- BioFrontiers Institute, University of ColoradoBoulderUnited States
| | - Dylan J Taatjes
- Department of Biochemistry, University of ColoradoBoulderUnited States
| |
Collapse
|
19
|
Zhao F, Shao M, Li M, Li T, Zheng Y, Sun W, Ni C, Li L. Sphingolipid metabolites involved in the pathogenesis of atherosclerosis: perspectives on sphingolipids in atherosclerosis. Cell Mol Biol Lett 2025; 30:18. [PMID: 39920588 PMCID: PMC11804087 DOI: 10.1186/s11658-024-00679-2] [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: 07/29/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025] Open
Abstract
Atherosclerosis, with its complex pathogenesis, is a leading underlying cause of many cardiovascular diseases, which are increasingly prevalent in the population. Sphingolipids play an important role in the development of atherosclerosis. Key metabolites and enzymes in sphingolipid metabolism influence the pathogenesis of atherosclerosis in a variety of ways, including inflammatory responses and oxidative stress. Thus, an investigation of sphingolipid metabolism-related metabolites and key enzymes may provide novel insights and treatment targets for atherosclerosis. This review discusses various mechanisms and research progress on the relationship between various sphingolipid metabolites, related enzymes, and atherosclerosis. Finally, we look into the future research direction of phytosphingolipids.
Collapse
Affiliation(s)
- Fufangyu Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mingyan Shao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingrui Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianxing Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yanfei Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China.
| | - Cheng Ni
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Lingru Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
20
|
Parthasarathy G, Venkatesan N, Sidhu GS, Song MJ, Liao CY, Barrow F, Mauer A, Sehrawat T, Nakao Y, Daniel PV, Dasgupta D, Pavelko K, Revelo XS, Malhi H. Deletion of sphingosine 1-phosphate receptor 1 in myeloid cells reduces hepatic inflammatory macrophages and attenuates MASH. Hepatol Commun 2025; 9:e0613. [PMID: 39899672 DOI: 10.1097/hc9.0000000000000613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/02/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Immune cell-driven inflammation is a key mediator of metabolic dysfunction-associated steatohepatitis (MASH) progression. We have previously demonstrated that pharmacological sphingosine 1-phosphate (S1P) receptor modulation ameliorates MASH and is associated with attenuated accumulation of intrahepatic macrophage and T-cell subsets. Although S1P receptors are expressed on several immune cell types, given the prominent role of monocyte-derived recruited macrophages in the sterile inflammation of MASH, we hypothesized that deletion of S1P receptor 1 (S1P1) on myeloid cells may ameliorate MASH by reducing the accumulation of proinflammatory monocyte-derived macrophages in the liver. METHODS The LyzMCre approach was used to generate myeloid cell-specific knockout mice, termed S1pr1MKO. Littermate S1pr1loxp/loxp mice were used as wild-type controls. MASH was established by feeding mice a high-fat, -fructose, and -cholesterol (FFC) diet for 24 weeks, which led to the development of steatohepatitis and MASH-defining cardiometabolic risk factors. Liver injury and inflammation were determined by histological and gene expression analyses. Intrahepatic leukocyte populations were analyzed by mass cytometry and immunohistochemistry. RESULTS Histological examination demonstrated a reduction in liver inflammatory infiltrates and fibrosis in high-fat, -fructose, and -cholesterol-fed S1pr1MKO compared to wild-type. There was a corresponding reduction in alanine aminotransferase, a sensitive marker for liver injury. As determined by mass cytometry, a significant decrease in recruited macrophages was noted in the livers of high-fat, -fructose, and -cholesterol-fed S1pr1MKO mice compared to wild-type. Gene ontology pathway analysis revealed significant suppression of the peroxisome proliferator-activated receptor gamma and mitogen-activated protein kinase pathways in S1pr1MKO consistent with attenuated MASH in mice. CONCLUSIONS Deletion of S1P1 in myeloid cells is sufficient to attenuate intrahepatic accumulation of monocyte-derived macrophages and ameliorate murine MASH.
Collapse
Affiliation(s)
- Gopanandan Parthasarathy
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nanditha Venkatesan
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Guneet Singh Sidhu
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Myeong Jun Song
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chieh-Yu Liao
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Fanta Barrow
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Amy Mauer
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tejasav Sehrawat
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yasuhiko Nakao
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - P Vineeth Daniel
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Debanjali Dasgupta
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Xavier S Revelo
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
21
|
Wan X, Zhang Y, Zhang K, Mou Y, Jin X, Huang X. The alterations of ocular surface metabolism and the related immunity inflammation in dry eye. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2025; 5:1-12. [PMID: 39758836 PMCID: PMC11699629 DOI: 10.1016/j.aopr.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 01/07/2025]
Abstract
Background Dry eye disease (DED) stands as a prominent ocular condition of global prevalence, emerging as a growing concern within public health. However, the underlying mechanisms involved in its pathogenesis remain largely unknown. In recent years, with the development of metabolomics, numerous studies have reported alterations in ocular surface metabolism in DED and offered fresh perspectives on the development of DED. Main text The metabolic changes of the ocular surface of DED patients are closely intertwined with the cellular metabolism process and immune inflammation changes. This article expounds upon the correlation between ocular surface metabolism and immune inflammation alterations in DED in terms of glycolysis, lipid metabolism, amino acid metabolism, cellular signaling pathways, and immune inflammation regulation. Conclusions The alterations in ocular surface metabolism of patients with dry eye are closely associated with their inflammatory status. Our work contributes novel insights into the pathogenesis of dry eye diseases and offers innovative molecular targets for diagnosing, detecting, and managing DED patients.
Collapse
Affiliation(s)
- Xiaojie Wan
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Yu Zhang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Kaiye Zhang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Yujie Mou
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Xiuming Jin
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Xiaodan Huang
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| |
Collapse
|
22
|
Li X, Duan W, Zhu Y, Ji R, Feng K, Kathuria Y, Xiao H, Yu Y, Cao Y. Transcriptomics and metabolomics reveal the alleviation effect of pectic polysaccharide on dextran sodium sulfate-induced colitis mice. Int J Biol Macromol 2025; 288:138755. [PMID: 39674473 DOI: 10.1016/j.ijbiomac.2024.138755] [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: 10/03/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Ulcerative colitis (UC) is a relapsing disease with an increasing morbidity and prevalence. Dietary polysaccharides have recently become a research hotspot because of their therapeutic effects and safety on UC. Our previous research elucidated that pectic polysaccharide from Phyllanthus emblica L. (PEP-1) could alleviate dextran sodium sulfate-induced UC mice. Herein, metabolomics and transcriptomics were further applied to disclose the underlying mechanisms behind PEP-1's anti-inflammatory effects. PEP-1 intervention altered the serum metabolite contents and pathways represented by decreasing xanthine and sphinganine levels. Changes in gene expressions correlated with metabolite variations led by the suppression of the expression of the inflammatory factors, colorectal cancer promoter, and NF-κB pathway as well as the enhancement of tight junctions. This study demonstrated that the ameliorating effect of chronic UC was partially ascribed to the alteration of the serum metabolites and changes in gene expression.
Collapse
Affiliation(s)
- Xiaoqing Li
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Wen Duan
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yi Zhu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Ruya Ji
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Konglong Feng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Foshan University, Foshan 528000, China
| | - Yukti Kathuria
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA.
| | - Yigang Yu
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China.
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
23
|
Jawale D, Khandibharad S, Singh S. Innate Immune Response and Epigenetic Regulation: A Closely Intertwined Tale in Inflammation. Adv Biol (Weinh) 2025; 9:e2400278. [PMID: 39267219 DOI: 10.1002/adbi.202400278] [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: 05/23/2024] [Revised: 07/08/2024] [Indexed: 09/17/2024]
Abstract
Maintenance of delicate homeostasis is very important in various diseases because it ensures appropriate immune surveillance against pathogens and prevents excessive inflammation. In a disturbed homeostatic condition, hyperactivation of immune cells takes place and interplay between these cells triggers a plethora of signaling pathways, releasing various pro-inflammatory cytokines such as Tumor necrosis factor alpha (TNFα), Interferon-gamma (IFNƴ), Interleukin-6 (IL-6), and Interleukin-1 beta (IL-1β), which marks cytokine storm formation. To be precise, dysregulated balance can impede or increase susceptibility to various pathogens. Pathogens have the ability to hijack the host immune system by interfering with the host's chromatin architecture for their survival and replication in the host cell. Cytokines, particularly IL-6, Interleukin-17 (IL-17), and Interleukin-23 (IL-23), play a key role in orchestrating innate immune responses and shaping adaptive immunity. Understanding the interplay between immune response and the role of epigenetic modification to maintain immune homeostasis and the structural aspects of IL-6, IL-17, and IL-23 can be illuminating for a novel therapeutic regimen to treat various infectious diseases. In this review, the light is shed on how the orchestration of epigenetic regulation facilitates immune homeostasis.
Collapse
Affiliation(s)
- Diksha Jawale
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), NCCS Complex, SPPU Campus, Ganeshkhind, Pune, 411007, India
| | - Shweta Khandibharad
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), NCCS Complex, SPPU Campus, Ganeshkhind, Pune, 411007, India
| | - Shailza Singh
- Systems Medicine Laboratory, Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), NCCS Complex, SPPU Campus, Ganeshkhind, Pune, 411007, India
| |
Collapse
|
24
|
Zhang MY, Yin C, Ding L, Cheng L, Lv Q, Wang P, Zhang SB, You QY. Mechanism of Panax notoginseng saponins in improving cognitive impairment induced by chronic sleep deprivation based on the integrative analysis of serum metabolomics and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118988. [PMID: 39447711 DOI: 10.1016/j.jep.2024.118988] [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: 08/03/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax notoginseng saponin (PNS) has a variety of biological activities, such as improvement of myocardial ischemia, improvement of learning and memory, hypolipidemia, and immunomodulation. However, its protective mechanism on the central nervous system (CNS) is not clear. AIM OF THE STUDY The present study initially evaluated the possible mechanism of PNS to improve cognitive dysfunction due to chronic sleep deprivation (CSD). MATERIALS AND METHODS In the present study, we used a modified multi-platform aquatic environment sleep deprivation method to induce a cognitively impaired rat model, and explored the mechanism of action of PNS by integrating serum metabolomics and network pharmacology, which was further verified by molecular docking and experiments. RESULTS The results showed that PNS significantly shortened the escape latency, increased the target quadrant time and the number of traversing platforms, and attenuated the inflammatory damage in the hippocampal Cornu Ammonis 1 (CA1) region in CSD rats. The non-targeted metabolomics results indicated that 35 biomarkers significantly altered following PNS therapy intervention, with metabolic pathways enriched for the effects of One carbon pool by folate, Riboflavin metabolism, Glycerophospholipid metabolism, Sphingolipid metabolism, Glycerolipid metabolism, Arachidonic acid metabolism, and Tryptophan metabolism. In addition, network pharmacology identified 234 potential targets for PNS intervention in CSD with cognitive impairment. Metabolite-response-enzyme-gene network was constructed by MetaScape and matched with the network pharmacology results to identify a total of five shared targets (LPL, GPAM, HSD11B1, HSD11B2, and SULT2A1) and two metabolic pathways (Sphingolipid metabolism and Steroid hormone biosynthesis). The results of molecular docking revealed that the five active ingredients had good binding ability with the five core targets. qPCR analysis confirmed the ability of PNS to modulate the above five targets. CONCLUSIONS The combination of metabolomics and network analysis provides a scientific basis for promoting the clinical application of PNS in cognitive impairment.
Collapse
Affiliation(s)
- Mei-Ya Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Chao Yin
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Li Ding
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Li Cheng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Qing Lv
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Ping Wang
- Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Shun-Bo Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Qiu-Yun You
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
25
|
Patil AS, Xu Y. Comprehensive Metabolomics in Mouse Mast Cell Model of Allergic Rhinitis for Profiling, Modulation, Semiquantitative Analysis, and Pathway Analysis. Biomolecules 2025; 15:109. [PMID: 39858503 PMCID: PMC11763337 DOI: 10.3390/biom15010109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
Allergic rhinitis affects millions globally, causing significant discomfort and reducing the quality of life. This study investigates the metabolic alterations in murine mast cells (MC/9) under allergic rhinitis conditions induced by lipopolysaccharide (LPS) stimulation, employing UHPLC-QTOF-MS-based untargeted and targeted metabolomics. The analysis identified 44 significantly regulated metabolites, including histamine, leukotrienes, prostaglandins, thromboxanes, and ceramides. Key metabolic pathways such as arachidonic acid, histidine, and sphingolipid metabolisms were notably modulated. The study further examined the therapeutic effects of triprolidine and zileuton, demonstrating their capacity to reverse LPS-induced metabolic shifts. Triprolidine primarily modulated histidine and sphingolipid metabolism, while zileuton targeted arachidonic acid and sphingolipid metabolism. These findings underscore the utility of metabolomics analysis in elucidating the complex biochemical pathways involved in allergic rhinitis and highlight the potential of metabolomics for evaluating therapeutic interventions. This study enhances our understanding of mast cell metabolism in allergic responses and provides a robust model for assessing the efficacy of anti-allergic agents, paving the way for more effective treatments.
Collapse
Affiliation(s)
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA;
| |
Collapse
|
26
|
Miyata K, Inoue Y, Yamane M, Honda H. Fish environmental RNA sequencing sensitively captures accumulative stress responses through short-term aquarium sampling. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 959:178182. [PMID: 39719761 DOI: 10.1016/j.scitotenv.2024.178182] [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: 10/15/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
The utility of environmental RNA (eRNA) in capturing biological responses to stresses has been discussed previously; however, the limited number of genes detected remains a significant hindrance to its widespread implementation. Here, we investigated the potential of eRNA to assess the health status of Japanese medaka fish exposed to linear alkylbenzene sulfonate. Analyzing eRNA and organismal RNA (oRNA) in aquarium water within 12 h, we achieved high mapping rates and 10 times more differentially expressed genes than previously reported. This advancement has facilitated the previously unattainable capability of gene ontology (GO) analysis. The GO analysis revealed that eRNA can detect nuclear genes associated with cellular components and reflect cumulative gene expression signatures over time, while oRNA provided short-term gene expression signatures in biological process. Moreover, eRNA exhibited high sensitivity in responding to genes associated with sphingolipid and ceramide biosynthesis, which are involved in inflammatory responses possibly originating from impaired cells. This finding aligns with the observations made in oRNA. In conclusion, eRNA-sequencing (eRNA-seq) using aquarium water emerges as a valuable high sensitivity tool for analyzing physiological stress. The findings of this study lay the foundation for further development of eRNA-seq technologies.
Collapse
Affiliation(s)
- Kaede Miyata
- R&D Safety Science Research, Kao Corporation, Ichikai-Machi, Haga-Gun, Tochigi 321-3497, Japan.
| | - Yasuaki Inoue
- R&D Safety Science Research, Kao Corporation, Ichikai-Machi, Haga-Gun, Tochigi 321-3497, Japan.
| | - Masayuki Yamane
- R&D Safety Science Research, Kao Corporation, Ichikai-Machi, Haga-Gun, Tochigi 321-3497, Japan
| | - Hiroshi Honda
- R&D Safety Science Research, Kao Corporation, Ichikai-Machi, Haga-Gun, Tochigi 321-3497, Japan
| |
Collapse
|
27
|
Wang R, Gan C, Mao R, Chen Y, Yan R, Li G, Xiong T, Guo J. Rat models of postintracerebral hemorrhage pneumonia induced by nasal inoculation with Klebsiella pneumoniae or intratracheal inoculation with LPS. Front Immunol 2025; 15:1477902. [PMID: 39845950 PMCID: PMC11750689 DOI: 10.3389/fimmu.2024.1477902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background A stable and reproducible experimental bacterial pneumonia model postintracerebral hemorrhage (ICH) is necessary to help investigating the pathogenesis and novel treatments of Stroke-associated pneumonia (SAP). Aim To establish a Gram-negative bacterial pneumonia-complicating ICH rat model and an acute lung injury (ALI)-complicating ICH rat model. Methods We established two standardized models of post-ICH pneumonia by nasal inoculation with Klebsiella pneumoniae (Kp) or intratracheal inoculation with lipopolysaccharide (LPS). Survival and neurological scores were monitored. Magnetic resonance imaging was performed to evaluate hematoma volume. Abdominal aortic blood was collected for leukocyte counting, serum was isolated to determine concentrations of S100β and proinflammatory cytokines using ELISAs. Histopathological changes of brain, lung and gut were assessed using hematoxylin-eosin staining. Lung was isolated for immunofluorescence staining for myeloperoxidase (MPO). Bronchoalveolar lavage fluid was collected for leukocyte counting, and supernatant was prepared to measure MPO activity. Ileum was isolated for immunofluorescence staining for tight junction proteins ZO-1 and γδ TCRs/IL-17A and for Alcian blue-nuclear fast red staining of acidic mucins. Feces were collected, 16S rRNA sequencing, untargeted metabolomics and Spearman's correlation analyses were performed to explore changes of gut microbiota, metabolites and their interactions. Results In Kp-induced bacterial pneumonia-complicating ICH rats, we demonstrated that Kp challenge caused more severe neurological deficits, brain damage, neuroinflammation, and aggravated pneumonia and lung injury. Disruptions of the intestinal structure and gut barrier and the reductions of the protective intestinal IL-17A-producing γδT cells were also observed. Kp challenge exacerbated the gut microbiota dysbiosis and fecal metabolic profile disorders, which were characterized by abnormal sphingolipid metabolism especially elevated ceramide levels; increased levels of neurotoxic quinolinic acid and an upregulation of tryptophan (Trp)-serotonin-melatonin pathway. Spearman's correlation analyses further revealed that the reduction or depletion of some beneficial bacteria, such as Allobaculum and Faecalitalea, and the blooming of some opportunistic pathogens, such as Turicibacter, Dietzia, Corynebacterium and Clostridium_sensu_stricto_1 in Kp-induced SAP rats were associated with the disordered sphingolipid and Trp metabolism. Using an LPS-induced ALI complicating ICH model, we also characterized SAP-induced brain, lung and gut histopathology injuries; peripheral immune disorders and intense pulmonary inflammatory responses. Conclusions These two models may be highly useful for investigating the pathogenesis and screening and optimizing potential treatments for SAP. Moreover, the differential genera and sphingolipid or Trp metabolites identified above seem to be promising therapeutic targets.
Collapse
Affiliation(s)
- Ruihua Wang
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changlian Gan
- School of Traditional Dai Medicine, West Yunnan University of Applied Science, Xishuangbanna, China
| | - Rui Mao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Chen
- Department of Bioinformatics, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianqin Xiong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianwen Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Neurology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
28
|
Ke Y, Gannaban R, Liu J, Zhou Y. STIM1 and lipid interactions at ER-PM contact sites. Am J Physiol Cell Physiol 2025; 328:C107-C114. [PMID: 39620863 DOI: 10.1152/ajpcell.00634.2024] [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: 09/03/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024]
Abstract
Store-operated calcium (Ca2+) entry (SOCE) represents a major route of Ca2+ permeation across the plasma membrane (PM) in nonexcitable cells, which plays an indispensable role in maintaining intracellular Ca2+ homeostasis. This process is orchestrated through the dynamic coupling between the endoplasmic reticulum (ER)-localized Ca2+ sensor stromal interaction molecule 1 (STIM1) and the PM-resident ORAI1 channel. Upon depletion of ER Ca2+ stores, STIM1 undergoes conformational rearrangements and oligomerization, leading to the translocation of activated STIM1 toward the PM. This movement is facilitated by the physical interactions between positively charged cytosolic domains within STIM1 and negatively charged phospholipids embedded in the PM, ultimately enabling its binding to and activation of the PM-embedded ORAI1 channel. In this mini-review, we provide an overview of STIM1-mediated Ca2+ signaling at ER-PM contact sites, highlighting the regulatory roles of phospholipids in the inner leaflet and sphingolipids in the outer leaflet of the PM. We also discuss the development of molecular tools that enable real-time visualization and manipulation of membrane contact sites (MCSs) at ER-PM junctions. Finally, we highlight recent progress in developing targeted therapies for human diseases linked to STIM1 mutations and dysregulated Ca2+ signaling at ER-PM MCSs.
Collapse
Affiliation(s)
- Yuepeng Ke
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, United States
| | - Ritchel Gannaban
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, United States
| | - Junchen Liu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, United States
- Graduate School of Biological Sciences, M. D. Anderson Cancer Center, University of Texas Health Science Center, Houston, Texas, United States
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, United States
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, Texas, United States
| |
Collapse
|
29
|
Palencia-Campos A, Ruiz-Cañas L, Abal-Sanisidro M, López-Gil JC, Batres-Ramos S, Saraiva SM, Yagüe B, Navarro D, Alcalá S, Rubiolo JA, Bidan N, Sánchez L, Mura S, Hermann PC, de la Fuente M, Sainz B. Reprogramming tumor-associated macrophages with lipid nanosystems reduces PDAC tumor burden and liver metastasis. J Nanobiotechnology 2024; 22:795. [PMID: 39719597 DOI: 10.1186/s12951-024-03010-5] [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: 05/15/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) requires innovative therapeutic strategies to counteract its progression and metastatic potential. Since the majority of patients are diagnosed with advanced metastatic disease, treatment strategies targeting not only the primary tumor but also metastatic lesions are needed. Tumor-Associated Macrophages (TAMs) have emerged as central players, significantly influencing PDAC progression and metastasis. Our objective was to validate an innovative therapeutic strategy involving the reprogramming of TAMs using lipid nanosystems to prevent the formation of a pro-metastatic microenvironment in the liver. RESULTS In vitro results demonstrate that M2-polarized macrophages lose their M2-phenotype following treatment with lipid nanoemulsions composed of vitamin E and sphingomyelin (VitE:SM), transitioning to an M0/M1 state. Specifically, VitE:SM nanoemulsion treatment decreased the expression of macrophage M2 markers such as Arg1 and Egr2, while M1 markers such as Cd86, Il-1b and Il-12b increased. Additionally, the TGF-βR1 inhibitor Galunisertib (LY2157299) was loaded into VitE:SM nanoemulsions and delivered to C57BL/6 mice orthotopically injected with KPC PDAC tumor cells. Treated mice showed diminished primary tumor growth and reduced TAM infiltration in the liver. Moreover, we observed a decrease in liver metastasis with the nanoemulsion treatment in an intrasplenic model of PDAC liver metastasis. Finally, we validated the translatability of our VitE:SM nanosystem therapy in a human cell-based 3D co-culture model in vivo, underscoring the pivotal role of macrophages in the nanosystem's therapeutic effect in the context of human PDAC metastasis. CONCLUSIONS The demonstrated effectiveness and safety of our nanosystem therapy highlights a promising therapeutic approach for PDAC, showcasing its potential in reprogramming TAMs and mitigating the occurrence of liver metastasis.
Collapse
Affiliation(s)
- Adrián Palencia-Campos
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Laura Ruiz-Cañas
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
- Biobanco Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Marcelina Abal-Sanisidro
- Nano-Oncology and Translational Therapeutics Group, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
- University of Santiago de Compostela (USC), 15782, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain
| | - Juan Carlos López-Gil
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
- Department of Biochemistry, Autónoma University of Madrid (UAM), 28029, Madrid, Spain
| | - Sandra Batres-Ramos
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Sofia Mendes Saraiva
- Nano-Oncology and Translational Therapeutics Group, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain
| | - Balbino Yagüe
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Diego Navarro
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
- Department of Biochemistry, Autónoma University of Madrid (UAM), 28029, Madrid, Spain
| | - Sonia Alcalá
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain
| | - Juan A Rubiolo
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, University of Santiago de Compostela (USC), Lugo, Spain
- Laboratorio Mixto de Biotecnología Acuática, Facultad de Ciencias Bioquímicas y Farmacéuticas, UNR, 2000, Rosario, Argentina
| | - Nadège Bidan
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Faculty of Veterinary, University of Santiago de Compostela (USC), Lugo, Spain
| | - Simona Mura
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Group, IDIS, Complexo Hospitalario Universitario de Santiago de Compostela, 15706, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain
- DIVERSA Technologies S.L, Edificio Emprendia, Campus Sur, 15782, Santiago de Compostela, Spain
| | - Bruno Sainz
- Cancer Stem Cells and Fibroinflammatory Microenvironment Group, Instituto de Investigaciones Biomédicas (IIBm) Sols-Morreale CSIC-UAM, 28029, Madrid, Spain.
- Biomarkers and Personalized Approach to Cancer Group (BIOPAC), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Area 3 Cancer, 28049, Madrid, Spain.
- Centro de Investigación Biomédica en Red, CIBERONC, ISCIII, Área Cáncer, Madrid, Spain.
| |
Collapse
|
30
|
Cui Y, Luo S, Wu B, Li Q, Han F, Wang Z. Immunomodulatory Effects of SPHK1 and Its Interaction with TFAP2A in Yellow Drum ( Nibea albiflora). Int J Mol Sci 2024; 25:13641. [PMID: 39769404 PMCID: PMC11728317 DOI: 10.3390/ijms252413641] [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: 11/05/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Sphingosine kinases (SPHKs) are essential enzymes that catalyze the phosphorylation of sphingosine to produce sphingosine-1-phosphate (S1P), which plays pivotal roles in inflammation and immune regulation. In this study, genome-wide association analysis (GWAS) identified the Ydsphk1 gene as closely associated with the resistance of yellow drum (Nibea albiflora) to Vibrio harveyi. Structural prediction showed that YDSPHK1 contains a typical diacylglycerol kinase catalytic (DAGKc) domain (154-291 aa). By constructing and transfecting Ydsphk1 expression plasmids into yellow drum kidney cells, we found that YDSPHK1 is localized in the cytoplasm. Subsequent RNA-Seq analysis of an overexpression plasmid identified 25 differentially expressed genes (DEGs), including 13 upregulated and 12 downregulated. Notably, nsun5 and hsp90aa1 were significantly upregulated, while Nfkbia and hmox1 were downregulated. Promoter analysis indicated that the core regulatory regions of Ydsphk1 are located between -1931~-1679 bp and -419~+92 bp, with two predicted TFAP2A binding sites in the -419~+92 bp region. Further studies demonstrated that varying concentrations of TFAP2A significantly reduced Ydsphk1 promoter activity. These findings underscore the pivotal role of Ydsphk1 in regulating immune responses in yellow drum, particularly through its impact on key immune-related genes and pathways such as NF-κB signaling and ferroptosis. The identification of Ydsphk1 as a mediator of immune regulation provides valuable insights into the molecular mechanisms of immune defense and highlights its potential as a target for enhancing pathogen resistance in aquaculture practices. This study lays a strong foundation for future research aimed at developing innovative strategies for disease management in aquaculture species.
Collapse
Affiliation(s)
- Yu Cui
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Shuai Luo
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Baolan Wu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Qiaoying Li
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
| | - Zhiyong Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Healthy Mariculture for the East China Sea, Fisheries College, Jimei University, Xiamen 361021, China; (Y.C.); (S.L.); (B.W.); (Q.L.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
31
|
Heymann CJF, Mak AL, Holleboom AG, Verheij J, Shiri-Sverdlov R, van Mil SWC, Tushuizen ME, Koek GH, Grefhorst A. The plasma lipidome varies with the severity of metabolic dysfunction-associated steatotic liver disease. Lipids Health Dis 2024; 23:402. [PMID: 39696394 DOI: 10.1186/s12944-024-02380-x] [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: 09/13/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is closely associated with many aspects of disturbed metabolic health. MASLD encompasses a wide spectrum of liver diseases, ranging from isolated steatosis to metabolic dysfunction-associated steatohepatitis (MASH), up to fibrosis, cirrhosis, and ultimately hepatocellular carcinoma. Limited noninvasive diagnostic tools are currently available to distinguish the various stages of MASLD and as such liver biopsy remains the gold standard for MASLD diagnostics. We aimed to explore whether the plasma lipidome and its variations can serve as a biomarker for MASLD stages. METHODS We investigated the plasma lipidome of 7 MASLD-free subjects and 32 individuals with MASLD, of whom 11 had MASH based on biopsy scoring. RESULTS Compared with the MASLD-free subjects, individuals with MASLD had higher plasma concentrations of sphingolipids, glycerolipids, and glycerophospholipids. Only plasma concentrations of ceramide-1-phosphate C1P(d45:1) and phosphatidylcholine PC(O-36:3), PC(O-38:3), and PC(36:2) differed significantly between presence of MASH in individuals with MASLD. Of these lipids, the first three have a very low relative plasma abundance, thus only PC(36:2) might serve as a biomarker with higher plasma concentrations in MASLD individuals without MASH compared to those with MASH. CONCLUSIONS Plasma lipids hold promise as biomarkers of MASLD stages, whereas plasma PC(36:2) concentrations would be able to distinguish individuals with MASH from those with MASLD without MASH.
Collapse
Affiliation(s)
- Clément J F Heymann
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Adriaan G Holleboom
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Ronit Shiri-Sverdlov
- Department of Genetics and Cell Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Saskia W C van Mil
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Maarten E Tushuizen
- Department of Gastroenterology and Hepatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ger H Koek
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Pondelick AM, Moncayo LV, Donvito G, McLane VD, Gillespie JC, Hauser KF, Spiegel S, Lichtman AH, Sim-Selley LJ, Selley DE. Dissociation between the anti-allodynic effects of fingolimod (FTY720) and desensitization of S1P 1 receptor-mediated G-protein activation in a mouse model of sciatic nerve injury. Neuropharmacology 2024; 261:110165. [PMID: 39303855 DOI: 10.1016/j.neuropharm.2024.110165] [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: 06/04/2024] [Revised: 08/28/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Sphingosine-1-phosphate (S1P) receptor (S1PR) agonists, such as fingolimod (FTY720), alleviate nociception in preclinical pain models by either activation (agonism) or inhibition (functional antagonism) of S1PR type-1 (S1PR1). However, the dose-dependence and temporal relationship between reversal of nociception and modulation of S1PR1 signaling has not been systematically investigated. This study examined the relationship between FTY720-induced antinociception and S1PR1 adaptation using a sciatic nerve chronic constriction injury (CCI) model of neuropathic pain in male and female C57Bl/6J mice. Daily injections of FTY720 for 14 days dose-dependently reversed CCI-induced mechanical allodynia without tolerance development, and concomitantly resulted in a dose-dependent reduction of G-protein activation by the S1PR1-selective agonist SEW2871 in the lumbar spinal cord and brain. These findings indicate FTY720-induced desensitization of S1PR1 signaling coincides with its anti-allodynic effects. Consistent with this finding, a single injection of FTY720 reversed mechanical allodynia while concomitantly producing partial desensitization of S1PR1-stimulated G-protein activation in the CNS. However, mechanical allodynia returned 24-hr post injection, despite S1PR1 desensitization at that time, demonstrating a dissociation between these measures. Furthermore, CCI surgery led to elevations of sphingolipid metabolites, including S1P, which were unaffected by daily FTY720 administration, suggesting FTY720 reversed mechanical allodynia by targeting S1PR1 rather than sphingolipid metabolism. Supporting this hypothesis, acute administration of the S1PR1-selective agonist CYM-5442 mimicked the anti-allodynic effect of FTY720. In contrast, the S1PR1-selective antagonist NIBR-0213 prevented the anti-allodynic effect of FTY720, but NIBR-0213 given alone did not affect nociception. These results indicate that FTY720 alleviates CCI-induced allodynia through a mechanism distinct from functional antagonism.
Collapse
Affiliation(s)
- Abby M Pondelick
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Lauren V Moncayo
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Giulia Donvito
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Virginia D McLane
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - James C Gillespie
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Laura J Sim-Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
33
|
Xie P, Chen J, Dan A, Lin Z, He Y, Cai Z. Long-term exposure to triclocarban induces splenic injuries in mice: Insights from spatial metabolomics and lipidomics. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136370. [PMID: 39486321 DOI: 10.1016/j.jhazmat.2024.136370] [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: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Triclocarban (TCC) is a widely used antimicrobial agent and known endocrine-disrupting chemical found in various products. While its potential toxicities on endocrine-related organs have been highlighted in previous studies, the effects of TCC on non-endocrine organs, particularly the spleen, remain largely unknown. Here, we employed a novel approach combining long-term TCC exposure in a mouse model with spatial metabolomics and lipidomics to investigate the effects of TCC on the spleen. Our results showed that TCC exposure significantly altered the splenic organ weight and coefficient and induced obvious pathological alterations. Omic analysis revealed that TCC exposure disrupted the splenic homeostasis, as indicated by the upregulation of glutathione metabolism, ceramide-to-sphingomyelin signaling and biosynthesis of glycerophospholipids. Notably, the data of mass spectrometry imaging (MSI) revealed that TCC accumulated in the red pulp of the mouse spleen, while its metabolites concentrated in the white pulp. Further MSI analyses identified region-specific metabolic disruptions, including upregulated ceramide signaling in the red pulp, indicating localized inflammation, and upregulated glutathione metabolism throughout the spleen, suggesting widespread oxidative damage. Our findings provide crucial insights into the spatial distribution and biochemical impact of TCC on mice spleens, highlighting the potential risks of long-term TCC exposure to immune function.
Collapse
Affiliation(s)
- Peisi Xie
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Jing Chen
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Akang Dan
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Zian Lin
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Yu He
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Zongwei Cai
- Ministry of Education Key Laboratory of Analytical Science for Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China; State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
34
|
Wang L, Bi L, Qiu Y, Huang G, Ye P, Liu Y, Li A, Yang X, Shen P, Wang J, Zeng Q, Zhang H, Li S, Jin H. Effectiveness of electro-acupuncture for cognitive improvement on Alzheimer's disease quantified via PET imaging of sphingosine-1-phosphate receptor 1. Alzheimers Dement 2024; 20:8331-8345. [PMID: 39320044 DOI: 10.1002/alz.14260] [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: 01/25/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Electro-acupuncture (EA) has demonstrated potential in improving mild-to-moderate dementia in clinics, but the underlying scientific target remains unclear. METHODS EA was administered to APP/PS1 Alzheimer's disease (AD) mice, with untreated AD, and wild type (WT) mice serving as controls. The efficacy of EA was assessed by the Morris water maze cognitive functional tests. Brain magnetic resonance imaging-positron emission tomography (PET) scans using [18F]TZ4877 targeting sphingosine-1-phosphate receptor 1 (S1PR1) and [18F]AV45 targeting amyloid beta fibrils were conducted. The correlation between regional brain PET quantifications and cognitive functions was analyzed. RESULTS EA significantly improved cognitive and memory functions of AD (p = 0.04) and reduced the uptake of [18F]TZ4877 in the cortex (p = 0.02) and hippocampus (p = 0.03). Immunofluorescence confirmed colocalizations of S1PR1 with glial fibrillary acidic protein and ionized calcium-binding adaptor molecule-1. Furthermore, immunohistochemistry showed a significant reduction of interleukin 1β and tumor necrosis factor α after EA treatment. DISCUSSION EA may reverse AD by suppressing neuroinflammation, and the PET imaging of S1PR1 seemed potent in evaluating the treatment for AD patients HIGHLIGHTS: Electro-acupuncture (EA) was administered to APP/PS1 Alzheimer's disease (AD) mice, with untreated AD, and wild type (WT) mice serving as controls. The efficacy of EA was assessed by the Morris water maze cognitive functional tests and positron emission tomography (PET) imaging quantifications. PET tracer [18F]AV45 was used to detect amyloid beta deposition. An increased uptake of [18F]AV45 was found in AD compared to WT mice, with significance observed only in the cortex and not in the hippocampus. EA treatment exhibited a trend toward reduced [18F]AV45 uptake in AD mouse brains post-treatment. However, statistical difference was not attained in most brain regions. EA "Baihui (DU20) and Sishencong (EX-HN1)" significantly improved cognitive and memory functions of AD (p = 0.04). Brain magnetic resonance imaging p(MRI)-positron emission tomography (PET) quantifications revealed that significantly reduced the uptake of [18F]TZ4877 in the cortex (p = 0.02) and hippocampus (p = 0.03) after EA treatment. The correlation between PET quantifications and cognitive functions was analyzed and the most notable correlations were found between escape latency (reaction cognitive and memory behavior) and volume distribution (VT) quantifications of [18F]TZ4877. VT quantifications of [18F]TZ4877 in key brain regions for cognitive and memory ability, such as the cortex and hippocampus, positively correlated with platform latency (cortex p < 0.01, r = 0.7102; hippocampus p < 0.01, r = 0.6891). Immunofluorescence confirmed colocalizations of S1PR1 with glial fibrillary acidic protein and ionized calcium-binding adaptor molecule-1 in the AD brain. And the EA treatment significantly reduced the signals in the cortex and hippocampus. Immunohistochemistry showed a significant reduction of interleukin 1β and tumor necrosis factor α after EA treatment. EA reversed AD by suppressing neuroinflammation in the cortex and hippocampus. The S1PR1 targeting PET tracer [18F]TZ4877 showed promise in evaluating the pathological progression of AD in clinical settings.
Collapse
Affiliation(s)
- Lu Wang
- Department of Chinese Medicine Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Lei Bi
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yifan Qiu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Guolong Huang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Peizhen Ye
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yongshan Liu
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - An Li
- Department of Chinese Medicine Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xuan Yang
- Department of Chinese Medicine Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Peining Shen
- Pharmaceutical Clinical Trails Office, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Junfeng Wang
- Department of Neurology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Qi Zeng
- Department of Chinese Medicine Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hongyu Zhang
- Department of Chinese Medicine Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Shengqiao Li
- Department of Chinese Medicine Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hongjun Jin
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
35
|
Liu B, Zhou J, Jiang B, Tang B, Liu T, Lei P. The role of ACER2 in intestinal sphingolipid metabolism and gastrointestinal cancers. Front Immunol 2024; 15:1511283. [PMID: 39650647 PMCID: PMC11621088 DOI: 10.3389/fimmu.2024.1511283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Sphingolipids, particularly sphingosine-1-phosphate (S1P), are bioactive lipids involved in regulating cellular processes such as proliferation, apoptosis, inflammation, and tumor progression. Alkaline ceramidase 2 (ACER2) plays a critical role in sphingolipid metabolism by catalyzing the hydrolysis of ceramide to sphingosine, which is subsequently converted to S1P. Dysregulation of ACER2 has been implicated in various gastrointestinal cancers, including colorectal cancer, gastric cancer, and hepatocellular carcinoma. ACER2-mediated sphingolipid signaling, particularly through the SphK/S1P pathway, influences cancer development by modulating immune responses, inflammation, and the balance between cell survival and death. This review examines the physiological functions of ACER2, and its role in sphingolipid metabolism, and its contribution to the pathogenesis of gastrointestinal cancers. Understanding the mechanisms by which ACER2 regulates tumor progression and immune modulation may open new avenues for targeted therapies in gastrointestinal malignancies.
Collapse
Affiliation(s)
- Binggang Liu
- Department of Gastrointestinal Surgery, the Central Hospital of Yongzhou, Yongzhou, China
| | | | | | | | | | | |
Collapse
|
36
|
Cozzolino K, Sanford L, Hunter S, Molison K, Erickson B, Courvan MCS, Jones T, Ajit D, Galbraith MD, Espinosa JM, Bentley DL, Allen MA, Dowell RD, Taatjes DJ. Mediator kinase inhibition suppresses hyperactive interferon signaling in Down syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.05.547813. [PMID: 37461585 PMCID: PMC10349994 DOI: 10.1101/2023.07.05.547813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Hyperactive interferon (IFN) signaling is a hallmark of Down syndrome (DS), a condition caused by trisomy 21 (T21); strategies that normalize IFN signaling could benefit this population. Mediator-associated kinases CDK8 and CDK19 drive inflammatory responses through incompletely understood mechanisms. Using sibling-matched cell lines with/without T21, we investigated Mediator kinase function in the context of hyperactive IFN in DS over a 75min - 24h timeframe. Activation of IFN-response genes was suppressed in cells treated with the CDK8/CDK19 inhibitor cortistatin A (CA), via rapid suppression of IFN-responsive transcription factor (TF) activity. We also discovered that CDK8/CDK19 affect splicing, a novel means by which Mediator kinases control gene expression. To further probe Mediator kinase function, we completed cytokine screens and metabolomics experiments. Cytokines are master regulators of inflammatory responses; by screening 105 different cytokine proteins, we show that Mediator kinases help drive IFN-dependent cytokine responses at least in part through transcriptional regulation of cytokine genes and receptors. Metabolomics revealed that Mediator kinase inhibition altered core metabolic pathways in cell type-specific ways, and broad up-regulation of anti-inflammatory lipid mediators occurred specifically in kinase-inhibited cells during hyperactive IFNγ signaling. A subset of these lipids (e.g. oleamide, desmosterol) serve as ligands for nuclear receptors PPAR and LXR, and activation of these receptors occurred specifically during hyperactive IFN signaling in CA-treated cells, revealing mechanistic links between Mediator kinases, lipid metabolism, and nuclear receptor function. Collectively, our results establish CDK8/CDK19 as context-specific metabolic regulators, and reveal that these kinases control gene expression not only via TFs, but also through metabolic changes and splicing. Moreover, we establish that Mediator kinase inhibition antagonizes IFN signaling through transcriptional, metabolic, and cytokine responses, with implications for DS and other chronic inflammatory conditions.
Collapse
Affiliation(s)
- Kira Cozzolino
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| | - Lynn Sanford
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Samuel Hunter
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Kayla Molison
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| | - Benjamin Erickson
- Dept. Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- UC-Denver RNA Bioscience Initiative
| | - Meaghan C S Courvan
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
- Crnic Institute Boulder Branch
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Taylor Jones
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| | - Deepa Ajit
- Metabolon, Inc., Durham, North Carolina, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Dept. of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Dept. of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David L Bentley
- Dept. Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO 80045, USA
- UC-Denver RNA Bioscience Initiative
| | - Mary A Allen
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Robin D Dowell
- Dept. of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO, 80303, USA
- BioFrontiers Institute, University of Colorado, Boulder, CO, 80303, USA
| | - Dylan J Taatjes
- Dept. of Biochemistry, University of Colorado, Boulder, CO, 80303, USA
| |
Collapse
|
37
|
Augusto SN, Suresh A, Tang WHW. Ceramides as Biomarkers of Cardiovascular Diseases and Heart Failure. Curr Heart Fail Rep 2024; 22:2. [PMID: 39560878 DOI: 10.1007/s11897-024-00689-3] [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] [Accepted: 09/17/2024] [Indexed: 11/20/2024]
Abstract
PURPOSE OF REVIEW Ceramides are lipid species that play several physiological roles in the body, including stress response, inflammation, and apoptosis, and their involvement in lipid metabolism and energy production makes them crucial in the pathophysiology of heart failure (HF). RECENT FINDINGS Several species of ceramides and ceramide signatures have recently been investigated as possible biomarkers of cardiovascular disease (CVD), and risk scores have demonstrated prognostic value in stratifying patients by risk and possibly predicting adverse cardiac events. With growing interest in targeting metabolic dysfunction, understanding the role of ceramides in CVD also opens the possibility of novel therapeutics that target ceramide metabolism to improve cardiac function and cardiac outcomes in patients. Understanding the role of ceramides in CVD opens the possibility of novel diagnostics and theragnostic targeting ceramide metabolism to improve cardiac function and cardiac outcomes in patients with heart failure.
Collapse
Affiliation(s)
- Silvio N Augusto
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA, 9500 Euclid Avenue, Desk J3-4, 44195
| | - Abhilash Suresh
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - W H Wilson Tang
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA, 9500 Euclid Avenue, Desk J3-4, 44195.
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
38
|
Wu Q, Liu K, Hou R, Wu X, Ruan X, Wang M, Sun Z, Meng L, Dai G, Li C, Wu J, Mu G. Integrative Lipid Pseudotargeted Metabolomics and Amino Acids Targeted Metabolomics Unravel the Therapeutic Mechanism of Rhizoma Paridis Saponins on Experimental Colitis of Damp-Heat Type. Drug Des Devel Ther 2024; 18:5087-5108. [PMID: 39554759 PMCID: PMC11568858 DOI: 10.2147/dddt.s476494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Inflammatory bowel disease (IBD) is a serious disease that affects the metabolism and inflammatory responses of human beings. From the perspective of traditional Chinese medicine, damp-heat syndrome is one of the main syndromes of IBD. Rhizoma Paridis, also known as the root of Paris polyphylla, a well-known herbal medicine used in China, is used to treat IBD with damp-heat syndrome (IBD-DH). However, uncertainty still exists regarding the underlying mechanisms and the impact of Rhizoma Paridis on IBD-DH. Methods The rats in the model (DAT) and medication administration (Rhizoma Paridis total saponins (RPTS) and Pennogenin (PN)) groups were given a high temperature and high humidity environment, high fat and high sugar diet combined with 2,4,6-trinitrobenzene sulfonic acid (TNBS) to establish the model of experimental colitis of damp-heat type, and the normal control group (RNC) rats were given a normal diet at normal temperature and humidity. Damp-heat control group (DNC) was set with the same condition as DAT without TNBS. Hematoxylin-Eosin (HE) staining was used to observe the histopathological morphology of the rat colorectum. The expression of the metabolism-related genes (Phospholipase A2 (sPLA2, cPLA2), and phosphatidylethanolamine N-methyltransferase (PEMT)) was assessed by using real-time quantitative PCR analysis (RT-qPCR). And the levels of the metabolism-related proteins (sPLA2, cPLA2), S100A8/9, Arg-1, and cytokines were detected by enzyme-linked immunosorbent assay (ELISA) kit. To investigate lipids and amino acids which closely associated with the IBD and IBD-DH, lipid pseudotargeted metabolomics with UHPLC-TQ/MS analysis method, as well as targeted quantitative amino acid analysis were performed. Results Our data showed that RPTS (50 mg/kg) and PN (20 mg/kg) significantly ameliorated the severity of TNBS-induced colitis and downregulated the levels of circulating proinflammatory cytokines. Compared with RNC group, lipid pseudotargeted metabolomics demonstrated that glycerophospholipids, sphingolipids, carnitine, and glycerolipids were the four most perturbed lipid classes, and amino acids targeted metabolomics demonstrated that serine, N-acetylneuraminic acid, histidine, proline, taurine, and kynurenine changed significantly in DAT group . Correlation analyses showed tight associations between most of differential metabolites and proinflammatory cytokines. RPTS and PN both regulated glycerophospholipid metabolism and sphingolipid metabolism. However, both of them did not have a significant effect on amino acid modulation. RPTS and PN potentially regulated sPLA2, cPLA2, and PEMT. Conclusion These results showed that RPTS (50 mg/kg) and PN (20 mg/kg) effectively alleviated rats' colitis of damp-heat type, affected cytokines, and altered lipid metabolism without significant modulation on amino acid metabolism.
Collapse
Affiliation(s)
- Qi Wu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Kexin Liu
- Department of Gastroenterology, The 981th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Chengde, Hebei, 067000, People’s Republic of China
| | - Ruijuan Hou
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Xingxing Wu
- Department of Gastroenterology, Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, 450053, People’s Republic of China
| | - Xiaoyu Ruan
- Internal Medicine Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Mao Wang
- Ethics Committee, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Zhiting Sun
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Lingchang Meng
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Guoliang Dai
- Clinical Pharmacology Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Changyin Li
- Clinical Pharmacology Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, People’s Republic of China
| | - Jing Wu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| | - Genglin Mu
- Institute of Chinese Medicine of Nanjing University, Nanjing Drum Tower Hospital, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, 210008, People’s Republic of China
| |
Collapse
|
39
|
Feng L, Zhao X, Song J, Yang S, Xiang J, Zhang M, Tu C, Song X. Association between the plasma ceramide and coronary microvascular resistance. Cardiovasc Diabetol 2024; 23:395. [PMID: 39497178 PMCID: PMC11536972 DOI: 10.1186/s12933-024-02495-6] [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: 09/10/2024] [Accepted: 10/30/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Plasma ceramide plays a potentially significant role in the pathogenesis of coronary microvascular dysfunction. However, the relationship between plasma ceramide and coronary microvascular resistance in patients remains unclear. This study aimed to evaluate the association between plasma ceramide levels, as well as their distinct ratios, and coronary microvascular resistance. METHODS This single-center observational study retrospectively enrolled patients who underwent both ceramide measurement and coronary angiography during hospitalization. The microvascular resistance of the coronary arteries was assessed in all patients using the angiography-derived index of microcirculatory resistance (Angio-IMR). The cumulative coronary microvascular resistance was calculated by summing the microvascular resistance of the three main coronary arteries. Multiple linear and logistic regression analyses were employed to evaluate the relationship between plasma ceramide and cumulative coronary microvascular resistance. Restricted cubic spline (RCS) analysis was conducted to investigate the association between plasma ceramide levels and cumulative coronary microvascular resistance. Receiver operating characteristic (ROC) curves were employed to evaluate the predictive value of plasma ceramide for coronary microvascular resistance. Additionally, subgroup analyses and interaction tests were performed. RESULTS A total of 225 patients were included in this study, with a median cumulative coronary microvascular resistance of 48.04 (40.32-56.73). After adjusting for potential confounding factors, both plasma 16:0 ceramide and the 16:0/24:0 ceramide ratio were positively associated with cumulative coronary microvascular resistance [standardized β ± standard error: 75.05 ± 8.46 (P < 0.001) and 91.72 ± 20.41 (P < 0.001), respectively]. Similar independent associations were observed in predicting high cumulative microvascular resistance [β = 8.03 ± 1.91 (P < 0.001) and 9.98 ± 3.88 (P = 0.010), respectively]. Additionally, a significant nonlinear relationship was observed between plasma 16:0 ceramide, the 16:0/24:0 ceramide ratio, and cumulative coronary microvascular resistance (P for nonlinear < 0.05). The ROC analysis revealed that the optimal cut-off for plasma 16:0 ceramide is 0.178 µmol/L, with a specificity of 57.1% and a sensitivity of 91.2%. For the 16:0/24:0 ceramide ratio, the optimal cut-off is 0.072, yielding a specificity of 73.2% and a sensitivity of 54.9%. Subgroup analysis indicated that the association between plasma ceramide and coronary microvascular resistance was trending toward non-significance in patients with acute coronary syndrome (ACS). CONCLUSIONS A significant nonlinear relationship exists between plasma ceramide and coronary microvascular resistance, which holds important clinical implications for the risk stratification of coronary microvascular disease. New insights into the potential effects of ceramides enhance our understanding of the complex mechanisms underlying coronary microvascular disease and warrant further investigation in a broader population.
Collapse
Affiliation(s)
- Lanxin Feng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jianqiao Song
- Sun yat sen university, Zhongshan school of medicine, Guangzhou, People's Republic of China
| | - Shuwen Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jianping Xiang
- ArteryFlow Technology Co., Ltd, Hangzhou, People's Republic of China
| | - Min Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Chenchen Tu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China.
| |
Collapse
|
40
|
Esplin ED, Hanson C, Wu S, Horning AM, Barapour N, Nevins SA, Jiang L, Contrepois K, Lee H, Guha TK, Hu Z, Laquindanum R, Mills MA, Chaib H, Chiu R, Jian R, Chan J, Ellenberger M, Becker WR, Bahmani B, Khan A, Michael B, Weimer AK, Esplin DG, Shen J, Lancaster S, Monte E, Karathanos TV, Ladabaum U, Longacre TA, Kundaje A, Curtis C, Greenleaf WJ, Ford JM, Snyder MP. Multiomic analysis of familial adenomatous polyposis reveals molecular pathways associated with early tumorigenesis. NATURE CANCER 2024; 5:1737-1753. [PMID: 39478120 PMCID: PMC11584401 DOI: 10.1038/s43018-024-00831-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 08/29/2024] [Indexed: 11/24/2024]
Abstract
Familial adenomatous polyposis (FAP) is a genetic disease causing hundreds of premalignant polyps in affected persons and is an ideal model to study transitions of early precancer states to colorectal cancer (CRC). We performed deep multiomic profiling of 93 samples, including normal mucosa, benign polyps and dysplastic polyps, from six persons with FAP. Transcriptomic, proteomic, metabolomic and lipidomic analyses revealed a dynamic choreography of thousands of molecular and cellular events that occur during precancerous transitions toward cancer formation. These involve processes such as cell proliferation, immune response, metabolic alterations (including amino acids and lipids), hormones and extracellular matrix proteins. Interestingly, activation of the arachidonic acid pathway was found to occur early in hyperplasia; this pathway is targeted by aspirin and other nonsteroidal anti-inflammatory drugs, a preventative treatment under investigation in persons with FAP. Overall, our results reveal key genomic, cellular and molecular events during the earliest steps in CRC formation and potential mechanisms of pharmaceutical prophylaxis.
Collapse
Affiliation(s)
- Edward D Esplin
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Casey Hanson
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Si Wu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Aaron M Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Nasim Barapour
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Lihua Jiang
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Hayan Lee
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Tuhin K Guha
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Zheng Hu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | | | - Meredith A Mills
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Hassan Chaib
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Roxanne Chiu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Joanne Chan
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Winston R Becker
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Bahareh Bahmani
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Aziz Khan
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Basil Michael
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Annika K Weimer
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jeanne Shen
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Samuel Lancaster
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Emma Monte
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Uri Ladabaum
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Teri A Longacre
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Christina Curtis
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - James M Ford
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA.
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
41
|
Taru V, Szabo G, Mehal W, Reiberger T. Inflammasomes in chronic liver disease: Hepatic injury, fibrosis progression and systemic inflammation. J Hepatol 2024; 81:895-910. [PMID: 38908436 PMCID: PMC11881887 DOI: 10.1016/j.jhep.2024.06.016] [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: 02/18/2024] [Revised: 05/23/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Chronic liver disease leads to hepatocellular injury that triggers a pro-inflammatory state in several parenchymal and non-parenchymal hepatic cell types, ultimately resulting in liver fibrosis, cirrhosis, portal hypertension and liver failure. Thus, an improved understanding of inflammasomes - as key molecular drivers of liver injury - may result in the development of novel diagnostic or prognostic biomarkers and effective therapeutics. In liver disease, innate immune cells respond to hepatic insults by activating cell-intrinsic inflammasomes via toll-like receptors and NF-κB, and by releasing pro-inflammatory cytokines (such as IL-1β, IL-18, TNF-α and IL-6). Subsequently, cells of the adaptive immune system are recruited to fuel hepatic inflammation and hepatic parenchymal cells may undergo gasdermin D-mediated programmed cell death, termed pyroptosis. With liver disease progression, there is a shift towards a type 2 inflammatory response, which promotes tissue repair but also fibrogenesis. Inflammasome activation may also occur at extrahepatic sites, such as the white adipose tissue in MASH (metabolic dysfunction-associated steatohepatitis). In end-stage liver disease, flares of inflammation (e.g., in severe alcohol-related hepatitis) that spark on a dysfunctional immune system, contribute to inflammasome-mediated liver injury and potentially result in organ dysfunction/failure, as seen in ACLF (acute-on-chronic liver failure). This review provides an overview of current concepts regarding inflammasome activation in liver disease progression, with a focus on related biomarkers and therapeutic approaches that are being developed for patients with liver disease.
Collapse
Affiliation(s)
- Vlad Taru
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Christian-Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria; Iuliu Hatieganu University of Medicine and Pharmacy, 4(th) Dept. of Internal Medicine, Cluj-Napoca, Romania
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Wajahat Mehal
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA; West Haven Veterans Medical Center, West Haven, CT, USA.
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Christian-Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria; Center for Molecular Medicine (CeMM) of the Austrian Academy of Science, Vienna, Austria
| |
Collapse
|
42
|
Wang T, Tian T, Zhu Z, Fang S, Zhang L, Peng X, Shi R, Li Y, Wu J, Ma Y. Gardenia jasminoides Ellis. Polysaccharides Alleviated Cholestatic Liver Injury by Increasing the Production of Butyric Acid and FXR Activation. Phytother Res 2024; 38:5363-5375. [PMID: 39237123 DOI: 10.1002/ptr.8326] [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: 11/22/2023] [Revised: 07/21/2024] [Accepted: 08/17/2024] [Indexed: 09/07/2024]
Abstract
Gardenia jasminoides Ellis. polysaccharide (GPS) can protect against cholestatic liver injury (CLI) by regulating nuclear farnesoid X receptor (FXR).However, the mechanism via which GPS mediates the FXR pathway remains unclear. The aim of this study was to investigate the mechanism. Firstly, an alpha-naphthylisothiocyanate-induced cholestatic mouse model was administered with GPS to evaluate its hepatoprotective effects. The metabolic pathways influenced by GPS in cholestatic mice were detected by serum metabolomics. The effect of GPS on bile acid (BA) homeostasis, FXR expression, and liver inflammation were investigated. Second, the intestinal bacteria metabolites affected by GPS in vivo and in vitro were determined. The activation of FXR by sodium butyrate (NaB) was measured. Finally, the effects of NaB on cholestatic mice were demonstrated. The main pathways influenced by GPS involved BA biosynthesis. GPS upregulated hepatic FXR expression, improved BA homeostasis, reduced F4/80+ and Ly6G+ positive areas in the liver, and inhibited liver inflammation in cholestatic mice. Butyric acid was the most notable intestinal bacterial metabolite following GPS intervention. NaB activated the transcriptional activity of FXR in vitro, upregulated hepatic FXR and its downstream efflux transporter expression, and ameliorated disordered BA homeostasis in CLI mice. NaB inhibited the toll-like receptor 4/nuclear factor (TLR4/NF-κB) pathway and reduced inflammation and CLI in mice. An FXR antagonist suppressed the effects. In conclusion, GPS increased butyric acid production, which can activate hepatic FXR, reverse BA homeostasis disorder, and inhibit the TLR4/NF-κB inflammatory pathway, exerting protective effects against CLI.
Collapse
Affiliation(s)
- Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian Tian
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenyun Zhu
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Su Fang
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lincong Zhang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaotian Peng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Shi
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
43
|
Older EA, Zhang J, Ferris ZE, Xue D, Zhong Z, Mitchell MK, Madden M, Wang Y, Chen H, Nagarkatti P, Nagarkatti M, Fan D, Ellermann M, Li YX, Li J. Biosynthetic enzyme analysis identifies a protective role for TLR4-acting gut microbial sulfonolipids in inflammatory bowel disease. Nat Commun 2024; 15:9371. [PMID: 39477928 PMCID: PMC11525784 DOI: 10.1038/s41467-024-53670-y] [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: 05/10/2023] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
The trillions of microorganisms inhabiting the human gut are intricately linked to human health. While specific microbes have been associated with diseases, microbial abundance alone cannot reveal the molecular mechanisms involved. One such important mechanism is the biosynthesis of functional metabolites. Here, we develop a biosynthetic enzyme-guided disease correlation approach to uncover microbial functional metabolites linked to disease. Applying this approach, we negatively correlate the expression of gut microbial sulfonolipid (SoL) biosynthetic enzymes to inflammatory bowel disease (IBD). Targeted chemoinformatics and metabolomics then confirm that SoL abundance is significantly decreased in IBD patient data and samples. In a mouse model of IBD, we further validate that SoL abundance is decreased while inflammation is increased in diseased mice. We show that SoLs consistently contribute to the immunoregulatory activity of different SoL-producing human microbes. We further reveal that sulfobacins A and B, representative SoLs, act on Toll-like receptor 4 (TLR4) and block lipopolysaccharide (LPS) binding, suppressing both LPS-induced inflammation and macrophage M1 polarization. Together, these results suggest that SoLs mediate a protective effect against IBD through TLR4 signaling and showcase a widely applicable biosynthetic enzyme-guided disease correlation approach to directly link the biosynthesis of gut microbial functional metabolites to human health.
Collapse
Affiliation(s)
- Ethan A Older
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, 29208, USA
| | - Jian Zhang
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Zachary E Ferris
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, 29208, USA
| | - Dan Xue
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, 29208, USA
| | - Zheng Zhong
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Mary K Mitchell
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina, 29208, USA
| | - Michael Madden
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, 29208, USA
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, 29209, USA
| | - Hexin Chen
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina, 29208, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, 29209, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, 29209, USA
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina, 29209, USA
| | - Melissa Ellermann
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina, 29208, USA
| | - Yong-Xin Li
- Department of Chemistry and The Swire Institute of Marine Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China.
| | - Jie Li
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina, 29208, USA.
| |
Collapse
|
44
|
Wang D, Xu R, Wang Z. Protective Role of Sphingosine-1-Phosphate During Radiation-Induced Testicular Injury. Antioxidants (Basel) 2024; 13:1322. [PMID: 39594464 PMCID: PMC11591009 DOI: 10.3390/antiox13111322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
The impact of ionizing radiation on the male reproductive system is gaining increasing attention, particularly when it comes to testicular damage, which may result in decreased sperm quality and hormonal imbalances. Finding effective protective measures to mitigate testicular damage caused by radiation has become a focal point in the biomedical field. S1P, an essential biological signaling molecule, has garnered significant interest due to its multiple roles in regulating cellular functions and its protective effects against radiation-induced testicular injury. S1P not only effectively reduces the generation of ROS induced by radiation but also alleviates oxidative stress by enhancing the activity of antioxidant enzymes. Furthermore, S1P inhibits radiation-induced cell apoptosis by regulating the expression of anti-apoptotic and pro-apoptotic proteins. Additionally, S1P alleviates radiation-induced inflammation by inhibiting the production of inflammatory factors, thereby further protecting testicular tissue. In summary, S1P effectively reduces radiation-induced testicular damage through multiple mechanisms, offering a promising therapeutic approach to safeguard male reproductive health. Future research should explore the specific mechanisms of action and clinical application potential of S1P, aiming to contribute significantly to the prevention and treatment of radiation damage.
Collapse
Affiliation(s)
- Defan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen 361102, China;
| | - Renfeng Xu
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China;
| | - Zhengchao Wang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China;
| |
Collapse
|
45
|
Wang D, Zhu L, Liu H, Feng X, Zhang C, Liu B, Li T, Liu L, Chang H, Sun J, Yang L, Yang W. Altered gut metabolites and metabolic reprogramming involved in the pathogenesis of colitis-associated colorectal cancer and the transition of colon "inflammation to cancer". J Pharm Biomed Anal 2024; 253:116553. [PMID: 39486392 DOI: 10.1016/j.jpba.2024.116553] [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: 07/03/2024] [Revised: 10/27/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Colitis-associated colorectal cancer (CAC) is fatal and can develop spontaneously or as a complication of inflammatory bowel diseases. Although co-administration of azoxymethane/dextran sulfate sodium (AOM/DSS) is a classic method for CAC modeling, its limitations need to be addressed. Accordingly, we aimed to optimize the AOM/DSS model to study CAC extensively and further investigate its pathogenic mechanisms relative to microbiota and metabolism. We optimized the CAC model via a single or enhanced injection of AOM combined with different administration modes and varying DSS concentrations. Subsequently, the fecal-microbiota composition was examined using 16S RNA sequencing, and fecal-colon-metabolome profiles were evaluated via ultra-high performance liquid chromatography-mass spectrometry. Two interval injections of AOM combined with 1.5 % DSS-free drinking resulted in a high tumor formation rate, uniform tumor formation, and low mortality. Based on this model, we innovatively divided the pathogenesis of CAC into three stages, namely inflammation induction, proliferation initiation, and tumorigenesis, and examined the pathological characteristics in each stage. Gut microbial dysbiosis and metabolic alteration drove colorectal tumorigenesis by aggravating inflammation while promoting cell proliferation and carcinogenesis in mice. For the first time, we dynamically demonstrated the process of colon "inflammation to cancer" transformation and provided novel insights to clarify the role of amino acid metabolism in the formation of CAC.
Collapse
Affiliation(s)
- Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Bin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hao Chang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jingwei Sun
- Beijing University of Chinese Medicine, Beijing 100700, China
| | - Lei Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
46
|
Deng J, Wu S, Huang Y, Deng Y, Yu K. Esophageal cancer risk is influenced by genetically determined blood metabolites. Medicine (Baltimore) 2024; 103:e40122. [PMID: 39470544 PMCID: PMC11521038 DOI: 10.1097/md.0000000000040122] [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: 05/24/2024] [Revised: 07/13/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
It remains unclear what causes esophageal cancer (EC), but blood metabolites have been connected to it. Our study performed a Mendelian randomization (MR) analysis to assess the causality from genetically proxied 1400 blood metabolites to EC level. A two-sample MR analysis was employed to evaluate the causal relationship between 1400 blood metabolites and EC. Initially, the EC genome-wide association study (GWAS) data (from Jiang L et al) were examined, leading to the identification of certain metabolites. Subsequently, another set of EC GWAS data from FINNGEN was utilized to validate the findings. Causality was primarily determined through inverse variance weighting, with additional support from the MR-Egger, weighted median, and MR-PRESSO models. Heterogeneity was assessed using the MR Cochran Q test. The MR-Egger intercept and MR-PRESSO global methods were employed to detect multicollinearity. In this study, Bonferroni corrected P value was used for significance threshold. We found 2 metabolites with overlaps, which are lipids. Docosatrienoate (22:3n3) was found to be causally associated with a decreased risk of EC, as evidenced by the EC GWAS data (from Jiang et al) (odds ratio [OR] = 0.620, 95% confidence interval [CI] = 0.390-0.986, P = .044) and the EC GWAS data (from FINNGEN) (OR = 0.77, 95% CI = 0.6-0.99, P = .042), these results were consistent across both data sets. Another overlapping metabolite, glycosyl-N-(2-hydroxyneuramoyl)-sphingosine, was associated with the risk of ES, with EC GWAS data (from Jiang L et al) (OR = 1.536, 95% CI = 1.000-2.360, P = .049), while EC GWAS data (from FINNGEN) (OR = 0.733, 95% CI = 0.574-0.937, P = .013), the 2 data had opposite conclusions. The findings of this study indicate a potential association between lipid metabolites (Docosatrienoate (22:3n3) and glycosyl-N-(2-hydroxynervonoyl)-sphingosine (d18:1/24:1 (2OH))) and the risk of esophageal carcinogenesis.
Collapse
Affiliation(s)
- Jieyin Deng
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| | - Silin Wu
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
- School of Clinical Medicine, North Sichuan Medical College, Sichuan, China
| | - Ye Huang
- Department of Nursing, Nursing School, Chengdu Medical College, Chengdu, China
| | - Yi Deng
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| | - Ke Yu
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| |
Collapse
|
47
|
Liu PY, Liaw J, Soutter F, Ortiz JJ, Tomley FM, Werling D, Gundogdu O, Blake DP, Xia D. Multi-omics analysis reveals regime shifts in the gastrointestinal ecosystem in chickens following anticoccidial vaccination and Eimeria tenella challenge. mSystems 2024; 9:e0094724. [PMID: 39287379 PMCID: PMC11494932 DOI: 10.1128/msystems.00947-24] [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: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Coccidiosis, caused by Eimeria parasites, significantly impacts poultry farm economics and animal welfare. Beyond its direct impact on health, Eimeria infection disrupts enteric microbial populations leading to dysbiosis and increases vulnerability to secondary diseases such as necrotic enteritis, caused by Clostridium perfringens. The impact of Eimeria infection or anticoccidial vaccination on host gastrointestinal phenotypes and enteric microbiota remains understudied. In this study, the metabolomic profiles and microbiota composition of chicken caecal tissue and contents were evaluated concurrently during a controlled experimental vaccination and challenge trial. Cobb500 broilers were vaccinated with a Saccharomyces cerevisiae-vectored anticoccidial vaccine and challenged with 15,000 Eimeria tenella oocysts. Assessment of caecal pathology and quantification of parasite load revealed correlations with alterations to caecal microbiota and caecal metabolome linked to infection and vaccination status. Infection heightened microbiota richness with increases in potentially pathogenic species, while vaccination elevated beneficial Bifidobacterium. Using a multi-omics factor analysis, data on caecal microbiota and metabolome were integrated and distinct profiles for healthy, infected, and recovering chickens were identified. Healthy and recovering chickens exhibited higher vitamin B metabolism linked to short-chain fatty acid-producing bacteria, whereas essential amino acid and cell membrane lipid metabolisms were prominent in infected and vaccinated chickens. Notably, vaccinated chickens showed distinct metabolites related to the enrichment of sphingolipids, important components of nerve cells and cell membranes. Our integrated multi-omics model revealed latent biomarkers indicative of vaccination and infection status, offering potential tools for diagnosing infection, monitoring vaccination efficacy, and guiding the development of novel treatments or controls.IMPORTANCEAdvances in anticoccidial vaccines have garnered significant attention in poultry health management. However, the intricacies of vaccine-induced alterations in the chicken gut microbiome and its subsequent impact on host metabolism remain inadequately explored. This study delves into the metabolic and microbiotic shifts in chickens post-vaccination, employing a multi-omics integration analysis. Our findings highlight a notable synergy between the microbiome composition and host-microbe interacted metabolic pathways in vaccinated chickens, differentiating them from infected or non-vaccinated cohorts. These insights pave the way for more targeted and efficient approaches in poultry disease control, enhancing both the efficacy of vaccines and the overall health of poultry populations.
Collapse
Affiliation(s)
- Po-Yu Liu
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Janie Liaw
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - José Jaramillo Ortiz
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- Centre for Vaccinology and Regenerative Medicine, Royal Veterinary College, London, United Kingdom
| | - Fiona M. Tomley
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
| | - Dirk Werling
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- Centre for Vaccinology and Regenerative Medicine, Royal Veterinary College, London, United Kingdom
| | - Ozan Gundogdu
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Damer P. Blake
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
- Centre for Vaccinology and Regenerative Medicine, Royal Veterinary College, London, United Kingdom
| | - Dong Xia
- Pathobiology and Population Sciences, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
48
|
Qiu X, Lu Y, Mu C, Tang P, Liu Y, Huang Y, Luo H, Liu JY, Li X. The Biomarkers in Extreme Longevity: Insights Gained from Metabolomics and Proteomics. Int J Med Sci 2024; 21:2725-2744. [PMID: 39512690 PMCID: PMC11539388 DOI: 10.7150/ijms.98778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/10/2024] [Indexed: 11/15/2024] Open
Abstract
The pursuit of extreme longevity is a popular topic. Advanced technologies such as metabolomics and proteomics have played a crucial role in unraveling complex molecular interactions and identifying novel longevity-related biomarkers in long-lived individuals. This review summarizes key longevity-related biomarkers identified through metabolomics, including high levels of omega-3 polyunsaturated fatty acids (PUFAs), short-chain fatty acids (SCFAs) and sphingolipids, as well as low levels of tryptophan. Proteomics analyses have highlighted longevity-related proteins such as apolipoprotein E (APOE) and pleiotrophin (PTN), along with lower S-nitrosylated and higher glycosylated proteins found from post-translational modification proteomics as potential biomarkers. We discuss the molecular mechanisms that could support the above biomarkers' potential for healthy longevity, including metabolic regulation, immune homeostasis maintenance, and resistance to cellular oxidative stress. Moreover, multi-omics studies of various long-lived cohorts are encompassed, focusing on how the integration of various omics technologies has contributed to the understanding of longevity. This comprehensive review aims to provide new biological insights and pave the way for promoting health span.
Collapse
Affiliation(s)
- Xiaorou Qiu
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Yixian Lu
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Chao Mu
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Peihua Tang
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Yueli Liu
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Yongmei Huang
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Hui Luo
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Jun-Yan Liu
- CNTTI of the Institute of Life Sciences & Anesthesia Department of the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Chongqing Medical University, Chongqing, 400016, China
| | - Xuemeng Li
- Zhanjiang Key Laboratory of Human Microecology and Clinical Translation Research, the Marine Biomedical Research Institute, College of Basic Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| |
Collapse
|
49
|
Li Z, Li Y, Tang X, Xing A, Lin J, Li J, Ji J, Cai T, Zheng K, Lingampelly SS, Li K. Causal Metabolomic and Lipidomic Analysis of Circulating Plasma Metabolites in Autism: A Comprehensive Mendelian Randomization Study with Independent Cohort Validation. Metabolites 2024; 14:557. [PMID: 39452938 PMCID: PMC11509474 DOI: 10.3390/metabo14100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The increasing prevalence of autism spectrum disorder (ASD) highlights the need for objective diagnostic markers and a better understanding of its pathogenesis. Metabolic differences have been observed between individuals with and without ASD, but their causal relevance remains unclear. METHODS Bidirectional two-sample Mendelian randomization (MR) was used to assess causal associations between circulating plasma metabolites and ASD using large-scale genome-wide association study (GWAS) datasets-comprising 1091 metabolites, 309 ratios, and 179 lipids-and three European autism datasets (PGC 2015: n = 10,610 and 10,263; 2017: n = 46,351). Inverse-variance weighted (IVW) and weighted median methods were employed, along with robust sensitivity and power analyses followed by independent cohort validation. RESULTS Higher genetically predicted levels of sphingomyelin (SM) (d17:1/16:0) (OR, 1.129; 95% CI, 1.024-1.245; p = 0.015) were causally linked to increased ASD risk. Additionally, ASD children had higher plasma creatine/carnitine ratios. These MR findings were validated in an independent US autism cohort using machine learning analysis. CONCLUSION Utilizing large datasets, two MR approaches, robust sensitivity analyses, and independent validation, our novel findings provide evidence for the potential roles of metabolomics and circulating metabolites in ASD diagnosis and etiology.
Collapse
Affiliation(s)
- Zhifan Li
- Big Data and Internet of Things Program, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Z.L.); (J.L.); (T.C.); (K.Z.)
| | - Yanrong Li
- Center for Artificial Intelligence-Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Y.L.); (A.X.); (J.L.); (J.J.)
| | - Xinrong Tang
- Yantai Special Education School, Yantai 264001, China;
| | - Abao Xing
- Center for Artificial Intelligence-Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Y.L.); (A.X.); (J.L.); (J.J.)
| | - Jianlin Lin
- Center for Artificial Intelligence-Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Y.L.); (A.X.); (J.L.); (J.J.)
| | - Junrong Li
- Big Data and Internet of Things Program, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Z.L.); (J.L.); (T.C.); (K.Z.)
| | - Junjun Ji
- Center for Artificial Intelligence-Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Y.L.); (A.X.); (J.L.); (J.J.)
| | - Tiantian Cai
- Big Data and Internet of Things Program, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Z.L.); (J.L.); (T.C.); (K.Z.)
| | - Ke Zheng
- Big Data and Internet of Things Program, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Z.L.); (J.L.); (T.C.); (K.Z.)
| | - Sai Sachin Lingampelly
- Department of Medicine, University of California, San Diego School of Medicine, San Diego, CA 92103-8467, USA;
| | - Kefeng Li
- Center for Artificial Intelligence-Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China; (Y.L.); (A.X.); (J.L.); (J.J.)
| |
Collapse
|
50
|
Dhanabalan AK, Devadasan V, Haribabu J, Krishnasamy G. Machine learning models to identify lead compound and substitution optimization to have derived energetics and conformational stability through docking and MD simulations for sphingosine kinase 1. Mol Divers 2024:10.1007/s11030-024-10997-4. [PMID: 39417979 DOI: 10.1007/s11030-024-10997-4] [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: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Sphingosine kinases (SphKs) are a group of important enzymes that circulate at low micromolar concentrations in mammals and have received considerable attention due to the roles they play in a broad array of biological processes including apoptosis, mutagenesis, lymphocyte migration, radio- and chemo-sensitization, and angiogenesis. In the present study, we constructed three classification models by four machine learning (ML) algorithms including naive bayes (NB), support vector machine (SVM), logistic regression, and random forest from 395 compounds. The generated ML models were validated by fivefold cross validation. Five different scaffold hit fragments resulted from SVM model-based virtual screening and docking results indicate that all the five fragments exhibit common hydrogen bond interaction a catalytic residue of SphK1. Further, molecular dynamics (MD) simulations and binding free energy calculation had been carried out with the identified five fragment leads and three cocrystal inhibitors. The best 15 fragments were selected. Molecular dynamics (MD) simulations showed that among these compounds, 7 compounds have favorable binding energy compared with cocrystal inhibitors. Hence, the study showed that the present lead fragments could act as potential inhibitors against therapeutic target of cancers and neurodegenerative disorders.
Collapse
Affiliation(s)
- Anantha Krishnan Dhanabalan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Velmurugan Devadasan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502, Copiapó, Chile
- Chennai Institute of Technology (CIT), Chennai, Tamil Nadu, 600069, India
| | - Gunasekaran Krishnasamy
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, Tamil Nadu, 600025, India.
| |
Collapse
|