1
|
Ruenkoed S, Pholoeng A, Nontasan S, Panprommin D, Mongkolwit K, Wangkahart E. Assessing the impact of acidifiers on growth performance, innate immune capacity, response to ammonia nitrogen stress, digestive enzyme activity, intestinal histology, and gene expression of Nile tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2025; 162:110315. [PMID: 40204243 DOI: 10.1016/j.fsi.2025.110315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/15/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
This study investigated the effects of acidifiers (ACD) supplementation level on the growth, immune response, intestinal histology and gene expression of Nile tilapia (Oreochromis niloticus). The experimental diets included three different levels of ACD concentrations: 0 g/kg (CTRL) as the control, 1 g/kg (ACD1), and 2 g/kg (ACD2). These ACD were sprayed onto the diets and fish were fed to Nile tilapia (7.12 ± 0.47 g) for 56 days. The results indicated that increasing the dietary content of ACD significantly enhanced growth performance, as reflected in higher final body weight, specific growth rate, average daily gain, and feed efficiency. The ACD2 diet showed improved outcomes compared to the CTRL diet. Fish fed ACD diets showed significantly higher antioxidant enzyme activity compared to the CTRL diet and were able to maintain their antioxidant status throughout the experiment. The activity of digestive enzymes, including protease, amylase, and lipase, was significantly increased in fish fed diets supplemented with varying levels of 1 and 2 g/kg diet compared to the CTRL diet (P < 0.05). Additionally, significant increases were observed in the thickness of the intestinal muscularis and the width of the intestinal villi in fish fed ACD at concentrations of 1 and 2 g/kg of the diet, compared to the CTRL diet (P < 0.05). Moreover, dietary supplementation with ACD at concentrations of 2 g/kg of the diet significantly upregulated the expression of immune-related genes (P < 0.05). Overall, the use of ACD in fish diets has shown improvements in growth performance, immune responses, and intestinal histology, indicating their potential as a beneficial feed additive for Nile tilapia aquaculture. To the best of our knowledge, recent evaluations of dietary ACD supplementation in practical diets also highlight the influence of ACD supplementation as a feed additive, which could benefit the aquaculture industry.
Collapse
Affiliation(s)
- Supranee Ruenkoed
- C.P. Vietnam Corporation, Dong Nai, 810000, Viet Nam; Advance Pharma Vietnam Co., Ltd., Ho Chi Minh City, 10000, Viet Nam
| | - Apichet Pholoeng
- Laboratory of Fish Immunology and Nutrigenomics, Applied Animal and Aquatic Sciences Research Unit, Division of Fisheries, Faculty of Technology, Mahasarakham University, Khamriang Sub-district, Kantarawichai, Maha Sarakham, 44150, Thailand
| | - Supap Nontasan
- Laboratory of Fish Immunology and Nutrigenomics, Applied Animal and Aquatic Sciences Research Unit, Division of Fisheries, Faculty of Technology, Mahasarakham University, Khamriang Sub-district, Kantarawichai, Maha Sarakham, 44150, Thailand; Creative Food and Tourism Research Unit, Faculty of Tourism and Hotel Management, Mahasarakham University, Talad Sub-district, Muang, Maha Sarakham, 44000, Thailand
| | - Dutrudi Panprommin
- School of Agriculture and Natural Resources, University of Phayao, Phayao, 56000, Thailand
| | | | - Eakapol Wangkahart
- Laboratory of Fish Immunology and Nutrigenomics, Applied Animal and Aquatic Sciences Research Unit, Division of Fisheries, Faculty of Technology, Mahasarakham University, Khamriang Sub-district, Kantarawichai, Maha Sarakham, 44150, Thailand.
| |
Collapse
|
2
|
El-Belkasy RO, El-Kemary M, Hanafy NAN. Evaluating the role of targeted silymarin loaded hyaluronic acid/protein nanoparticles in activating hepatic progenitor stem cells for liver regeneration after CCl 4-induced liver damage. Int J Biol Macromol 2025; 309:142837. [PMID: 40188925 DOI: 10.1016/j.ijbiomac.2025.142837] [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: 08/02/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Silymarin is a natural flavonoid component isolated from the Silybum Marianum (Milk Thistle) plant with multiple pharmacological activities. We investigated its anti-fibrotic effect on the liver and demonstrated its role in activating hepatic progenitor stem cells during liver regeneration. METHODS Hybrid polymeric protein nanoparticles were prepared by loading silymarin with an albumin-hyaluronic acid complex to achieve stem cell targeting and increase silymarin's bioavailability. RESULTS TEM, Zeta potential, DLS, UV-visible spectrophotometer, Fluorescence analysis, and FTIR verified the successful formation of nanoparticles and efficient encapsulation. In the present study, The liver fibrotic model was induced by the intraperitoneal injection of carbon tetrachloride, followed by the injection of silymarin NPs into mice twice a week for 4 weeks. We evaluated the expression of hepatic fibrosis markers such as (Collagen I, TGF-β1, SMAD3, and MMP-3) and hepatic progenitor stem cell activation markers such as (HNF1β, FOXl1, CD90, Vimentin, and CD105). The results showed that the targeted silymarin NPs caused significant suppression and downregulation of Collagen I, TGF-β, SMAD-3, and MMP-3 and upregulation of the hepatic progenitor stem cells markers HNF1β, FOXl1, CD90, Vimentin, and CD105. They also didn't induce expression of IL-6, IL-1β, and TNF-α, proving that they cause no signs of inflammation. CONCLUSION The novel point is that these results demonstrated that the targeted Silymarin NPs not only could efficiently alleviate CCl4-induced liver fibrosis more than using only free silymarin; by inhibiting the TGF-β/Smad-3 signaling pathway, but also could activate hepatic progenitor stem cells causing liver regeneration.
Collapse
Affiliation(s)
- Rawan O El-Belkasy
- Nanomedicine Department, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Maged El-Kemary
- Nanomedicine Department, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; Nile Valley University, Fayoum 63518, Egypt
| | - Nemany A N Hanafy
- Group of Bionanotechnology and Molecular Cell Biology, Nanomedicine Department, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt; NanoBio4Can program, Koç University Research Center for Translational Medicine (KUTTAM), 34450 Istanbul, Turkey.
| |
Collapse
|
3
|
Song Q, Chen X, Jiang Q, He Z, Su X, Dong C, Xiang H, Song C, Xiong Y, Yang S. Oxalate stimulates macrophage secretion of prostaglandin E2 to promote renal tubular epithelial cell osteogenesis. Life Sci 2025; 366-367:123476. [PMID: 39986650 DOI: 10.1016/j.lfs.2025.123476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
Osteogenesis of renal tubular epithelial cells (RTEC) is an important trigger for calcium oxalate (CaOx) kidney stone formation, but whether macrophages are involved in RTEC osteogenesis is unclear. The purpose of this study was to investigate the role and mechanism of macrophages in CaOx kidney stones on RTEC osteogenesis. Oxalate or ethylene glycol was used to construct in vitro and in vivo CaOx kidney stone models, respectively. Macrophage-derived conditioned medium was used to induce osteogenesis in HK-2 cells, and genetic controls and pharmacological interventions were used to investigate the underlying mechanism. The results demonstrated that macrophage-conditioned medium under oxalate intervention facilitated the increase of alkaline phosphatase and calcium salts as well as the upregulation of osteogenic marker genes (BMP2 and RUNX2) expression in HK-2 cells. On the one hand, the knockdown of the JAK2 gene in HK-2 cells reverses the role of macrophage-derived conditioned medium in promoting osteogenesis in HK-2 cells. On the other hand, inhibition of prostaglandin E2 (PGE2) generation in macrophages reverses osteogenesis in HK-2 cells. Moreover, inhibition of PGE2 generation would cure ethylene glycol-induced renal injury and calcium salt deposition, as well as osteogenesis of RTEC. This study illustrates that in the presence of oxalate, macrophages secret PGE2 to activate JAK2/STAT3 signaling in RTEC, which could trigger osteogenesis. It provides new insights into the mechanism of CaOx kidney stone formation.
Collapse
Affiliation(s)
- Qianlin Song
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China; Central Laboratory, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China
| | - Xin Chen
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China; Central Laboratory, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China
| | - Qinhong Jiang
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China; Central Laboratory, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China
| | - Ziqi He
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China; Central Laboratory, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China
| | - Xiaozhe Su
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China; Central Laboratory, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China
| | - Caitao Dong
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China; Central Laboratory, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China
| | - Heng Xiang
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China; Central Laboratory, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China
| | - Chao Song
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China.
| | - Yunhe Xiong
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China.
| | - Sixing Yang
- Department of Urology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
4
|
Liu L, Bai J, Wang J, Fan J, Yin D, Chang H, Hui X, Yang P. Benzylurea Protects hPDLFs Against LPS-Induced Mitochondrial Dysfunction Through MTCH2. Oral Dis 2025; 31:1255-1267. [PMID: 39491029 DOI: 10.1111/odi.15172] [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: 08/01/2024] [Revised: 09/14/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE The aim of this study is to explore the mechanism of benzylurea in the inflammatory injury of human periodontal ligament fibroblasts (hPDLFs). METHODS An inflammation model of hPDLFs was established using LPS. Nuclear transport of nuclear transcription factor-κB (NF-κB), secretion of cytokines, and the morphology and distribution of F-actin were determined. Mitochondrial function was assessed by measuring mitochondrial membrane potential (MMP), mitochondrial permeability transition pore (mPTP), and reactive oxygen species (ROS) levels. The expression of mitochondrial carrier homolog 2 (MTCH2) and Cytochrome b5 type B (CYB5B) was detected. RESULTS Benzylurea alleviated the effects of lipopolysaccharide (LPS) on the proliferation and apoptosis of hPDLFs. It reduced the release of inflammatory cytokines and inhibited NF-κB nuclear translocation. Benzylurea improved mitochondrial function by regulating MMP and preventing excessive mPTP opening. Furthermore, LPS elevated the expression of MTCH2 and reduced the expression of CYB5B in hPDLFs. However, these effects can be inhibited by benzylurea. The altered expression of MTCH2 directly affected CYB5B expression, the release of inflammatory cytokines, and the activation of nuclear translocation of NF-κB. CONCLUSION CYB5B may act as an effector of MTCH2, with benzylurea enhancing mitochondrial function and protecting hPDLFs from LPS-induced injury through MTCH2.
Collapse
Affiliation(s)
- Li Liu
- Department of Stomatology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Jing Bai
- Department of Stomatology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Jiyun Wang
- Department of Pharmacy, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Junheng Fan
- Department of Stomatology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Dong Yin
- Department of Stomatology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Hong Chang
- School of Stomatology, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xuancheng Hui
- Department of Maxillofacial Surgery, Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Pengfei Yang
- Center for Biomedical Engineering, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
5
|
Xu W, Huang Y, Zhou R. NLRP3 inflammasome in neuroinflammation and central nervous system diseases. Cell Mol Immunol 2025; 22:341-355. [PMID: 40075143 PMCID: PMC11955557 DOI: 10.1038/s41423-025-01275-w] [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: 12/06/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Neuroinflammation plays an important role in the pathogenesis of various central nervous system (CNS) diseases. The NLRP3 inflammasome is an important intracellular multiprotein complex composed of the innate immune receptor NLRP3, the adaptor protein ASC, and the protease caspase-1. The activation of the NLRP3 inflammasome can induce pyroptosis and the release of the proinflammatory cytokines IL-1β and IL-18, thus playing a central role in immune and inflammatory responses. Recent studies have revealed that the NLRP3 inflammasome is activated in the brain to induce neuroinflammation, leading to further neuronal damage and functional impairment, and contributes to the pathological process of various neurological diseases, such as multiple sclerosis, Parkinson's disease, Alzheimer's disease, and stroke. In this review, we summarize the important role of the NLRP3 inflammasome in the pathogenesis of neuroinflammation and the pathological course of CNS diseases and discuss potential approaches to target the NLRP3 inflammasome for the treatment of CNS diseases.
Collapse
Grants
- 81821001, 82130107, 82330052, 82202038, U20A20359 National Natural Science Foundation of China (National Science Foundation of China)
- National Key research and development program of China (grant number (2020YFA0509101), The Strategic Priority Research Program of the Chinese Academy of Sciences (XDB0940000),
- MEXT | JST | Strategic Promotion of Innovative R and D (Strategic Promotion of Innovative R&D)
- the CAS Project for Young Scientists in Basic Research (YSBR-074) and the Fundamental Research Funds for the Central Universities, the outstanding Youth Project of Anhui Provincial Natural Science Foundation (2408085Y049), the Research Start-up Funding of the Institute of Health and Medicine, Hefei Comprehensive National Science Center (2024KYQD004), the Natural Science Foundation of Jiangsu Province (BK20221085),
- The key project of Anhui Provincial Department of Education Fund (2024AH052060).
Collapse
Affiliation(s)
- Wen Xu
- Neurology Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Yi Huang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China.
| | - Rongbin Zhou
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
6
|
Ma Z, Zhang J, Li Z, Zhu Y, Han X, Lei L, Cheng K, Liu W. Interleukin-1β Inhibits Ovarian Cancer Cell Proliferation and Metastasis Through the MAPK/MMP12 Pathway. Int J Mol Sci 2025; 26:3287. [PMID: 40244135 PMCID: PMC11989259 DOI: 10.3390/ijms26073287] [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: 02/10/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Epithelial ovarian cancer (EOC) is a gynecological tumor with high mortality. Despite aggressive treatment, survival rates for patients with advanced EOC are low, and more effective methods of diagnosis and treatment are urgently needed. Inflammation and cancer are strongly associated; however, the mechanisms that mediate this relationship are not fully understood. In this study, we found that the expression of interleukin-1β (IL-1β), a proinflammatory cytokine, increased in an ovarian cancer tissue microarray (TMA) and inhibited A2780 and SKOV3 cell viability and metastasis. Recombinant IL-1β protein and the overexpression of IL-1β decreased the proliferation and metastasis of ovarian cancer cells. IL-1β deficiency promoted proliferation and metastasis. Moreover, transcriptome sequencing revealed that IL-1β downregulates the expression of matrix metalloproteinase 12 (MMP12). The signaling pathway involving MAPK/AP-1/MMP12 is involved in IL-1β-regulated ovarian cancer progression. Overall, we found that the proinflammatory cytokine IL-1β inhibits ovarian cancer cell viability and metastasis. These findings provided deeper insights into inflammation and cancer progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450002, China; (Z.M.); (J.Z.); (Z.L.); (Y.Z.); (X.H.); (L.L.); (K.C.)
| |
Collapse
|
7
|
Hong Y, Li W, Xing Z, Lu M, Tang T, Zhu L, Xiong W, Zhang H, Liu W, Ren S. LRRK2 reduces the sensitivity to TKI and PD-1 blockade in ccRCC via activating LPCAT1. Oncogene 2025:10.1038/s41388-025-03289-0. [PMID: 40121376 DOI: 10.1038/s41388-025-03289-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 03/25/2025]
Abstract
Tyrosine kinase inhibitor (TKI) and immune checkpoint inhibitor (ICI) combination therapy is emerging as a major therapeutic strategy for advanced clear cell renal cell carcinoma (ccRCC). To define the druggable targets for improvement of TKI and ICI combination therapy in ccRCC, we analyzed a commercial protein kinase inhibitor dataset and a public ccRCC dataset and identified LRRK2 as a potential candidate that can be targeted by a small molecule inhibitor. We demonstrated that LRRK2 was transcriptionally upregulated by HIF2A and enabled to drive proliferation of ccRCC cells in a manner independent of its kinase activity. LRRK2 inhibits the RBX1-mediated degradation of lipid metabolism modulator LPCAT1 to reducing the sensitivity to TKI and PD-1 blockade in ccRCC. Specifically, LRRK2/LPCAT1 upregulated IL-1β expression levels through AKT and also increased IL-1β shearing by activating inflammasome. To target the kinase-independent activity of LRRK2, we developed an LR-protac and showed that LR-protac decreased LRRK2 protein level and enhanced the antitumor effect of PD-1 blockade and TKI in ccRCC. These data indicate that LRRK2 is a viable target for improvement of the efficacy of PD-1 blockade and TKI in ccRCC.
Collapse
Affiliation(s)
- Yulong Hong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, 410011, China
| | - Wei Li
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, 410011, China
| | - Zhuo Xing
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, 410011, China
| | - Minghao Lu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Institute of Urologic Science and Technology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tianyu Tang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Institute of Urologic Science and Technology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liang Zhu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, 410011, China
| | - Wei Xiong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, 410011, China
| | - Huan Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wentao Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, 410011, China.
| | - Shangqing Ren
- Robotic Minimally Invasive Surgery Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
8
|
Urbanelli L, Delo F, Cerrotti G, Albertini E, Lucci J, Buratta S, Calzoni E, Giovagnoli S, Lugini L, Federici C, Fratini F, Mercati V, Emiliani C. Cross-Kingdom Communication via Plant-Derived Extracellular Vesicle Nucleic Acids in Genetically Engineered Nicotiana tabacum. Genes (Basel) 2025; 16:356. [PMID: 40149507 PMCID: PMC11942166 DOI: 10.3390/genes16030356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Plants release extracellularly lipid bilayer-enclosed vesicles of nanometric size that can be retrieved in their fluids. Plant-derived extracellular vesicles (PDEVs) have mostly been involved in modulating host-pathogen interaction, making them a tool for cross-kingdom communication with a key role in plant immunity. In addition, PDEVs have demonstrated promising therapeutic features, not only in terms of intrinsic nutraceutical properties but also of active molecules' delivery. Transgenic plants have been developed for a variety of purposes, i.e., to improve their functional properties like crops, but also to produce therapeutic molecules. However, it is unclear whether transgenes can end up in PDEVs, thus making them a vehicle for their cross-kingdom diffusion into the environment. METHODS Here, we investigated the association of transgenic DNA and RNA with PDEVs secreted by tobacco (Nicotiana tabacum) engineered to express the neomycine phosphotransferase II (Npt-II) gene. PDEVs were isolated from leaf apoplastic fluid by ultracentrifugation and characterized for their morphology and size. The association of DNA and RNA was assessed by qRT-PCR and their immunomodulatory properties by assaying PDEVs-induced IL1β and IL10 on THP1 monocytes. RESULTS Npt-II RNA, but not DNA, could be amplified from PDEVs, whereas no differences were observed between wt and transgenic tobacco PDEVs in terms of immunomodulatory properties. CONCLUSIONS Although a different behaviour by other types of RNAs or DNAs could still be possible, our findings indicate that in this model, PDEVs are not associated with transgenic DNA, but they can protect RNA, including transgenic RNA, from degradation, contributing to their cross-kingdom spreading.
Collapse
Affiliation(s)
- Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (L.U.); (G.C.); (S.B.); (E.C.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Federica Delo
- Bios-Therapy, Physiological Systems for Health S.p.A., Loc. Aboca 20, Sansepolcro, 52037 Arezzo, Italy; (F.D.); (J.L.)
| | - Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (L.U.); (G.C.); (S.B.); (E.C.)
| | - Emidio Albertini
- Department of Agricultural, Food and Environmental Sciences, University of Perugia, Borgo XX Giugno 74, 06121 Perugia, Italy;
| | - Jacopo Lucci
- Bios-Therapy, Physiological Systems for Health S.p.A., Loc. Aboca 20, Sansepolcro, 52037 Arezzo, Italy; (F.D.); (J.L.)
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (L.U.); (G.C.); (S.B.); (E.C.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (L.U.); (G.C.); (S.B.); (E.C.)
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy;
| | - Luana Lugini
- Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy; (L.L.); (C.F.); (F.F.)
| | - Cristina Federici
- Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy; (L.L.); (C.F.); (F.F.)
| | - Federica Fratini
- Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy; (L.L.); (C.F.); (F.F.)
| | - Valentino Mercati
- Bios-Therapy, Physiological Systems for Health S.p.A., Loc. Aboca 20, Sansepolcro, 52037 Arezzo, Italy; (F.D.); (J.L.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (L.U.); (G.C.); (S.B.); (E.C.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| |
Collapse
|
9
|
Erasha AM, EL-Gendy H, Aly AS, Fernández-Ortiz M, Sayed RKA. The Role of the Tumor Microenvironment (TME) in Advancing Cancer Therapies: Immune System Interactions, Tumor-Infiltrating Lymphocytes (TILs), and the Role of Exosomes and Inflammasomes. Int J Mol Sci 2025; 26:2716. [PMID: 40141358 PMCID: PMC11942452 DOI: 10.3390/ijms26062716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Understanding how different contributors within the tumor microenvironment (TME) function and communicate is essential for effective cancer detection and treatment. The TME encompasses all the surroundings of a tumor such as blood vessels, fibroblasts, immune cells, signaling molecules, exosomes, and the extracellular matrix (ECM). Subsequently, effective cancer therapy relies on addressing TME alterations, known drivers of tumor progression, immune evasion, and metastasis. Immune cells and other cell types act differently under cancerous conditions, either driving or hindering cancer progression. For instance, tumor-infiltrating lymphocytes (TILs) include lymphocytes of B and T cell types that can invade malignancies, bringing in and enhancing the ability of immune system to recognize and destroy cancer cells. Therefore, TILs display a promising approach to tackling the TME alterations and have the capability to significantly hinder cancer progression. Similarly, exosomes and inflammasomes exhibit a dual effect, resulting in either tumor progression or inhibition depending on the origin of exosomes, type of inflammasome and tumor. This review will explore how cells function in the presence of a tumor, the communication between cancer cells and immune cells, and the role of TILs, exosomes and inflammasomes within the TME. The efforts in this review are aimed at garnering interest in safer and durable therapies for cancer, in addition to providing a promising avenue for advancing cancer therapy and consequently improving survival rates.
Collapse
Affiliation(s)
- Atef M. Erasha
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Hanem EL-Gendy
- Department of Pharmacology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Ahmed S. Aly
- Department of Animal Production, Faculty of Agriculture, Ain Shams University, Cairo 11241, Egypt;
| | - Marisol Fernández-Ortiz
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Ramy K. A. Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt;
| |
Collapse
|
10
|
Ibrahim S, Yousef EH, El-Dessouki AM, Raslan NA, Alzokaky AA. Melatonin augments anti-tumor activity and alleviates nephrotoxicity of gemcitabine in a pancreatic cancer xenograft model targeting P62/Keap1 pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03938-x. [PMID: 40100373 DOI: 10.1007/s00210-025-03938-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 02/16/2025] [Indexed: 03/20/2025]
Abstract
Although gemcitabine is a primary chemotherapy for pancreatic cancer, its effectiveness is limited by chemoresistance and nephrotoxicity, posing significant clinical challenges. Therefore, the development of novel therapeutic approaches to prevent pancreatic malignancy remains crucial. This study aimed to investigate the potential of melatonin in enhancing gemcitabine's anticancer efficacy while mitigating its nephrotoxic effects through modulation of the Keap1/p62 pathway. A pancreatic cancer xenograft model was established in rats, which received either gemcitabine (50 mg/kg, I.P.), melatonin (50 mg/kg, I.P.), or their combination three times per week for 2 weeks. Our findings demonstrate that melatonin potentiates gemcitabine's cancer-suppressing effects via modulation of the Kelch-like-ECH associated protein-1 (Keap1)/p62 pathway, resulting in reduced fibrosis, oxidative stress, and inflammatory markers. Additionally, melatonin significantly mitigated gemcitabine-induced nephrotoxicity. These results suggest that melatonin may serve as an adjuvant therapy in pancreatic cancer treatment, enhancing chemotherapy efficacy while reducing its adverse effects.
Collapse
Affiliation(s)
- Samar Ibrahim
- Pharmacy Practice and Clinical Pharmacy Department, Faculty of Pharmacy, Galala University, Ataka, Egypt
| | - Eman H Yousef
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Ahmed M El-Dessouki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Giza, 12566, Egypt
| | - Nahed A Raslan
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11651, Egypt
- Department of Clinical Pharmacy Program, College of Health Sciences and Nursing, Al-Rayan Colleges, AL-Madina AL-Munawarah, Saudi Arabia
| | - Amany A Alzokaky
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt.
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11651, Egypt.
| |
Collapse
|
11
|
Coelho MM, Moreira FC, Zuccherato LW, Ventura LHDA, Camatta GC, Starling-Soares B, Torres L, Durso DF, Sato HI, da Costa MS, Guimarães HC, Barbuto RC, Júnior MLO, Speziali E, Tupinambas U, Teixeira SMR, Silveira-Nunes G, Teixeira-Carvalho A, Maioli TU, Faria AMC. Living in endemic area for infectious diseases is associated to differences in immunosenescence and inflammatory signatures. Front Immunol 2025; 16:1547854. [PMID: 40165959 PMCID: PMC11955481 DOI: 10.3389/fimmu.2025.1547854] [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: 12/18/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Research on aged individuals from developed countries show that lifestyle factors such as diet, physical activity, stress, smoking, and sleep quality impact aging. However, other relevant factors may influence aging in less-studied populations, such as Brazilian cohorts. This study aimed to analyze immunosenescence profile of individuals living in an endemic area for several infectious diseases in Brazil. We showed that these individuals exhibited accelerated epigenetic aging and increased production of IL-12p70, IL-17A, and IL-9. Production of inflammatory mediators IL-12p70, IL-6, IL-1β, IL-2, and IL-1ra in individuals with flu-like symptoms and those with COVID-19 was higher among residents in endemic areas than in residents from a control non-endemic area. Furthermore, residents of the endemic area had a more prominent inflammatory profile during viral infection and a different pattern of plasma mediators when compared to residents of a non-endemic area. Our data suggests that these two cohorts had specific immune signatures regardless of the presence or the type of infection at study. Therefore, we demonstrated that there were distinct patterns of immune responses and epigenetic aging depending on the environment the individuals live in. These observations add a layer of diversity to the studies of human aging by including individuals from less represented regions.
Collapse
Affiliation(s)
- Monique Macedo Coelho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Caixeta Moreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana Werneck Zuccherato
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Haniel de Araújo Ventura
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Giovanna Caliman Camatta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bernardo Starling-Soares
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lícia Torres
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danielle Fernandes Durso
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Hugo Itaru Sato
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Murilo Soares da Costa
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | - Elaine Speziali
- Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
- Universidade Edson Antônio Velano, Fundação de Ensino e Tecnologia de Alfenas, Belo Horizonte, Brazil
| | - Unaí Tupinambas
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Santuza Maria Ribeiro Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Silveira-Nunes
- Departamento de Medicina, Universidade Federal de Juiz de Fora, Governador Valadares, Brazil
| | | | - Tatiani Uceli Maioli
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
12
|
Jin Y, Zhang C, Jia M, Chen M. Enhanced Dermal Delivery of Nanoparticulate Formulation of Cutibacterium acnes Using Sponge Spicules for Atopic Dermatitis Treatment. Int J Nanomedicine 2025; 20:3235-3249. [PMID: 40103747 PMCID: PMC11917440 DOI: 10.2147/ijn.s509798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Introduction The pathogenesis of atopic dermatitis (AD) is closely linked to both genetic and environmental factors, with patients often exhibiting a range of immunological abnormalities, including a pronounced Th2-type overreaction, which is a key feature of the disease. Purpose Cutibacterium acnes has been shown to induce a robust Th1 immune response through intraperitoneal injections, potentially preventing the development of AD. In this study, a novel nanoparticulate formulation of Cutibacterium acnes (NFCA) was developed with the formulation optimization for the dermal delivery. Materials and Methods Sponge Haliclona sp. spicules (SHS) were isolated from the explants of sponge Haliclona sp. with our proprietary method. The NFCA was prepared by high-speed grinding followed by film extrusion. The skin penetration of the model drugs in NFCA with SHS were visualized using confocal microscopy. The therapeutic effects of NFCA coupled with SHSs against AD in mice were assessed by using pathohistological examination and cytokine ELISA assay. Results The NFCA particle size was 254.1±39.4 nm, with a PDI of 0.29±0.08 and a Zeta potential of -7.9±0.6 mV. SHS significantly enhanced total skin absorption of FD10K (39.6±6.7%, p=0.00076) as well as deposition in the viable epidermis (3.2±1.6%, p=0.08) and deep skin (dermis & receptor) (36.0±5.9%, p=1.82E-5) compared to the control. In vitro cytotoxicity tests showed that NFCA had low toxicity to HaCaT cells (IC50=63.8 mg/mL). The study confirmed that NFCA can activate immune signaling pathways, promoting the high expression of IL-6 and IL-8 in keratinocytes, enhancing TNF-α and IL-1β expression in macrophages, and inducing Th1 and Th17-type immune responses. Furthermore, we demonstrated that the dermal delivery of NFCA using SHS in vivo significantly reduced epidermal thickness, serum IgE levels, and tissue IL-4 levels, thereby accelerating skin repair and mitigating Th2 polarization. Conclusion SHS were employed to effectively deliver NFCA to the deeper skin layers to exert its immune functions. Moreover, the combination of SHS and NFCA can significantly cure mice with atopic dermatitis.
Collapse
Affiliation(s)
- Youmei Jin
- Department of Marine Biological Science & Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Chi Zhang
- Department of Marine Biological Science & Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Mengnan Jia
- Department of Marine Biological Science & Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Ming Chen
- Department of Marine Biological Science & Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, 361102, People's Republic of China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, 361102, People's Republic of China
- Pingtan Research Institute of Xiamen University, Pingtan, 350400, People's Republic of China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518000, People's Republic of China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang'an Biomedicine Laboratory, Xiamen, 361102, People's Republic of China
| |
Collapse
|
13
|
Arias AM, Reinartz DM, Sairs C, Kumar SS, Wilson JE. Streptococcus anginosus Activates the NLRP3 Inflammasome to Promote Inflammatory Responses from Macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642696. [PMID: 40161672 PMCID: PMC11952393 DOI: 10.1101/2025.03.12.642696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chronic inflammation and oral dysbiosis are common features of oral squamous cell carcinoma (OSCC). The commensal streptococci, S. anginosus, is increased in oral diseases including OSCC. Our previous work revealed that S. anginosus promotes inflammatory responses from macrophage cell lines, however the molecular mechanism by which S. anginosus interacts with macrophages to instigate this response remains to be investigated. Here, we expand on our previous findings by investigating the effects of S. anginosus infection of primary bone marrow derived macrophages (BMMs) and during in vivo infection. We found S. anginosus activated primary BMMs, which presented an enlarged cellular area, increased NF-κB activation and downstream inflammatory cytokines TNF⍰, IL-6 and IL-1β at 24 hours post infection. S. anginosus viability was dispensable for NF-κB activation, but essential for the induction of downstream inflammatory proteins and cytokines. S. anginosus persisted intracellularly within BMMs and induced the expression of inflammasome sensors AIM2, NLRC4 and NLRP3. Further, BMMs lacking the inflammasome adapter protein ASC ( Asc -/- ) had significantly diminished IL-1β production compared to wild type BMMs, indicating that S. anginosus activated the inflammasome. S. anginosus primarily triggered the inflammasome through NLRP3 as S. anginosus -infected Nlrp3 -/- BMMs and NLRP3 inhibitor (MCC950)-treated wild type BMMs displayed diminished IL-1β production compared to wild type controls. Lastly, S. anginosus -infected Asc -/- and Nlrp3 -/- mice displayed reduced weight loss compared to C57BL/6 mice. These overall findings indicate that S. anginosus replicates within macrophages and promotes a proinflammatory response in part through activation of the NLRP3 inflammasome. brief summary sentence: S. anginosus replicates intracellularly within macrophages and is sensed by the NLRP3 inflammasome to promote proinflammatory response.
Collapse
|
14
|
Zhou S, Zhou X, Zhang P, Zhang W, Huang J, Jia X, He X, Sun X, Su H. The gut microbiota-inflammation-HFpEF axis: deciphering the role of gut microbiota dysregulation in the pathogenesis and management of HFpEF. Front Cell Infect Microbiol 2025; 15:1537576. [PMID: 40182777 PMCID: PMC11965942 DOI: 10.3389/fcimb.2025.1537576] [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: 12/01/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Heart failure with preserved left ventricular ejection fraction (HFpEF) is a disease that affects multiple organs throughout the body, accounting for over 50% of heart failure cases. HFpEF has a significant impact on individuals' life expectancy and quality of life, but the exact pathogenesis remains unclear. Emerging evidence implicates low-grade systemic inflammation as a crucial role in the onset and progression of HFpEF. Gut microbiota dysregulation and associated metabolites alteration, including short-chain fatty acids, trimethylamine N-oxides, amino acids, and bile acids can exacerbate chronic systemic inflammatory responses and potentially contribute to HFpEF. In light of these findings, we propose the hypothesis of a "gut microbiota-inflammation-HFpEF axis", positing that the interplay within this axis could be a crucial factor in the development and progression of HFpEF. This review focuses on the role of gut microbiota dysregulation-induced inflammation in HFpEF's etiology. It explores the potential mechanisms linking dysregulation of the gut microbiota to cardiac dysfunction and evaluates the therapeutic potential of restoring gut microbiota balance in mitigating HFpEF severity. The objective is to offer novel insights and strategies for the management of HFpEF.
Collapse
Affiliation(s)
- Shenghua Zhou
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xuan Zhou
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Panpan Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Wei Zhang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Jinli Huang
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xuzhao Jia
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xiaole He
- Department of General Practice, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Xin Sun
- Department of Pediatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| | - Hui Su
- Department of Geriatrics, Xijing Hospital, the Fourth Military Medical University, Xi’an, China
| |
Collapse
|
15
|
Chen P, Wang H, Tang Z, Shi J, Cheng L, Zhao C, Li X, Zhou C. Selective Depletion of CCR8+Treg Cells Enhances the Antitumor Immunity of Cytotoxic T Cells in Lung Cancer by Dendritic Cells. J Thorac Oncol 2025:S1556-0864(25)00109-1. [PMID: 40056978 DOI: 10.1016/j.jtho.2025.02.029] [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: 09/24/2024] [Revised: 01/17/2025] [Accepted: 02/22/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Accumulation of regulatory T (Treg) cells, an immunosuppressive population, limits the efficacy of immunotherapy in NSCLC. C-C motif chemokine receptor 8 (CCR8) is selectively expressed in tumor-infiltrating Treg cells and is, therefore, considered an ideal target. METHODS The efficacy and safety of anti-CCR8 monotherapy and its combination with programmed cell death protein-1 (PD1) inhibitor were evaluated in four NSCLC-bearing mice models. To track the dynamic changes in tumor microenvironment, we performed the single-cell RNA sequencing, the single-cell T-cell receptor sequencing analysis, the flow cytometry, the multi-color immunofluorescence, and the Luminex assay on tumors after three, seven, 14, and 21 days of different treatment regimens. Then, in vitro and in vivo experiments were applied to validate our findings and explore molecular mechanisms of the synergistic effects. RESULTS Across four NSCLC-bearing mice models, the combination of CCR8 antibody and PD1 inhibitor significantly reduced tumor growth (p < 0.05) without obvious mouse body weight drops and systemic cytokine storm. The anti-CCR8 therapy synergizes with PD1 blockade by remodeling the tumor microenvironment and disrupting CCR8+Treg-C-C motif chemokine ligand 5 (CCL5)+ dendritic cells (DC) interaction. Mechanistically, therapeutic depletion of CCR8+Treg cells combined with PD1 inhibitor extremely increased interleukin-12 secretion by the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway activation on CCL5+ DCs, thereby promoting cytotoxic activity of CD8+ T cells. The therapeutic potential of the CCR8 antibody LM-108 in combination with immunotherapy was observed in clinical patients with advanced NSCLC. CONCLUSION Overall, CCR8 expression on tumor-infiltrating Treg cells is correlated with immunosuppressive function on DCs and CD8+ T cells, thus impeding antitumor immunity.
Collapse
Affiliation(s)
- Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Zhuoran Tang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Jinpeng Shi
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Lei Cheng
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China; Department of Medical Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China.
| |
Collapse
|
16
|
Ahmadi P, Mahmoudi M, Rafatpanah H, Rezaieyazdi Z, Ahmadi‐Khorram M, Javanmardi Z, Tabasi NS, Esmaeili S. The Impacts of Lactobacillus delbrueckii and Lactobacillus rhamnosus to Promote In Vitro Anti-Inflammatory Profile of RA-Macrophages. Food Sci Nutr 2025; 13:e70068. [PMID: 40099178 PMCID: PMC11911130 DOI: 10.1002/fsn3.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Rheumatoid arthritis (RA) is a prevalent and debilitating autoimmune disease. Numerous studies have demonstrated promising results regarding the use of probiotics as a therapeutic approach to alleviate RA symptoms. This study isolated monocytes from the PBMCs of RA patients and healthy donors. These monocytes were then differentiated into macrophages and divided into five groups: untreated, LPS-treated, L. delbrueckii (Del)-treated, L. rhamnosus (Ram)-treated, and a mixed treatment group. Three macrophage subpopulations-M0, M1, and M2-were identified in all treatment groups, with variations observed in the population percentages of each subpopulation and the expression levels of CD14, CD80, and HLA-DR. Flow cytometry results indicated that, compared to the untreated and LPS-treated groups, treatment with probiotic bacteria (Del, Ram, and Mix) stimulated the polarization of macrophages toward the M2 phenotype while suppressing the percentage of the M1 population. Additionally, the expression of CD14, a Pathogen-Associated Molecular Pattern (PAMP) and phagocytosis-inducing receptor, was significantly reduced in the probiotic-treated groups. Probiotic treatment also profoundly influenced antigen presentation by suppressing CD80, a ligand for the CD28 co-stimulatory marker on T cells, and HLA-DR, which presents antigens to the T cell receptors of Th4 cells. Interestingly, quantitative real-time PCR results indicated that probiotic treatment of macrophages significantly increased the expression of IL-10 and TGF-β, both anti-inflammatory cytokines, while significantly decreasing the expression of inflammatory cytokines, including IL-12, IL-1β, and TNF-α, in both healthy controls and RA patients. It seems that these probiotics may have a regulatory effect on macrophages, affecting their polarization, antigen presentation patterns, phagocytosis, and cytokine secretion profiles. This suggests that these probiotics may have therapeutic and prophylactic effects on RA.
Collapse
Affiliation(s)
- Parisa Ahmadi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
| | - Mahmoud Mahmoudi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Houshang Rafatpanah
- Division of Inflammation and Inflammatory Diseases, Immunology Research CentreMashhad University of Medical SciencesMashhadIran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research CenterMashhad University of Medical SciencesMashhadIran
| | - Maryam Ahmadi‐Khorram
- Department of Nutrition, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Zahra Javanmardi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
| | | | - Seyed‐Alireza Esmaeili
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Immunology Department, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
17
|
Liu M, Wang C, Hu Q, Wu X, Wang Q, Wang J, Xu K, Lu X, Tian W. Single-cell sequencing revealed the necessity of macrophages in brain microenvironment remodeling by breast cancer metastasis. Transl Oncol 2025; 53:102287. [PMID: 39837060 PMCID: PMC11788856 DOI: 10.1016/j.tranon.2025.102287] [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: 11/30/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
Breast cancer is one of the most common cancers worldwide, 30-50 % of patients with advanced breast cancer develop brain metastasis, causing severe damage to their life quality. Due to the existence of the blood-brain barrier (BBB), brain lesions were recognized to be a unique microenvironment with limited infiltration of circulating immune cells and drugs. However, emerging studies reported the immunology of the brain tumor microenvironment (TME) and indicated the potential of immunotherapy against brain metastases. Therefore, it is of great value to comprehensively investigate the TME and identify the pro-tumoral mechanisms facilitating brain metastases and the crucial molecules involved in this process. In this research, we re-analyzed public data on three brain surgical specimens of breast cancer metastases and identified the immunosuppressive roles of macrophages in the metastatic TME. Then, we conducted the first single-cell RNA sequencing on a murine model of breast cancer brain metastasis. In the brain TME, immune cells showed prominent heterogeneity, especially the mononuclear phagocyte system (MPS). We identified the alteration of macrophage subclusters in the central nerve system (CNS) after breast cancer invasion and found that metastatic cancer cells re-shaped the TME cellular interactions for immune evasion and nutrition supply. Finally, this research could serve as a reference for further analysis of new therapies against brain metastatic lesions.
Collapse
Affiliation(s)
- Maotang Liu
- Department of Neurosurgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, 300041, China; Department of Neurosurgery, The Affiliated Wuxi No.2 People's Hospital, Wuxi 214002, China; Wuxi Neurosurgical Institute, Wuxi, Jiangsu Province, 214002, China
| | - CenZhu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Qin Hu
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - XueChao Wu
- Department of Neurosurgery, The Affiliated Wuxi No.2 People's Hospital, Wuxi 214002, China; Wuxi Neurosurgical Institute, Wuxi, Jiangsu Province, 214002, China
| | - Qing Wang
- Department of Neurosurgery, The Affiliated Wuxi No.2 People's Hospital, Wuxi 214002, China; Wuxi Neurosurgical Institute, Wuxi, Jiangsu Province, 214002, China
| | - Jing Wang
- Department of Neurosurgery, The Affiliated Wuxi No.2 People's Hospital, Wuxi 214002, China; Wuxi Neurosurgical Institute, Wuxi, Jiangsu Province, 214002, China
| | - Kun Xu
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China.
| | - XiaoJie Lu
- Department of Neurosurgery, General Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, 300041, China; Department of Neurosurgery, The Affiliated Wuxi No.2 People's Hospital, Wuxi 214002, China; Wuxi Neurosurgical Institute, Wuxi, Jiangsu Province, 214002, China; Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China.
| | - Wei Tian
- Department of Neurosurgery, The Affiliated Wuxi No.2 People's Hospital, Wuxi 214002, China; Wuxi Neurosurgical Institute, Wuxi, Jiangsu Province, 214002, China; Nanjing Medical University, Nanjing, Jiangsu Province, 211166, China.
| |
Collapse
|
18
|
Casso-Chapa B, González NAV, Le NT, Palaskas NL, Nead KT, Eutsey LP, Samanthapudi VSK, Osborn AM, Lee J, Mejia G, Hoang O, Lin SH, Deswal A, Herrmann J, Wang G, Kirkland JL, Krishnan S, Wehrens XH, Chini EN, Yusuf SW, Iliescu CA, Jain A, Burks JK, Seeley E, Lorenzi PL, Chau KM, Mendoza KCO, Grumbach IM, Brookes PS, Hanssen NM, de Winther MP, Yvan-Charvet L, Kotla S, Schadler K, Abe JI. Reevaluating Anti-Inflammatory Therapy: Targeting Senescence to Balance Anti-Cancer Efficacy and Vascular Disease. Arterioscler Thromb Vasc Biol 2025; 45:372-385. [PMID: 39817327 PMCID: PMC11864897 DOI: 10.1161/atvbaha.124.319870] [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: 06/18/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 01/18/2025]
Abstract
Modulating immune function is a critical strategy in cancer and atherosclerosis treatments. For cancer, boosting or maintaining the immune system is crucial to prevent tumor growth. However, in vascular disease, mitigating immune responses can decrease inflammation and slow atherosclerosis progression. Anti-inflammatory therapy, therefore, presents a unique dilemma for cancer survivors: while it may decrease cardiovascular risk, it might also promote cancer growth and metastasis by suppressing the immune response. Senescence presents a potentially targetable solution to this challenge; senescence increases the risk of both cancer therapy resistance and vascular disease. Exercise, notably, shows promise in delaying this premature senescence, potentially improving cancer outcomes and lowering vascular disease risk post-treatment. This review focuses on the long-term impact of cancer therapies on vascular health. We underscore the importance of modulating senescence to balance cancer treatment's effectiveness and its vascular impact, and we emphasize investigating the role of exercise-mediated suppression of senescence in improving cancer survivorship.
Collapse
Affiliation(s)
- Bernardo Casso-Chapa
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Norma Alicia Vazquez González
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kevin T. Nead
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lydia P. Eutsey
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Division of Cancer Center Support Grant & Extramural Research Development, UT MD Anderson Cancer Center, Houston, TX
| | | | - Abigail M Osborn
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jonghae Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gilbert Mejia
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Oanh Hoang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - James L. Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xander H.T. Wehrens
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cezar A. Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Jared K. Burks
- Department of Leukemia, Division of Center Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Erin Seeley
- Department of Chemistry, University of Texas at Austin, Austin, Texas, USA
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Khanh M. Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Keila Carolina Ostos Mendoza
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | | | - Paul S. Brookes
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Nordin M.J. Hanssen
- Department of (Experimental) Vascular and Internal Medicine, Amsterdam UMC, Amsterdam, the Netherlands
- Diabeter Centrum Amsterdam, Amsterdam, the Netherlands
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam Institute for Immunology and Infectious Diseases (AII), Inflammatory Diseases Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d’Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), 06204 Nice, France
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keri Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
19
|
Živković E, Mitrović-Ajtić O, Subotički T, Ivanović J, Otašević V, Đikić D, Diklić M, Vukotić M, Dragojević T, Stanisavljević D, Antić D, Čokić VP. Thromboinflammatory Biomarkers in Lymphomas: Linking Inflammation to Thrombosis Risk. Int J Mol Sci 2025; 26:2058. [PMID: 40076681 PMCID: PMC11900196 DOI: 10.3390/ijms26052058] [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: 12/27/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Thrombosis is a critical complication in lymphomas, driven by chronic inflammation. To observe this systemic mechanism, we evaluated inflammatory cytokines, neutrophil and monocyte activation, and platelet function in diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and Hodgkin lymphoma (HL), with and without thrombosis using ELISA and flow cytometry according to laboratory and clinical data. Interleukin-1β was elevated across lymphomas and inversely correlated with the Khorana score for venous thromboembolism, while increased tumor necrosis factor-alpha (TNF-α) was inversely associated with the International Prognostic Index (IPI) in thrombosis-associated lymphomas. Neutrophil activation was increased in DLBCL, while elevated neutrophil extracellular traps (NETs) biomarkers were inversely consistent with thrombosis and the ThroLy score. NETs were elevated in HL. Classical monocytes were increased in all lymphoma subtypes, with intermediate and tissue factor (TF)-carrying monocytes elevated in DLBCL and HL. Platelet activation was pronounced, with platelet-monocyte aggregates and platelet-associated TF elevated in DLBCL and FL but not HL. P-selectin was increased in lymphomas with thrombosis, aligned with Khorana and ThroLy scores, and reflected clinical stage while inversely correlating with IPI in non-thrombotic lymphomas. These findings highlight distinct thromboinflammatory mechanisms across lymphoma subtypes, providing insights into biomarkers for thrombosis risk and therapeutic targets in lymphoma management.
Collapse
Affiliation(s)
- Emilija Živković
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| | - Olivera Mitrović-Ajtić
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| | - Tijana Subotički
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| | - Jelena Ivanović
- Lymphoma Center, Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (J.I.); (D.A.)
| | | | - Dragoslava Đikić
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| | - Miloš Diklić
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| | - Milica Vukotić
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| | - Teodora Dragojević
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| | - Dejana Stanisavljević
- Institute for Medical Statistics and Informatics, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Darko Antić
- Lymphoma Center, Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (J.I.); (D.A.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vladan P. Čokić
- Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (E.Ž.); (O.M.-A.); (T.S.); (D.Đ.); (M.D.); (M.V.); (T.D.)
| |
Collapse
|
20
|
Śniadach J, Kicman A, Michalska-Falkowska A, Jończyk K, Waszkiewicz N. Changes in Concentration of Selected Biomarkers of Exposure in Users of Classic Cigarettes, E-Cigarettes, and Heated Tobacco Products-A Narrative Review. Int J Mol Sci 2025; 26:1796. [PMID: 40076424 PMCID: PMC11898610 DOI: 10.3390/ijms26051796] [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: 01/07/2025] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Currently, the number of e-cigarette and heated tobacco product (HTP) users are steadily increasing, while the number of classic cigarette users are decreasing. The effects of smoking classic cigarettes on human health have been thoroughly described in the literature, but the negative health effects of e-cigarettes and HTPs on the human body are not clearly defined. Among users of different forms of tobacco, those at a particularly high risk of developing particular disease entities should be identified, allowing for the faster implementation of potential treatments, including psychotherapeutic ones. Biomarkers are used for this purpose. This paper summarizes the potential of these compounds from the different exposure groups of classic cigarettes, e-cigarettes, and HTPs, and presents changes in their concentrations in the body fluids of different tobacco users. This review discusses the impact of tobacco use in relation to levels of the following biomarkers: TNF-α, IL-1β, IL-6, IL-8, IL-17, IFN-γ, IL-10, IL-4, Il-13, TGF-β, VEGF EGF, HGF, BDNF, MMP-9, CRP, microplastics, and selected parameters of oxidative stress. This review also includes suggested forms of treatment, including Tobacco Product Use Reduction Programs, to minimize the potential negative effects of the above-mentioned products.
Collapse
Affiliation(s)
- Justyna Śniadach
- Department of Psychiatry, The Faculty of Medicine, Medical University of Bialystok, 15-272 Bialystok, Poland;
| | - Aleksandra Kicman
- Department of Aesthetic Medicine, The Faculty of Pharmacy, Medical University of Bialystok, 15-267 Bialystok, Poland;
| | | | - Kamila Jończyk
- Department of Psychiatry, The Faculty of Medicine, Medical University of Bialystok, 15-272 Bialystok, Poland;
| | - Napoleon Waszkiewicz
- Department of Psychiatry, The Faculty of Medicine, Medical University of Bialystok, 15-272 Bialystok, Poland;
| |
Collapse
|
21
|
Zhao S, Li X, Wang Y, Xu R, Li X, Liu J, Hou X, Liu H. Comparison of the Immune Enhancing Activity and Chemical Constituents Between Imitation Wild and Cultivated Astragali Radix. Molecules 2025; 30:923. [PMID: 40005233 PMCID: PMC11858062 DOI: 10.3390/molecules30040923] [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: 01/08/2025] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Astragali Radix (AR), a traditional food and medicinal herb used for thousands of years, is widely recognized for its role in enhancing immunity, particularly when combined with adjuvant chemotherapy. The two primary types of AR available in the market are imitation wild AR (grown for seven years) and cultivated AR (grown for two years). However, whether differences exist in their immune-enhancing effects and chemical constituents remains unclear. In this study, a comparative analysis was performed to evaluate the immune activity and chemical composition of cultivated and imitation wild AR. Immune activity was assessed through in vivo animal studies, while metabolomic analysis was used to characterize their chemical profiles. The results demonstrate that AR possesses significant antitumor and immune-enhancing activities, with imitation wild AR showing superior efficacy compared with cultivated AR. Following 16 days of daily AR treatment, both the thymus and spleen coefficients were significantly increased, effectively reversing the immune dysfunction induced by cyclophosphamide (CTX). Moreover, the administration of AR showed no significant toxicity, as evidenced by the stable liver and kidney function indicators, including ALT, UREA, and CRE levels. To investigate chemical differences, a customized chemotaxonomic-based in-house library containing 215 compounds was developed and integrated with the Progenesis QI informatics platform for metabolite annotation. Using multivariate analysis, 48 constituents were identified in total: 46 unique to the imitation wild AR and 45 specific to the cultivated AR. The correlation between chemical constituents and the pharmacological effects of AR was evaluated. Based on orthogonal partial least-squares discriminant analysis (OPLS-DA) and S-plot analysis, five potential biomarkers were identified, including Calycosin-7-glucoside, Rhamnocitrin-3-O-β-D-glucopyranoside, Ononin, 3,5-Dicaffeoylquinic acid, and Acetylastragaloside I. These biomarkers likely account for the differences in immune-enhancing effects between the two AR types. This study provides a scientific foundation for the rational use of Astragali Radix.
Collapse
Affiliation(s)
- Shuo Zhao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (S.Z.); (Y.W.); (X.L.)
| | - Xueting Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; (X.L.); (R.X.)
| | - Yumeng Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (S.Z.); (Y.W.); (X.L.)
| | - Rui Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; (X.L.); (R.X.)
| | - Xu Li
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (S.Z.); (Y.W.); (X.L.)
| | - Jiushi Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; (X.L.); (R.X.)
| | - Xiaolin Hou
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (S.Z.); (Y.W.); (X.L.)
| | - Haitao Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; (X.L.); (R.X.)
| |
Collapse
|
22
|
Yadav S, Prasannan A, Venkatachalam K, Binesh A. Exploring the mechanism and crosstalk between IL-6 and IL- 1β on M2 macrophages under metabolic stress conditions. Cytokine 2025; 186:156852. [PMID: 39765025 DOI: 10.1016/j.cyto.2024.156852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/14/2025]
Abstract
Macrophages are highly variable immune cells that are important in controlling inflammation and maintaining tissue balance. The ability to polarize into two major types-M1, promoting inflammation, and M2, resolving inflammation and contributing to tissue repair-determines their specific roles in health and disease. M2 macrophages are particularly important for reducing inflammation and promoting tissue regeneration, but their function is shaped mainly by surrounding cells. This is evident in obesity, diabetes, and chronic inflammation. Although many cytokines regulate macrophage polarization, interleukin-6 (IL-6) and interleukin-1β (IL-1β) are major players, but their effects on M2 macrophage behavior under metabolic stress remain unclear. This study describes the intricacies within M2 macrophages concerning IL-6 and IL-1β signaling when under metabolic stress. Though, more frequently than not, IL-6 is labelled as pro-inflammatory, it can also behave as an anti-inflammatory mediator. On the other hand, IL-1β is the main pro-inflammatory agent, particularly in metabolic disorders. The relationship between these cytokines and the macrophages is mediated through important pathways such as JAK/STAT and NFκB, which get perturbed by metabolic stress. Therefore, metabolic stress also alters the functional parameters of macrophages, including alterations in mitochondrial metabolism, glycolytic and oxidative metabolism. Phosphorylation alters the kinetics involved in energy consumption and affects their polarization and their function. However, it has been suggested that IL-6 and IL-1β may work in concert or competition when inducing M2 polarization and, importantly, implicate cytokine release, phagocytic activity, and tissue repair processes. In this review, we discuss the recent literature on the participation of IL-6 and IL-1β cytokines in macrophage polarization and how metabolic stress changes cytokine functions and synergistic relations. A better understanding of these cytokines would serve as an important step toward exploring alternative antiviral strategies directed against metabolic disturbance and, hence, approve further endeavors.
Collapse
Affiliation(s)
- Shawna Yadav
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Anusha Prasannan
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Ambika Binesh
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India.
| |
Collapse
|
23
|
Zhang W, Chen Y, Yao Z, Ouyang M, Sun M, Zou S. Post-Marketing Pharmacovigilance of Canakinumab from the FDA Adverse Event Reporting System (FAERS). Pharmaceuticals (Basel) 2025; 18:114. [PMID: 39861175 PMCID: PMC11768236 DOI: 10.3390/ph18010114] [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: 12/13/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Canakinumab, a humanized anti-IL-1β monoclonal antibody, is known for its ability to suppress IL-1β-mediated inflammation. However, continuous monitoring of its safety remains essential. Thus, we comprehensively evaluated the safety signals of canakinumab by data mining from FAERS. Methods: We used a disproportionate analysis to quantify canakinumab-related adverse events (AEs) using four algorithms. Clinical prioritization of the detected signals was assessed with a semiquantitative score method. Serious and non-serious outcomes were compared by statistical methods. Additionally, a stratification analysis of serious infections was conducted at the system organ class (SOC) level. Results: A total of 28,496 canakinumab-related AEs were collected, and 71 suspicious signals detected. Among these, 19 preferred terms (PTs) were identified as unexpected signals, including deafness, appendicitis, brain oedema, cushingoid, cellulitis, and papilledema. Of the AEs, 16 were more likely reported as serious outcomes, such as pneumonia, abdominal pain, deafness, and infection. Based on clinical priority score, 44 PTs were classified as weak, 27 as moderate, and none as strong. Furthermore, 30 PTs demonstrated a high level of evidence, primarily derived from FDA prescribing information, randomized controlled trials, and systematic reviews. Stratification analysis of infections and infestations (serious outcomes) revealed a stronger association of severe infections with canakinumab in older or heavier individuals. All positive signals followed an early failure pattern, with the incidence of canakinumab-associated AEs decreasing over time. Conclusions: We found that most of the suspicious signals were associated with infections. More attention should be paid to serious infections, particularly in males, individuals aged ≥60 years, or those weighing >100 kg, who demonstrated the highest risk of serious infections.
Collapse
Affiliation(s)
- Weidong Zhang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.Z.)
- Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yunzhou Chen
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.Z.)
| | - Zeyu Yao
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.Z.)
| | - Mengling Ouyang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.Z.)
| | - Minghui Sun
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.Z.)
| | - Shupeng Zou
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.Z.)
| |
Collapse
|
24
|
Gallucci G, Larocca M, Navazio A, Turazza FM, Inno A, Canale ML, Oliva S, Besutti G, Tedeschi A, Aschieri D, Russo A, Gori S, Silvestris N, Pinto C, Tarantini L. Atherosclerosis and the Bidirectional Relationship Between Cancer and Cardiovascular Disease: From Bench to Bedside, Part 2 Management. Int J Mol Sci 2025; 26:334. [PMID: 39796190 PMCID: PMC11719480 DOI: 10.3390/ijms26010334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
The first part of this review highlighted the evolving landscape of atherosclerosis, noting emerging cardiometabolic risk factors, the growing impact of exposomes, and social determinants of health. The prominent role of atherosclerosis in the bidirectional relationship between cardiovascular disease and cancer was also discussed. In this second part, we examine the complex interplay between multimorbid cardio-oncologic patients, cardiometabolic risk factors, and the harmful environments that lend a "syndemic" nature to these chronic diseases. We summarize management strategies targeting disordered cardiometabolic factors to mitigate cardiovascular disease and explore molecular mechanisms enabling more tailored therapies. Importantly, we emphasize the early interception of atherosclerosis through multifactorial interventions that detect subclinical signs (via biomarkers and imaging) to treat modifiable risk factors and prevent clinical events. A concerted preventive effort-referred to by some as a "preventome"-is essential to reduce the burden of atherosclerosis-driven chronic diseases, shifting from mere chronic disease management to the proactive promotion of "chronic health".
Collapse
Affiliation(s)
| | - Mario Larocca
- Provincial Medical Oncology, Department of Oncology and Advanced Technologies, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy; (M.L.); (C.P.)
| | - Alessandro Navazio
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| | | | - Alessandro Inno
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy; (A.I.)
| | - Maria Laura Canale
- Division of Cardiology, Azienda USL Toscana Nord-Ovest, Versilia Hospital, 55041 Lido di Camaiore, Italy;
| | - Stefano Oliva
- UOSD Cardiologia di Interesse Oncologico IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Giulia Besutti
- Radiology Unit, Department of Imaging and Laboratory Medicine, AUSL—IRCCS di Reggio Emilia, 42100 Reggio Emilia, Italy;
- Department of Surgical and Medical Sciences of Children and Adults, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Andrea Tedeschi
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29100 Piacenza, Italy; (A.T.); (D.A.)
| | - Daniela Aschieri
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29100 Piacenza, Italy; (A.T.); (D.A.)
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy;
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy; (A.I.)
| | - Nicola Silvestris
- Medical Oncology Department, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Carmine Pinto
- Provincial Medical Oncology, Department of Oncology and Advanced Technologies, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy; (M.L.); (C.P.)
| | - Luigi Tarantini
- Cardiologia Ospedaliera, Department of Specialized Medicine, AUSL—IRCCS in Tecnologie Avanzate e Modelli Assistenziali in Oncologia, 42100 Reggio Emilia, Italy;
| |
Collapse
|
25
|
Guo C, Sun H, Du Y, Dai X, Pang Y, Han Z, Xiong X, Li S, Zhang J, Zheng Q, Gui X. Specifically blocking αvβ8-mediated TGF-β signaling to reverse immunosuppression by modulating macrophage polarization. J Exp Clin Cancer Res 2025; 44:1. [PMID: 39743547 PMCID: PMC11697059 DOI: 10.1186/s13046-024-03250-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Targeting the TGF-β pathway in tumor therapy has proven challenging due to the highly context-dependent functions of TGF-β. Integrin αvβ8, a pivotal activator of TGF-β, has been implicated in TGF-β signaling within tumors, as demonstrated by the significant anti-tumor effects of anti-αvβ8 antibodies. Nevertheless, the expression profile of αvβ8 remains a subject of debate, and the precise mechanisms underlying the anti-tumor effects of anti-αvβ8 antibodies are not yet fully elucidated. METHODS We utilized single-cell RNA sequencing to assess αvβ8 expression across various human tumors. An anti-αvβ8 antibody was developed and characterized for its binding and blocking properties in vitro. Cryo-EM single-particle analysis was employed to study the detailed interaction between αvβ8 and the antibody Fab fragment. The anti-tumor efficacy of the antibody was evaluated in syngeneic mouse models with varying levels of αvβ8 expression, both as a monotherapy and in combination with PD-1 antibodies. Human PBMCs were isolated to investigate αvβ8 expression in myeloid cells, and macrophages were exposed to the antibody to study its impact on macrophage polarization. Pharmacokinetic studies of the αvβ8 antibody were conducted in cynomolgus monkeys. RESULTS Integrin αvβ8 is notably expressed in certain tumor types and tumor-infiltrating macrophages. The specific αvβ8 antibody 130H2 demonstrated high affinity, specificity, and blocking potency in vitro. Cryo-EM analysis further revealed that 130H2 interacts exclusively with the β8 subunit, without binding to the αv subunit. In vivo studies showed that this antibody significantly inhibited tumor growth and alleviated immunosuppression by promoting immune cell infiltration. Furthermore, combining the antibody with PD-1 inhibition produced a synergistic anti-tumor effect. In human PBMCs, monocytes exhibited high αvβ8 expression, and the antibody directly modulated macrophage polarization. Tumors with elevated αvβ8 expression were particularly responsive to 130H2 treatment. Additionally, favorable pharmacokinetic properties were observed in cynomolgus monkeys. CONCLUSIONS In summary, integrin αvβ8 is highly expressed in certain tumors and tumor-infiltrating macrophages. Targeting αvβ8 with a blocking antibody significantly inhibits tumor growth by modulating macrophage polarization and enhancing immune cell infiltration. Combining αvβ8 targeting with PD-1 treatment markedly increases the sensitivity of immune-excluded tumors. These results support further clinical evaluation of αvβ8 antibodies.
Collapse
Affiliation(s)
- Cuicui Guo
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Hui Sun
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Yulei Du
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Xiaodong Dai
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Yu Pang
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Zhen Han
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China
| | - Xinhui Xiong
- Nanjing Novoacine Biotechnology Co., Ltd, Nanjing, 210032, China
| | - Shaowei Li
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China
| | - Junhua Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China.
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, China.
| | - Xun Gui
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China.
| |
Collapse
|
26
|
Yin G, Chen X, Zhang Y, Huang Z, Guo W. Exploration of the role of AIM2/IL-1β in adult laryngeal papilloma. Sci Prog 2025; 108:368504241292457. [PMID: 40025877 PMCID: PMC11874212 DOI: 10.1177/00368504241292457] [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: 03/04/2025]
Abstract
OBJECTIVES Human papillomavirus (HPV) infection is closely related to upper respiratory mucosal lesions, and the most common disease is adult upper respiratory papilloma, which has a certain probability of cancer transformation.This study conducted in vitro tissue and cell experiments to explore the inflammatory mechanisms associated with HPV + adult laryngeal papilloma. METHODS We compared differential expression of AIM2 and IL-1β between HPV (High-risk) negative and positive adult laryngeal papilloma patients. In vitro experiments were conducted to investigate the differences in expression of AIM2, Caspase-1, and IL-1β in HPV- and HPV+ upper respiratory mucosal cells. RESULTS The expression level of AIM2 and IL-1β was higher in HPV (High-risk) positive papilloma tissue than HPV (High-risk) negative papilloma tissue. The expression of AIM2, Caspase-1, and IL-1β in HPV+ cells was also significantly higher than in HPV- cells. CONCLUSIONS The expression of IL-1β mediated by AIM2 was associated with chronic inflammation of upper respiratory mucosal tissue caused by HPV infection, and it may yet be associated with further pathological changes.
Collapse
Affiliation(s)
- Gaofei Yin
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaohong Chen
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhigang Huang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wei Guo
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Fernandes Q, Folorunsho OG. Unveiling the nexus: The tumor microenvironment as a strategic frontier in viral cancers. Cytokine 2025; 185:156827. [PMID: 39647395 DOI: 10.1016/j.cyto.2024.156827] [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: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Viral infections are a significant factor in the etiology of various cancers, with the tumor microenvironment (TME) playing a crucial role in disease progression. This review delves into the complex interactions between viruses and the TME, highlighting how these interactions shape the course of viral cancers. We explore the distinct roles of immune cells, including T-cells, B-cells, macrophages, and dendritic cells, within the TME and their influence on cancer progression. The review also examines how viral oncoproteins manipulate the TME to promote immune evasion and tumor survival. Unraveling these mechanisms highlights the emerging paradigm of targeting the TME as a novel approach to cancer treatment. Our analysis provides insights into the dynamic interplay between viruses and the TME, offering a roadmap for innovative treatments that leverage the unique characteristics of viral cancers.
Collapse
Affiliation(s)
- Queenie Fernandes
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, PO. Box 3050, Doha, Qatar.
| | - Oginni Gbenga Folorunsho
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 5000, Nova Gorica, Slovenia
| |
Collapse
|
28
|
Liu C, Wu K, Li C, Zhang Z, Zhai P, Guo H, Zhang J. SPP1+ macrophages promote head and neck squamous cell carcinoma progression by secreting TNF-α and IL-1β. J Exp Clin Cancer Res 2024; 43:332. [PMID: 39726047 DOI: 10.1186/s13046-024-03255-w] [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: 07/31/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a very aggressive disease characterized by a heterogeneous tumor immune microenvironment (TIME). Tumor-associated macrophages (TAMs) constitute the major innate immune population in the TIME where they facilitate crucial regulatory processes that participate in malignant tumor progression. SPP1 + macrophages (SPP1 + Macs) are found in many cancers, but their effects on HNSCC remain unknown. This study aimed to identify and validate the role and function of SPP1 + Macs in the malignant progression of HNSCC. METHODS In this study, we applied single-cell RNA sequencing (scRNA-seq) analyses of paired tumor and normal tissues from 5 HNSCC patients to identify tumor-specific SPP1 + Macs. RT-qPCR and multiplex immunohistochemical and multiplex immunofluorescence staining were used to verify the presence of SPP1 + Macs in the clinical samples. Gene set variation analysis suggested that SPP1 + Macs were actively involved in cytokine production. Thus, we constructed SPP1-OE macrophages and SPP1-KD macrophages (both differentiated from THP-1 cells), performed a Luminex liquid suspension chip detection assay to detect differential cytokines, and further assessed their biological functions and mechanisms in several HNSCC cell lines and adjacent macrophages. An in vivo experiment was used to verify the function of SPP1 + Macs in HNSCC progression. RESULTS The scRNA-seq results revealed that myeloid cells were heterogeneous and strongly correlated with tumor cells in the TIME in HNSCC and identified tumor-specific SPP1 + Macs, which were positively correlated with poor prognosis of HNSCC patients. Gene set variation analysis (GSVA) suggested that SPP1 + Macs were actively involved in cytokine production. Luminex liquid suspension chip detection assay indicated that SPP1 + Mac-derived TNF-α and IL-1β played important roles. Both in vitro and in vivo experiments and the use of VGX-1027, an inhibitor of macrophage-derived TNF-α and IL-1β, confirmed that SPP1 + Mac-derived TNF-α and IL-1β promoted HNSCC progression by supporting tumor cell proliferation and migration. Mechanistically, we found that TNF-α and IL-1β were upregulated due to NF-kappa B signaling pathway activation in SPP1 + Macs. Moreover, SPP1 + Mac-derived TNF-α and IL-1β promoted the expression of OPN in both tumor cells and other adjacent macrophages through different signaling pathways. CONCLUSIONS SPP1 + Macs increase the secretion of TNF-α and IL-1β via the NF-kappa B pathway to promote HNSCC cell proliferation, and TNF-α and IL-1β in turn upregulate the expression of OPN in tumor cells and macrophages; thus, SPP1 + Macs may be a candidate target through which antitumor efficacy can be enhanced.
Collapse
Affiliation(s)
- Chun Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Wu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
- Department of Oral and Maxillofacial Surgery, Second Xiangya Hospital of Central South University, Changsha, China
| | - Chuwen Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Zhen Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Peisong Zhai
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China
| | - Haiyan Guo
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianjun Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Center for Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- National Clinical Research Center for Oral Diseases, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, China.
- Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Center of Head and Neck Oncology Clinical and Translational Science, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
29
|
Xu L, Peng F, Luo Q, Ding Y, Yuan F, Zheng L, He W, Zhang SS, Fu X, Liu J, Mutlu AS, Wang S, Nehring RB, Li X, Tang Q, Li C, Lv X, Dobrolecki LE, Zhang W, Han D, Zhao N, Jaehnig E, Wang J, Wu W, Graham DA, Li Y, Chen R, Peng W, Chen Y, Catic A, Zhang Z, Zhang B, Mustoe AM, Koong AC, Miles G, Lewis MT, Wang MC, Rosenberg SM, O'Malley BW, Westbrook TF, Xu H, Zhang XHF, Osborne CK, Li JB, Ellis MJ, Rimawi MF, Rosen JM, Chen X. IRE1α silences dsRNA to prevent taxane-induced pyroptosis in triple-negative breast cancer. Cell 2024; 187:7248-7266.e34. [PMID: 39419025 PMCID: PMC11645245 DOI: 10.1016/j.cell.2024.09.032] [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: 12/06/2022] [Revised: 07/10/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Chemotherapy is often combined with immune checkpoint inhibitor (ICIs) to enhance immunotherapy responses. Despite the approval of chemo-immunotherapy in multiple human cancers, many immunologically cold tumors remain unresponsive. The mechanisms determining the immunogenicity of chemotherapy are elusive. Here, we identify the ER stress sensor IRE1α as a critical checkpoint that restricts the immunostimulatory effects of taxane chemotherapy and prevents the innate immune recognition of immunologically cold triple-negative breast cancer (TNBC). IRE1α RNase silences taxane-induced double-stranded RNA (dsRNA) through regulated IRE1-dependent decay (RIDD) to prevent NLRP3 inflammasome-dependent pyroptosis. Inhibition of IRE1α in Trp53-/- TNBC allows taxane to induce extensive dsRNAs that are sensed by ZBP1, which in turn activates NLRP3-GSDMD-mediated pyroptosis. Consequently, IRE1α RNase inhibitor plus taxane converts PD-L1-negative, ICI-unresponsive TNBC tumors into PD-L1high immunogenic tumors that are hyper-sensitive to ICI. We reveal IRE1α as a cancer cell defense mechanism that prevents taxane-induced danger signal accumulation and pyroptotic cell death.
Collapse
Affiliation(s)
- Longyong Xu
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Fanglue Peng
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qin Luo
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Yao Ding
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Fei Yuan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Liting Zheng
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wei He
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sophie S Zhang
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Xin Fu
- Department of Pathology, Xijing Hospital, Xi'an, Shaanxi 710032, China
| | - Jin Liu
- Department of Pathology, Xijing Hospital, Xi'an, Shaanxi 710032, China
| | - Ayse Sena Mutlu
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shuyue Wang
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ralf Bernd Nehring
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xingyu Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qianzi Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Catherine Li
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiangdong Lv
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Lacey E Dobrolecki
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weijie Zhang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dong Han
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Na Zhao
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric Jaehnig
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jingyi Wang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weiche Wu
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Davis A Graham
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yumei Li
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rui Chen
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Yiwen Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andre Catic
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhibin Zhang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bing Zhang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anthony M Mustoe
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Albert C Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George Miles
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael T Lewis
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C Wang
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | - Susan M Rosenberg
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas F Westbrook
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Han Xu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - C Kent Osborne
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Matthew J Ellis
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mothaffar F Rimawi
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi Chen
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Kim HW, Shin S, Park SH, Park JH, Kim SM, Lee YH, Lee MJ. Next-generation adjuvant systems containing furfurman drives potent adaptive immunity and host defense as a foot-and-mouth disease vaccine adjuvant. Front Immunol 2024; 15:1491043. [PMID: 39742276 PMCID: PMC11687127 DOI: 10.3389/fimmu.2024.1491043] [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: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Many countries use commercial foot-and-mouth disease (FMD) vaccines to prevent FMD pandemics, but these vaccines have disadvantages, such as repeated vaccinations due to the short persistence of antibody (Ab) titers and incomplete host defense despite high Ab titers. To address these shortcomings, we aimed to develop a novel FMD vaccine containing furfurman as an adjuvant. Method To demonstrate the efficacy of the test vaccine, adaptive immunity was evaluated by measuring Ab and neutralizing Ab titers and host defense against viral infections in experimental and target animals. In addition, the expression levels of cytokines [interferon (IFN)α, IFNβ, IFNγ, interleukin (IL)-1β, IL-2, and IL-12p40] were evaluated at the early stages of vaccination to confirm the simultaneous induction of cellular and humoral immune responses induced by the test vaccine. Result The groups that received vaccine containing furfurman showed a strong early, mid-term, and long-term immune response and host defense against viral infections compared to the control groups. The significant upregulation observed in cytokine levels in the furfurman group compared to those in the control groups strongly suggest that the test vaccine strengthens cellular immune response and effectively induces a humoral immune response. Conclusion Our study demonstrated that furfurman, as an FMD vaccine adjuvant, achieves long-lasting immunity and host defense against viral infections by eliciting potent cellular and humoral immune responses. Therefore, our findings contribute to the design of next-generation FMD vaccines and highlight the potential application of furfurman as an adjuvant for other viral diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Min Ja Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
31
|
Zhukova JV, Lopatnikova JA, Alshevskaya AA, Sennikov SV. Molecular mechanisms of regulation of IL-1 and its receptors. Cytokine Growth Factor Rev 2024; 80:59-71. [PMID: 39414547 DOI: 10.1016/j.cytogfr.2024.09.004] [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: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024]
Abstract
Interleukin 1 (IL-1) is a pro-inflammatory cytokine that plays a key role in the development and regulation of nonspecific defense and specific immunity. However, its regulatory influence extends beyond inflammation and impacts a range of immune and non-immune processes. The involvement of IL-1 in numerous biological processes, including modulation of inflammation, necessitates strict regulation at multiple levels. This review focuses on these regulatory processes and discusses their underlying mechanisms. IL-1 activity is controlled at various levels, including receptor binding, gene transcription, expression as inactive proforms, and regulated post-translational processing and secretion. Regulation at the level of the receptor expression - alternative splicing, tissue-specific isoforms, and gene polymorphism - is also crucial to IL-1 functional activity. Understanding these regulatory features of IL-1 will not only continue to shape future research directions but will also highlight promising therapeutic strategies to modulate the biological effects of IL-1.
Collapse
Affiliation(s)
- J V Zhukova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), Novosibirsk 630099, Russia; Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - J A Lopatnikova
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), Novosibirsk 630099, Russia; Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - A A Alshevskaya
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - S V Sennikov
- Federal State Budgetary Scientific Institution "Research Institute of Fundamental and Clinical Immunology" (RIFCI), Novosibirsk 630099, Russia; Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia.
| |
Collapse
|
32
|
Zhang Y, Wang C, Cheng S, Xu Y, Gu S, Zhao Y, Yang J, Wang Y. A Neutrophil Extracellular Traps-Related Signature Predicts Clinical Outcomes and Identifies Immune Landscape in Ovarian Cancer. J Cell Mol Med 2024; 28:e70302. [PMID: 39730971 DOI: 10.1111/jcmm.70302] [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: 08/03/2024] [Revised: 12/01/2024] [Accepted: 12/10/2024] [Indexed: 12/29/2024] Open
Abstract
Ovarian cancer (OvCa) is the most lethal gynaecology malignancies worldwide. Neutrophil extracellular traps (NETs), net-like protein structures produced by activated neutrophils and DNA-histone complexes, have a central role in tumours, though haven't been fully explored in OvCa. We obtained transcriptome data from TCGA-OvCa database (n = 376) as training, ICGC-OvCa database (n = 111) as validation and GTEx database (n = 180) as controls. Through LASSO-COX Regression analysis, we identified an eight-gene signature among 87 NETs-related genes, which was significantly related to poor prognosis in both TCGA-OvCa and ICGC-OvCa cohorts (Log-rank p-value = 0.0003 and 0.0014). Next, we constructed and validated a prognostic nomogram, consist of NETs-related signature and clinical features (C-index = 0.82). We evaluated 22 typical immune cell infiltration through CIBERSORT analysis, which implied upregulation of memory CD4 + T cells, follicular helper T cells and neutrophils in high-risk group. Additionally, we predicted therapy sensitivity through TIDE algorithm, indicating that high NETs-riskscore exhibited more sensitivity towards Sorafenib and less sensitivity towards immunotherapy. We initially reported that RAC2 upregulation was associated with NETs formation and poor prognosis (p-value < 0.05) through IHC analysis of tissue microarrays (n = 125). Conclusively, NETs-related signature was reliable for OvCa prognosis prediction and therapy assessment. Especially, RAC2 was predominantly related to NETs formation, thus providing hints towards anti-tumour mechanism of NETs in OvCa.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Chao Wang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Shanshan Cheng
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Yanna Xu
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Sijia Gu
- Department of Obstetrics and Gynecology, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yaqian Zhao
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Jiani Yang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| | - Yu Wang
- Department of Gynecology, School of Medicine, Shanghai First Maternity and Infant Hospital, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, School of Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, China
| |
Collapse
|
33
|
Postu PA, Boiangiu RS, Mihasan M, Stache AB, Tiron A, Hritcu L. The Distinct Biological Effects of 6-Hydroxy-L-Nicotine in Representative Cancer Cell Lines. Molecules 2024; 29:5593. [PMID: 39683752 DOI: 10.3390/molecules29235593] [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: 10/21/2024] [Revised: 11/12/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
6-hydroxy-L-nicotine (6HLN) is a nicotine (NIC) derivative with proven therapeutic potential in neurodegenerative disorders. Here, the impact of 6HLN on cell growth, migratory behavior, and inflammatory status of three different cancer cell lines (A549, MCF7, and U87) and two normal cell lines (16HBE14o and MCF10A) was investigated. In silico analyses were conducted to evaluate the binding affinity of 6HLN to nicotinic receptors (nAChRs) containing α9 and α5 subunits. The obtained in silico data revealed that 6HLN might act on the cholinergic system. Interestingly, the in vitro data showed the compound has cancer-stimulatory effects in U87 glioblastoma cells and cancer-inhibitory effects in MCF7 breast cancer cells. In A549 lung cancer cells, no changes were detected upon 6HLN administration. More importantly, 6HLN appears not to be deleterious for normal cells, with the viability of 16HBE14o pulmonary cells and MCF10A mammary cells remaining unchanged.
Collapse
Affiliation(s)
- Paula Alexandra Postu
- Center for Fundamental Research and Experimental Development in Translation Medicine-TRANSCEND, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Razvan Stefan Boiangiu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania
| | - Marius Mihasan
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania
| | - Alexandru Bogdan Stache
- Center for Fundamental Research and Experimental Development in Translation Medicine-TRANSCEND, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Adrian Tiron
- Center for Fundamental Research and Experimental Development in Translation Medicine-TRANSCEND, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Lucian Hritcu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania
| |
Collapse
|
34
|
Kim M, Powers CA, Fisher DT, Ku AW, Neznanov N, Safina AF, Wang J, Gautam A, Balachandran S, Krishnamurthy A, Gurova KV, Evans SS, Gudkov AV, Skitzki JJ. Enhancing Anti-PD-1 Immunotherapy by Targeting MDSCs via Hepatic Arterial Infusion in Breast Cancer Liver Metastases. Cancers (Basel) 2024; 16:3711. [PMID: 39518148 PMCID: PMC11545300 DOI: 10.3390/cancers16213711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Surgery, chemotherapy, and radiation often have limited utility for advanced metastatic disease in the liver, and despite its promising activity in select cancers, PD-1 blockade therapy similarly has minimal benefit in this setting. Curaxin, CBL0137, is an experimental anti-cancer drug that disrupts the binding of DNA to histones, destabilizes chromatin, and induces Z-DNA formation which may stimulate anti-tumor immune responses. METHODS Murine cell lines of colon (CT26) and breast (4T1) cancer were interrogated for survival and CBL0137-associated DNA changes in vitro. Immunocompetent models of liver metastases followed by CBL0137 hepatic arterial infusion (HAI) were used to examine in vivo tumor cell DNA alterations, treatment responses, and the immune contexture associated with CBL0137, both alone and in combination with anti-PD-1 therapy. RESULTS CBL0137 induced immediate changes to favor tumor cell death in vitro and in vivo with an efficient tumor uptake via the HAI route. Toxicity to CBL0137 was minimal and anti-tumor treatment effects were more efficient with HAI compared to intravenous delivery. Immune effects were pronounced with CBL0137 HAI with concurrent depletion of a specific population of myeloid-derived suppressor cells and maintenance of effector T cell populations. CONCLUSIONS Combination of CBL0137 HAI with PD-1 blockade improved survival in 4T1 tumors but not in CT26 tumors, and therapeutic efficacy relies on the finding of simultaneous and targeted depletion of myeloid-derived suppressor cells and skewing of T cell populations to produce synergy with PD-1 blockade therapy.
Collapse
Affiliation(s)
- Minhyung Kim
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (C.A.P.); (D.T.F.); (A.W.K.)
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Colin A. Powers
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (C.A.P.); (D.T.F.); (A.W.K.)
| | - Daniel T. Fisher
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (C.A.P.); (D.T.F.); (A.W.K.)
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Amy W. Ku
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (C.A.P.); (D.T.F.); (A.W.K.)
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Nickolay Neznanov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (N.N.); (A.F.S.); (K.V.G.); (A.V.G.)
| | - Alfiya F. Safina
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (N.N.); (A.F.S.); (K.V.G.); (A.V.G.)
| | - Jianmin Wang
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Avishekh Gautam
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (A.G.); (S.B.)
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; (A.G.); (S.B.)
| | - Anuradha Krishnamurthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Katerina V. Gurova
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (N.N.); (A.F.S.); (K.V.G.); (A.V.G.)
| | - Sharon S. Evans
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Andrei V. Gudkov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (N.N.); (A.F.S.); (K.V.G.); (A.V.G.)
| | - Joseph J. Skitzki
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (C.A.P.); (D.T.F.); (A.W.K.)
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| |
Collapse
|
35
|
Silva Z, Rabaça JA, Luz V, Lourenço RA, Salio M, Oliveira AC, Bule P, Springer S, Videira PA. New insights into the immunomodulatory potential of sialic acid on monocyte-derived dendritic cells. Cancer Immunol Immunother 2024; 74:9. [PMID: 39487861 PMCID: PMC11531459 DOI: 10.1007/s00262-024-03863-7] [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: 02/23/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Sialic acids at the cell surface of dendritic cells (DCs) play an important immunomodulatory role, and their manipulation enhances DC maturation, leading to heightened T cell activation. Particularly, at the molecular level, the increased stability of surface MHC-I molecules in monocyte-derived DCs (MoDCs) underpins an improved DC: T cell interaction. In this study, we focused on the impact of sialic acid remodelling by treatment with Clostridium perfringens sialidase on MoDCs' phenotypic and functional characteristics. Our investigation juxtaposes this novel approach with the conventional cytokine-based maturation regimen commonly employed in clinical settings.Notably, C. perfringens sialidase remarkably increased MHC-I levels compared to other sialidases having different specificities, supporting the idea that higher MHC-I is due to the cleavage of specific sialoglycans on cell surface proteins. Sialidase treatment induced rapid elevated surface expression of MHC-I, MHC-II and CD40 within an hour, a response not fully replicated by 48 h cytokine cocktail treatment. These increases were also observable 48 h post sialidase treatment. While CD86 and PD-L1 showed significant increases after 48 h of cytokine maturation, 48 h post sialidase treatment showed a higher increase in CD86 and shorter increase in PD-L1. CCR-7 expression was significantly increased 48 h after sialidase treatment but not significantly affected by cytokine maturation. Both treatments promoted higher secretion of the IL-12 cytokine. However, the cytokine cocktail induced a more pronounced IL-12 production. SNA lectin staining analysis demonstrated that the sialic acid profile is significantly altered by sialidase treatment, but not by the cytokine cocktail, which causes only slight sialic acid upregulation. Notably, the lipid-presenting molecules CD1a, CD1b and CD1c remained unaffected by sialidase treatment in MoDCs, a finding also further supported by experiments performed on C1R cells. Inhibition of endogenous sialidases Neu1 and Neu3 during MoDC differentiation did not affect surface MHC-I expression and cytokine secretion. Yet, sialidase activity in MoDCs was minimal, suggesting that sialidase inhibition does not significantly alter MHC-I-related functions. Our study highlights the unique maturation profile induced by sialic acid manipulation in MoDCs. These findings provide insights into the potential of sialic acid manipulation as a rapid immunomodulatory strategy, offering promising avenues for targeted interventions in inflammatory contexts.
Collapse
Affiliation(s)
- Zélia Silva
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - João Amorim Rabaça
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Vanessa Luz
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Rita Adubeiro Lourenço
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Mariolina Salio
- Medical Research Council Translational Immune Discovery Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX39DS, UK
| | - Alexandra Couto Oliveira
- CIISA‑Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300‑477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300‑477, Lisbon, Portugal
| | - Pedro Bule
- CIISA‑Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300‑477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300‑477, Lisbon, Portugal
| | | | - Paula Alexandra Videira
- Associate Laboratory i4HB, NOVA School of Science and Technology, Institute for Health and Bioeconomy, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
- Department of Life Sciences, Applied Molecular Biosciences Unit, UCIBIO, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
- Department of Life Sciences, CDG & Allies Professionals and Patient Associations International Network (CDG & Allies-PPAIN), NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| |
Collapse
|
36
|
Mumtaz F, Farag BM, Farahat MA, Farouk FA, Aarif MY, Eltresy MH, Amin MH, Habotta OA, Alneghery LM, Alawam AS, Almuqri EA, Aleissa MS, Alhudhaibi AM, Al-Olayan E, Abdel Moneim AE, Ramadan SS. Leek (Allium ampeloprasum var. kurrat) aqueous extract loaded on selenium nanoparticles protects against testis and brain injury induced by mercuric chloride in rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:9062-9075. [PMID: 38993070 DOI: 10.1002/jsfa.13733] [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: 02/13/2024] [Revised: 05/11/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Mercuric chloride (HgCl2) is poisonous to humans and animals and typically damages the nervous system and other organs. Mercuric chloride exposition disclosed to initiation of oxidative stress pathway can result in a defect in male fertility and testis tissue. Synthesized selenium nanoparticles (SeNPs) were characterized with a diameter range minimal than 100 nm, having the effective sets of the biological matter. The present study aimed to evaluate the effect of biosynthesized SeNPs, prepared by leek extract on Wistar rats' testicles and brain. METHODS Thirty-five Wistar male rats (120-150 g) were randomly split into five groups (n = 7), orally ingested with leek aqueous extract loaded on SeNPs, and then the animals were administered with mercury II chloride (HgCl2) to induce testis injury and damage the nervous system. RESULTS The used dose of mercuric chloride led to oxidative stress damage in the testis of the rats which was evidenced by a decrease in testosterone, luteinizing hormone (LH), follicle-stimulating hormone (FSH) and proliferating cell nuclear antigen (PCNA) levels, and an increase in nuclear factor-kappa B (NF-κB) and caspase-3. Also, HgCl2 decreased the levels of dopamine (DA), serotonin (5-HT), norepinephrine (NE) and brain-derived neurotrophic factor (BDNF) in the brains of rats. In addition, A decrease was observed in the levels of antioxidant markers, B-cell lymphoma-2 (Bcl-2), as well as an increase in malondialdehyde (MDA), nitric oxide (NO), NF-κB, tumor necrosis factor (TNF)-α, interleukin (IL)-1β and Bax in both testes and brains. Pre-treatment with leek extract loaded on SeNPs significantly ameliorated testosterone, LH, FSH, PCNA and caspase-3 levels in the testis and DA, 5-HT, NE and BDNF in brains. Although the contents of MDA, NO, TNF-α, IL-1β, NF-κB and Bax decreased significantly in both. glutathione, glutathione peroxidase, glutathione reductase, catalase, superoxide dismutase and Bcl-2 levels were significantly improved in both organs. CONCLUSION Our findings suggest that treatment with aqueous leek extract loaded on SeNPs may offer promising prospects for the advancement of anti-inflammation activity against testis injury and also have a very key role in neurobehavioral alterations as a result of mercury toxicity. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Farah Mumtaz
- Department of Biology, Collage of Science, University of Babylon, Babylon, Iraq
| | - Bahaa M Farag
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mennatullah A Farahat
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Fatma A Farouk
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Moataz Y Aarif
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mohamed H Eltresy
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Menna H Amin
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Abdullah S Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Eman A Almuqri
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Mohammed S Aleissa
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Abdulrahman M Alhudhaibi
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Shimaa S Ramadan
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
37
|
Shen Q, Murakami K, Sotov V, Butler M, Ohashi PS, Reedijk M. Inhibition of Notch enhances efficacy of immune checkpoint blockade in triple-negative breast cancer. SCIENCE ADVANCES 2024; 10:eado8275. [PMID: 39475614 PMCID: PMC11524187 DOI: 10.1126/sciadv.ado8275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Aberrant Notch, which is a defining feature of triple-negative breast cancer (TNBC) cells, regulates intercellular communication in the tumor immune microenvironment (TIME). This includes tumor-associated macrophage (TAM) recruitment through Notch-dependent cytokine secretion, contributing to an immunosuppressive TIME. Despite the low response rate of TNBC to immune checkpoint blockade (ICB), here, we report that inhibition of Notch-driven cytokine-mediated programs reduces TAMs and induces responsiveness to sequentially delivered ICB. This is characterized by the emergence of GrB+ cytotoxic T lymphocytes (CTLs) in the primary tumor. A more impressive effect of sequential treatment is observed in the lung where TAM depletion and increased CTLs are accompanied by near-complete abolition of metastases. This is due to (i) therapeutic reduction in Notch-dependent, prometastatic circulating factors released by the primary tumor, and (ii) elevated PD ligand 1 (PD-L1) in lung metastases, rendering them profoundly sensitive to ICB. These findings highlight the potential of combination cytokine inhibition and ICB as an immunotherapeutic strategy in TNBC.
Collapse
Affiliation(s)
- Qiang Shen
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Kiichi Murakami
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Valentin Sotov
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Marcus Butler
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Pamela S. Ohashi
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King’s College Circle, Room 7205, Toronto, Ontario M5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto Medical Discovery Tower, MaRS Centre, 101 College Street, Room 15-701, Toronto, Ontario M5G 2M9, Canada
| | - Michael Reedijk
- Ontario Cancer Institute, University Health Network, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
- Department of Medical Biophysics, University of Toronto, Toronto Medical Discovery Tower, MaRS Centre, 101 College Street, Room 15-701, Toronto, Ontario M5G 2M9, Canada
- Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Suite 8-411, Toronto, Ontario M5G 2M9, Canada
| |
Collapse
|
38
|
Skapinker E, Aucoin EB, Kombargi HL, Yaish AM, Li Y, Baghaie L, Szewczuk MR. Contemporaneous Inflammatory, Angiogenic, Fibrogenic, and Angiostatic Cytokine Profiles of the Time-to-Tumor Development by Cancer Cells to Orchestrate Tumor Neovascularization, Progression, and Metastasis. Cells 2024; 13:1739. [PMID: 39451257 PMCID: PMC11506673 DOI: 10.3390/cells13201739] [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: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytokines can promote various cancer processes, such as angiogenesis, epithelial to mesenchymal transition (EMT), invasion, and tumor progression, and maintain cancer stem-cell-like (CSCs) cells. The mechanism(s) that continuously promote(s) tumors to progress in the TME still need(s) to be investigated. The data in the present study analyzed the inflammatory, angiogenic, fibrogenic, and angiostatic cytokine profiles in the host serum during tumor development in a mouse model of human pancreatic cancer. Pancreatic MiaPaCa-2-eGFP cancer cells were subcutaneously implanted in RAG2xCγ double mutant mice. Blood samples were collected before cancer cell implantation and every week until the end point of the study. The extracted serum from the blood of each mouse at different time points during tumor development was analyzed using a Bio-Plex microarray analysis and a Bio-Plex 200 system for proinflammatory (IL-1β, IL-10, IFN-γ, and TNF-α) and angiogenic and fibrogenic (IL-15, IL-18, basic FGF, LIF, M-CSF, MIG, MIP-2, PDGF-BB, and VEGF) cytokines. Here, we find that during cancer cell colonization for tumor development, host angiogenic, fibrogenic, and proinflammatory cytokine profiling in the tumor-bearing mice has been shown to significantly reduce host angiostatic and proinflammatory cytokines that restrain tumor development and increase those for tumor growth. The proinflammatory cytokines IL-15, IL-18, and IL-1β profiles reveal a significant host serum increase after day 35 when the tumor began to progress in growth. In contrast, the angiostatic cytokine profiles of TNFα, MIG, M-CSF, IL-10, and IFNγ in the host serum revealed a dramatic and significant decrease after day 5 post-implantation of cancer cells. OP treatment of tumor-bearing mice on day 35 maintained high levels of angiostatic and fibrogenic cytokines. The data suggest an entirely new regulation by cancer cells for tumor development. The findings identify for the first time how pancreatic cancer cells use host cytokine profiling to orchestrate the initiation of tumor development.
Collapse
Affiliation(s)
- Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Haley L. Kombargi
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Abdulrahman M. Yaish
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| |
Collapse
|
39
|
Schermuly II, Romanet S, Patra AK, Mastrototaro L, Lemme A, Pieper R, Zentek J, Aschenbach JR. Transport of Neutral Amino Acids in the Jejunum of Pigs with Special Consideration of L-Methionine. Nutrients 2024; 16:3418. [PMID: 39408384 PMCID: PMC11478682 DOI: 10.3390/nu16193418] [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: 09/20/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Methionine (Met) is a popular nutritional supplement in humans and animals. It is routinely supplemented to pigs as L-Met, DL-Met, or DL-2-hydroxy-4-(methylthio) butanoic acid (DL-HMTBA). Methods: We investigated the effect of these Met supplements on jejunal amino acid (AA) transport in male castrated Piétrain × Danbred pigs, also including a non-supplemented group. The mucosal-to-serosal flux of ten [14C]-labeled AAs (L-glutamine, glycine, L-leucine, L-lysine, L-Met, L-serine, L-threonine, L-tryptophan, L-tyrosine and L-valine) was investigated at two concentrations (50 µM and 5 mM). Inhibition of apical uptake by mucosal L-Met was also measured for these AAs. The intestinal expression of apical AA transporters, angiotensin-converting enzyme II and inflammation-related genes were compared with those of a previous study. Results: Except for tryptophan and lysine at 5 mM, all AA fluxes were Na+-dependent (p ≤ 0.05), and the uptake of most AAs, except glycine and lysine, was inhibited by L-Met (p < 0.001). A correlation network existed between Na+-dependent fluxes of most AAs (except tryptophan and partly glycine). We observed the upregulation of B0AT1 (SLC6A19) (p < 0.001), the downregulation of ATB0,+ (SLC6A14) (p < 0.001) and a lower expression of CASP1, IL1β, IL8, TGFβ and TNFα in the present vs. the previous study (p < 0.001). Conclusions: The correlating AAs likely share the same Na+-dependent transporter(s). A varying effect of the Met supplement type on AA transport in the two studies might be related to a different level of supplementation or a different inflammatory status of the small intestine.
Collapse
Affiliation(s)
- Isabel I. Schermuly
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Stella Romanet
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| | - Amlan K. Patra
- American Institute for Goat Research, Langston University, Langston, OK 73050, USA;
| | - Lucia Mastrototaro
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Auf’m Hennekamp 65, 40225 Düsseldorf, Germany
| | - Andreas Lemme
- Animal Nutrition Services, Evonik Operations GmbH, Rodenbacher Chausee 4, 63457 Hanau-Wolfgang, Germany;
| | - Robert Pieper
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jürgen Zentek
- Institute of Animal Nutrition, Freie Universität Berlin, Königin-Luise-Straße 49, 14195 Berlin, Germany
| | - Jörg R. Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Königsweg 56, 14163 Berlin, Germany; (I.I.S.); (L.M.)
| |
Collapse
|
40
|
Tan D, Li E, Xiong S, Sun Y, Cheng W, Su Y, Lu Y. Transcriptomic and Metabolomic Analyses Reveal the Attenuating Role of Cordycepin and Cordyceps militaris Extract on Acute Liver Injury Induced by LPS in Piglets. Animals (Basel) 2024; 14:2873. [PMID: 39409822 PMCID: PMC11475243 DOI: 10.3390/ani14192873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Cordyceps militaris extract (CME) contains many bioactive compounds, mainly cordycepin (CPN). This study aimed to investigate the possible mechanisms underlying the amelioration of LPS-induced acute liver injury in piglets by CME or CPN supplementation using multi-omics analysis. Twenty-four weaned piglets were randomly distributed into 4 groups (n = 6): the control and LPS groups were fed basal diets; the CPN + LPS (CPN-LPS) and CME + LPS (CME-LPS) groups were fed the basal diets supplemented with CME or CPN. The results showed that CPN or CME supplementation significantly decreased the C-reactive protein level (p < 0.05) and improved liver tissue pathology to prevent acute liver injury after LPS treatment. Compared with LPS, the transcriptomic analysis indicated that CPN supplementation significantly downregulated cell adhesion molecules, while CME supplementation significantly downregulated inflammatory mediator regulation of TRP channels, complement and coagulation cascades and cytokine-cytokine receptor interaction. The metabolomic results showed that CPN or CME supplementation significantly reduced disease biomarker of bicyclo-prostaglandin E2, and increased levels of deoxyinosine and 3-hydroxyanthranilic acid (p < 0.05). The combined transcriptome and metabolome helped identify two metabolites PC 34:2 and PC 36:0, which may be associated with the restoration of liver cell morphology. In conclusion, CPN and CME could attenuate LPS-induced acute liver injury by regulating immune-related genes and metabolites. This study elucidates the potential protective mechanism of CPN or CME supplementation against acute liver injury.
Collapse
Affiliation(s)
- Ding Tan
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Endian Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Shijie Xiong
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Yue Sun
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Wenbo Cheng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (D.T.); (E.L.); (S.X.); (W.C.)
| | - Yang Lu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China
| |
Collapse
|
41
|
Zhang Y, Acosta FM, Jiang JX. Connexin 43 hemichannels and related diseases. Antib Ther 2024; 7:361-369. [PMID: 39678258 PMCID: PMC11646280 DOI: 10.1093/abt/tbae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/30/2024] [Accepted: 08/21/2024] [Indexed: 12/17/2024] Open
Abstract
Connexin 43 (Cx43) protein forms hemichannels (connexons) and gap junctions, with hemichannels consisting of six Cx43 molecules and gap junctions formed by two hemichannels. While gap junctions are prevalent in organs like the heart and liver, hemichannels are found in specific cell types, such as astrocytes and osteocytes. They allow the passage of small molecules (<1.5 kDa) between the cytoplasm and extracellular matrix. Cx43 hemichannels have emerged as potential therapeutic targets in various diseases, including central nervous system disorders, bone-related diseases, diabetic complications, wound healing, and cancers. Aberrant hemichannel opening can worsen conditions by releasing inflammatory elements, such as causing gliosis in neuronal cells. Conversely, functional hemichannels may inhibit cancer cell growth and metastasis. Recent studies are revealing new mechanisms of Cx43 hemichannels, broadening their therapeutic applications and highlighting the importance of regulating their activity for improved disease outcomes.
Collapse
Affiliation(s)
- Yanfeng Zhang
- AlaMab Therapeutics Inc, 302 Carnegie Center Dr Suite 100, Princeton, NJ 08540, United States
| | - Francisca M Acosta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, United States
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, United States
| |
Collapse
|
42
|
Zheng W, Marini W, Murakami K, Sotov V, Butler M, Gorrini C, Ohashi PS, Reedijk M. Caspase-1-dependent spatiality in triple-negative breast cancer and response to immunotherapy. Nat Commun 2024; 15:8514. [PMID: 39353903 PMCID: PMC11445480 DOI: 10.1038/s41467-024-52553-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024] Open
Abstract
Tumor immune microenvironment (TIME) spatial organization predicts outcome and therapy response in triple-negative breast cancer (TNBC). An immunosuppressive TIME containing elevated tumor-associated macrophages (TAM) and scarce CD8+ T cells is associated with poor outcome, but the regulatory mechanisms are poorly understood. Here we show that ETS1-driven caspase-1 expression, required for IL1β processing and TAM recruitment, is negatively regulated by estrogen receptors alpha (ERα) and a defining feature of TNBC. Elevated tumoral caspase-1 is associated with a distinct TIME characterized by increased pro-tumoral TAMs and CD8+ T cell exclusion from tumor nests. Mouse models prove the functional importance of ERα, ETS1, caspase-1 and IL1β in TIME conformation. Caspase-1 inhibition induces an immunoreactive TIME and reverses resistance to immune checkpoint blockade, identifying a therapeutically targetable mechanism that governs TNBC spatial organization.
Collapse
Affiliation(s)
- Weiyue Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wanda Marini
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Kiichi Murakami
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Valentin Sotov
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Marcus Butler
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of Medical Oncology, University of Toronto, Toronto, ON, Canada
| | - Chiara Gorrini
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Pamela S Ohashi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Michael Reedijk
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Department of Surgical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
43
|
Silva AD, Hwang J, Marciel MP, Bellis SL. The pro-inflammatory cytokines IL-1β and IL-6 promote upregulation of the ST6GAL1 sialyltransferase in pancreatic cancer cells. J Biol Chem 2024; 300:107752. [PMID: 39260693 PMCID: PMC11470512 DOI: 10.1016/j.jbc.2024.107752] [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: 03/11/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
The ST6GAL1 sialyltransferase is overexpressed in multiple cancers, including pancreatic ductal adenocarcinoma (PDAC). ST6GAL1 adds an α2-6-linked sialic acid to N-glycosylated membrane receptors, which consequently modulates receptor structure and function. While many studies have investigated the effects of ST6GAL1 on cell phenotype, there is a dearth of knowledge regarding mechanisms that regulate ST6GAL1 expression. In the current study, we evaluated the regulation of ST6GAL1 by two pro-inflammatory cytokines, IL-1β and IL-6, which are abundant within the PDAC tumor microenvironment. Cytokine activity was monitored using the Suit-2 PDAC cell line and two Suit-2-derived metastatic subclones, S2-013 and S2-LM7AA. For all three cell models, treatment with IL-1β or IL-6 increased the expression of ST6GAL1 protein and mRNA. Specifically, IL-1β and IL-6 induced expression of the ST6GAL1 YZ mRNA isoform, which is driven by the P3 promoter. The ST6GAL1 H and X isoforms were not detected. Promoter reporter assays confirmed that IL-1β and IL-6 activated transcription from the P3 promoter. We then examined downstream signaling mechanisms. IL-1β is known to signal through the NFκB transcription factor, whereas IL-6 signals through the STAT3 transcription factor. CUT&RUN experiments revealed that IL-1β promoted the binding of NFκB to the ST6GAL1 P3 promoter, and IL-6 induced the binding of STAT3 to the P3 promoter. Finally, we determined that inhibitors of NFκB and STAT3 blocked the upregulation of ST6GAL1 stimulated by IL-1β and IL-6, respectively. Together, these results highlight a novel molecular pathway by which cytokines within the tumor microenvironment stimulate the upregulation of ST6GAL1 in PDAC cells.
Collapse
Affiliation(s)
- Austin D Silva
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jihye Hwang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael P Marciel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Susan L Bellis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
44
|
Zhou LW, Li M, Li W. Regarding: IL-1β/DNA complex elevation distinguishes autoinflammatory disorders from autoimmune and infectious diseases. J Intern Med 2024; 296:377-378. [PMID: 38975626 DOI: 10.1111/joim.13818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Affiliation(s)
- Lian Wei Zhou
- Jiamusi University School of Clinical Medicine, Jiamusi, China
| | - Manling Li
- Jiaying College of Clinical Medicine, Meizhou, China
| | - Wenbo Li
- Jiamusi University School of Clinical Medicine, Jiamusi, China
| |
Collapse
|
45
|
Wang Y, Wang S, Li M, Zhang Q, Fang M, Zheng Q. Identification of molecular targets and underlying mechanisms of Fuzheng Shengbai Decoction against colon cancer based on network pharmacology. Am J Transl Res 2024; 16:4320-4342. [PMID: 39398618 PMCID: PMC11470291 DOI: 10.62347/vkmz3204] [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: 03/09/2024] [Accepted: 07/16/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVES To investigate the molecular targets and underlying mechanisms of Fuzheng Shengbai Decoction (FZSBD) against colon cancer (CC). METHODS Multiple network pharmacology approaches were used to predict the molecular targets and underlying mechanisms of FZSBD against CC. The expression of potential molecular targets was determined. The effects of FZSBD on cell viability, proliferation, migration, invasion, and the cell cycle of CC cells were investigated. The therapeutic efficacy, hematological, immunological, and inflammatory data in patients with CC were evaluated after treatment with the XELOX regimen with and without FZSBD. RESULTS A total of 912 potential targets in FZSBD and 2765 DEGs in CC specimens were screened. Five hub genes (TP53, MYC, VEGFA, CCND1, and IL1B) closely associated with immune-related signaling pathways and the cell cycle process were identified. The five hub genes were of prognostic value in CC. The gene and protein expression of the five hub genes was significantly higher in CC tumor tissue samples than that of normal tissue samples. Furthermore, with increasing doses, FZSBD increasingly inhibited growth, migration, and invasion, and suppressed the cell cycle process of CC cells. Supplementing of FZSBD to the XELOX regimen enhanced immune modulation and alleviated inflammatory responses. CONCLUSIONS This study identified the molecular targets and underlying mechanisms of FZSBD treatment against CC and may provide clues for future research on the treatment of CC with FZSBD.
Collapse
Affiliation(s)
- Yu Wang
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjing 210022, Jiangsu, China
| | - Shuiming Wang
- Department of Proctology, Nanjing Hospital of Traditional Chinese MedicineNanjing 210022, Jiangsu, China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Traditional Chinese MedicineNanjing 210022, Jiangsu, China
| | - Qijia Zhang
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjing 210003, Jiangsu, China
| | - Mingzhi Fang
- Department of Oncology, Nanjing Hospital of Traditional Chinese MedicineNanjing 210022, Jiangsu, China
| | - Qin Zheng
- Department of Oncology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese MedicineNanjing 210003, Jiangsu, China
| |
Collapse
|
46
|
Cai X, Yu X, Tang T, Xu Y, Wu T. JMJD2A promotes the development of castration-resistant prostate cancer by activating androgen receptor enhancer and inhibiting the cGAS-STING pathway. Mol Carcinog 2024; 63:1682-1696. [PMID: 38818897 DOI: 10.1002/mc.23753] [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: 02/28/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024]
Abstract
Exploring targets for inhibiting androgen receptor (AR) activity is an effective strategy for suppressing the development of castration-resistant prostate cancer (CRPC). Upregulation of histone demethylase JMJD2A activity is an important factor in increasing AR expression in CRPC. Based on our research, we found that the binding affinity between JMJD2A and AR increases in CRPC, while the level of AR histone methylation decreases and the H3K27ac level increases in the AR enhancer region. Further investigations revealed that overexpression of the histone demethylase JMJD2A increased the binding affinity between JMJD2A and AR, decreased AR histone methylation levels, upregulated H3K27ac in the AR enhancer region, and increased AR activity. Conversely, knocking down JMJD2A effectively reversed these effects. Additionally, in CRPC, JMJD2A expression was upregulated, the tumor-intrinsic immune cGAS-STING signaling pathway was suppressed, the tumor microenvironment was altered, and AR expression was upregulated. However, both knocking down JMJD2A and inhibiting the cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS-STING) signaling pathway reversed these effects. In summary, our study indicates that in CRPC, JMJD2A can directly bind to AR and activate residual AR enhancers through its demethylation activity, thereby promoting AR expression. Furthermore, upregulation of JMJD2A expression inhibits the innate immune cGAS-STING signaling pathway of the tumor, leading to a decrease in antitumor immune function, and further promoting AR expression.
Collapse
Affiliation(s)
- Xiang Cai
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaodong Yu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tielong Tang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yi Xu
- Department of Pharmacy, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Wu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
47
|
Ali NA, Elsayed GH, Mohamed SH, Abd Elkarim AS, Aly MS, Elgamal AM, Elsayed WM, El-Newary SA. Chia Seed ( Salvia hispanica) Attenuates Chemically Induced Lung Carcinomas in Rats through Suppression of Proliferation and Angiogenesis. Pharmaceuticals (Basel) 2024; 17:1129. [PMID: 39338293 PMCID: PMC11435337 DOI: 10.3390/ph17091129] [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: 07/11/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 09/30/2024] Open
Abstract
In 2022, 2.5 million cases of lung cancer were diagnosed, resulting in 1.8 million deaths. These statistics have motivated us to introduce a new natural product which is feasible in lung cancer therapies. This comprehensive study was performed to study the effects of chia seed extracts (70% ethanol and petroleum ether) on lung cancer in vitro and in vivo models. The invitro cytotoxicity activity of the chia extracts was studied in lung cancer cell lines (A549 cells). After 48 h, chia alcohol and ether extracts showed more inhibitory influence (IC50, 16.08, and 14.8 µg/mL, respectively) on A549 cells compared to Dox (IC50, 13.6 µg/mL). In vivo, administration of chia alcohol and ether extracts (500 mg/kg/day, orally for 20 weeks) recovered 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung cancer, as a significant reduction in the lung cancer biomarkers, including the relative weight of the lung (20.0 and 13.33%), ICAM(31.73 and 15.66%), and c-MYC (80 and 96%) and MMP9(60 and 69%) expression genes, and improvement in these changes were observed by histopathological examinations of the lung tissues compared to the lung control. Chia seeds fought lung cancer via suppression of proliferation, angiogenesis, inflammation, and activation apoptosis. These activities may be attributed to the chemical composition of chia, which is identified by LC-Mass, such as caffeic acid, vanillic acid, kaempferol-3-O-glucuronide, and taxifolin. Finally, we can conclude that chia seeds have an anti-lung cancer effect with a good safety margin.
Collapse
Affiliation(s)
- Naglaa A. Ali
- Hormones Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt; (N.A.A.); (G.H.E.); (S.H.M.)
| | - Ghada H. Elsayed
- Hormones Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt; (N.A.A.); (G.H.E.); (S.H.M.)
- Stem Cells Lab, Centre of Excellence for Advanced Sciences, National Research Centre, Dokki, Giza 12622, Egypt
| | - Safaa H. Mohamed
- Hormones Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt; (N.A.A.); (G.H.E.); (S.H.M.)
| | - Asmaa S. Abd Elkarim
- Chemistry of Tanning Materials and Leather Technology Department, National Research Centre, Giza 12622, Egypt;
| | - Mohamed S. Aly
- Department of Animal Reproduction and Artificial Insemination, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt;
| | - Abdelbaset M. Elgamal
- Department of Chemistry of Microbial and Natural Products, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt;
| | - Wael M. Elsayed
- Chemistry of Medicinal Plants Department, National Research Centre, Giza 12622, Egypt;
| | - Samah A. El-Newary
- Medicinal and Aromatic Plants Research Department, National Research Centre, El-Bouhoths St., Dokki, Giza 12622, Egypt
| |
Collapse
|
48
|
Shi R, Yu R, Lian F, Zheng Y, Feng S, Li C, Zheng X. Targeting HSP47 for cancer treatment. Anticancer Drugs 2024; 35:623-637. [PMID: 38718070 DOI: 10.1097/cad.0000000000001612] [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: 07/13/2024]
Abstract
Heat shock protein 47 (HSP47) serves as an endoplasmic reticulum residing collagen-specific chaperone and plays an important role in collagen biosynthesis and structural assembly. HSP47 is encoded by the SERPINH1 gene, which is located on chromosome 11q13.5, one of the most frequently amplified regions in human cancers. The expression of HSP47 is regulated by multiple cellular factors, including cytokines, transcription factors, microRNAs, and circular RNAs. HSP47 is frequently upregulated in a variety of cancers and plays an important role in tumor progression. HSP47 promotes tumor stemness, angiogenesis, growth, epithelial-mesenchymal transition, and metastatic capacity. HSP47 also regulates the efficacy of tumor therapies, such as chemotherapy, radiotherapy, and immunotherapy. Inhibition of HSP47 expression has antitumor effects, suggesting that targeting HSP47 is a feasible strategy for cancer treatment. In this review, we highlight the function and expression of regulatory mechanisms of HSP47 in cancer progression and point out the potential development of therapeutic strategies in targeting HSP47 in the future.
Collapse
Affiliation(s)
- Run Shi
- School of Medicine, Pingdingshan University, Pingdingshan, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Di L, Li M, Lei X, Xie W, Liu G, Wang Y, Zhang W, Zhu WG. Caspase-4 in glioma indicates deterioration and unfavorable prognosis by affecting tumor cell proliferation and immune cell recruitment. Sci Rep 2024; 14:17443. [PMID: 39075190 PMCID: PMC11286837 DOI: 10.1038/s41598-024-65018-z] [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: 12/13/2023] [Accepted: 06/16/2024] [Indexed: 07/31/2024] Open
Abstract
Gliomas are the most common malignant tumors of the central nervous system, accounting for approximately 80% of all malignant brain tumors. Accumulating evidence suggest that pyroptosis plays an essential role in the progression of cancer. Unfortunately, the effect of the pyroptosis-related factor caspase-4 (CASP4) on immunotherapy and drug therapy for tumors has not been comprehensively investigated. In this study, we systematically screened six hub genes by pooling differential pyroptosis-related genes in The Cancer Genome Atlas (TCGA) glioma data and the degree of centrality of index-related genes in the protein-protein interaction network. We performed functional and pathway enrichment analyses of the six hub genes to explore their biological functions and potential molecular mechanisms. We then investigated the importance of CASP4 using Kaplan-Meier survival analysis of glioma patients. TCGA and the Chinese Glioma Genome Atlas (CGGA) databases showed that reduced CASP4 expression leads to the potent clinical deterioration of glioma patients. Computational analysis of the effect of CASP4 on the infiltration level and recruitment of glioma immune cells revealed that CASP4 expression was closely associated with a series of tumor-suppressive immune checkpoint molecules, chemokines, and chemokine receptors. We also found that aberrant CASP4 expression correlated with chemotherapeutic drug sensitivity. Finally, analysis at the cellular and tissue levels indicated an increase in CASP4 expression in glioma, and that CASP4 inhibition significantly inhibited the proliferation of glioma cells. Thus, CASP4 is implicated as a new prognostic biomarker for gliomas with the potential to further guide immunotherapy and chemotherapy strategies for glioma patients.
Collapse
Affiliation(s)
- Longjiang Di
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Mengyan Li
- Guangdong Key Laboratory of Genomic Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518055, China
| | - Xianli Lei
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Faculty of Medicine, Peking University, Beijing, 100191, China
| | - Wenting Xie
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Guoqiang Liu
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Yongqing Wang
- Division of Rheumatology and Immunology, University of Toledo Medical Center, Toledo, OH, 43614, USA
| | - Wenjing Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.
| | - Wei-Guo Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Key Laboratory of Genomic Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
- College of Basic Medical Sciences, Wan Nan Medical College, Wuhu, 241006, China.
- International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
50
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|