1
|
Toba H, Jin D, Takai S. Suppressing SPARC gene with siRNA exerts therapeutic effects and inhibits MMP-2/9 and ADAMTS1 overexpression in a murine model of ischemia/reperfusion-induced acute kidney injury. J Pharmacol Sci 2025; 158:103-112. [PMID: 40288820 DOI: 10.1016/j.jphs.2025.03.010] [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: 01/14/2025] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC), a collagen-binding matricellular protein, is reported to facilitate inflammation and fibrosis in various tissues including the kidneys. Ischemia/reperfusion (I/R) is a major process of acute kidney injury. To investigate whether SPARC inhibition might attenuate renal I/R injury, we injected small interfering RNA (siRNA) targeting SPARC into male BALB/c mice one day before 45 min of renal ischemia followed by 72 h of reperfusion. Serum creatinine concentration, blood urea nitrogen, histological tubular damage, tubulointerstitial fibrosis, and expression of collagen I and transforming growth factor-β were increased after I/R. Expression of 4-hydroxy-2-nonenal, an oxidative stress marker, and the inflammatory cytokines monocyte chemoattractant protein-1 and tumor necrosis factor-α, were also upregulated in I/R kidneys. Overexpression of SPARC mRNA was observed after I/R, and immunohistochemistry revealed that SPARC was localized mainly in damaged tubuloepithelial cells. Additionally, a disintegrin and metalloproteinase with thrombospondin type 1 motif (ADAMTS1) expression colocalized with SPARC. Injection of siRNA targeting SPARC attenuated renal dysfunction, histological abnormalities, collagen deposition, oxidative stress, and renal inflammation. In addition, SPARC gene knockdown suppressed the I/R-induced increases in ADAMTS1 and matrix metalloproteinase-2/9 expression. In conclusion, I/R-induced SPARC could be a novel therapeutic target against acute kidney injury.
Collapse
Affiliation(s)
- Hiroe Toba
- Laboratory of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, 1 Misasagi Shichono-cho, Yamashina-ku, Kyoto, 607-8412, Japan; Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan.
| | - Denan Jin
- Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan
| | - Shinji Takai
- Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan
| |
Collapse
|
2
|
Jain P, Jain A, Deshmukh R, Samal P, Satapathy T, Ajazuddin. Metabolic dysfunction-associated steatotic liver disease (MASLD): Exploring systemic impacts and innovative therapies. Clin Res Hepatol Gastroenterol 2025; 49:102584. [PMID: 40157567 DOI: 10.1016/j.clinre.2025.102584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), which includes the inflammatory subtype metabolic dysfunction-associated steatohepatitis, is a prominent cause of chronic liver disease with systemic effects. Insulin resistance, obesity, and dyslipidaemia produce MASLD in over 30 % of adults. It is a global health issue. From MASLD to MASH, hepatic inflammation and fibrosis grow, leading to cirrhosis, hepatocellular cancer, and extrahepatic complications such CVD, CKD, and sarcopenia. Effects of MASLD to MASH are mediated through mechanisms that include inflammation, oxidative stress, dysbiosis, and predisposition through genetic makeup. Advances in diagnostic nomenclature in the past few years have moved the emphasis away from NAFLD to MASLD, focusing on the metabolic etiology and away from the stigma of an alcoholic-related condition. Epidemiological data show a large geographical variability and increasing prevalence in younger populations, particularly in regions with high carbohydrate-rich diets and central adiposity. Lifestyle modification is considered as the main management of MASLD currently. This may include dietary intervention, exercise, and weight loss management. Pharmaceutical management is primarily aimed at metabolic dysfunction with promising findings for GLP-1 receptor agonists, pioglitazone and SGLT-2 inhibitors, which can correct both hepatic and systemic outcome. However, it still depends on well-integrated multidisciplinary care models by considering complex relationships between MASLD and its effects on extrahepatic organs. Determining complications at an early stage; developing precision medicine strategies; exploring new therapeutic targets will represent crucial factors in improving their outcomes. This review discuss the systemic nature of MASLD and calls for multiple collaborations to reduce its far-reaching health impacts and our quest for understanding its pathological mechanisms. Thus, collective efforts that are required to address MASLD are under the public health, clinical care, and research angles toward effectively containing its rapidly increasing burden.
Collapse
Affiliation(s)
- Parag Jain
- Department of Pharmacology, Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India, 490024.
| | - Akanksha Jain
- Department of Biotechnology, Bharti University, Durg, C.G., India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, India, 281406
| | - Pradeep Samal
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Bilaspur, C.G., India
| | - Trilochan Satapathy
- Department of Pharmacy, Columbia Institute of Pharmaceutical Sciences, Raipur, C.G., India, 493111
| | - Ajazuddin
- Department of Pharmacology, Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India, 490024
| |
Collapse
|
3
|
Fabregat I, Cubero FJ. ECM1: a novel matricellular protein with promising antifibrotic potential. Gut 2025; 74:339-340. [PMID: 39448253 DOI: 10.1136/gutjnl-2024-333455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 10/26/2024]
Affiliation(s)
- Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de L'Hospitalet, Barcelona, Spain
- Centre for Biomedical Research, Network on Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
| | - Francisco Javier Cubero
- Centre for Biomedical Research, Network on Liver and Digestive Diseases (CIBEREHD), Madrid, Spain
- Immunology, Ophthalmology and ENT, Complutense University of Madrid Faculty of Medicine, Madrid, Spain
- Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
4
|
Link F, Li Y, Zhao J, Munker S, Fan W, Nwosu ZC, Yao Y, Wang S, Huang C, Liebe R, Hammad S, Liu H, Shao C, Gao C, Sun B, Török NJ, Ding H, Ebert MP, Weng H, Ten Dijke P, Drasdo D, Dooley S, Wang S. ECM1 attenuates hepatic fibrosis by interfering with mediators of latent TGF-β1 activation. Gut 2025; 74:424-439. [PMID: 39448254 DOI: 10.1136/gutjnl-2024-333213] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 07/24/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE Extracellular matrix protein 1 (ECM1) serves as a gatekeeper of hepatic fibrosis by maintaining transforming growth factor-β1 (TGF-β1) in its latent form. ECM1 knockout (KO) causes latent (L) TGF-β1 activation, resulting in hepatic fibrosis with rapid mortality. In chronic liver disease (CLD), ECM1 decreases with increasing CLD severity. We investigate the regulatory role of ECM1 in TGF-β1 bioavailability and its impact on CLD progression. DESIGN RNAseq was performed to analyse hepatic gene expression. Functional assays were performed using hepatic stellate cells (HSCs), Ecm1-KO and Fxr-KO mice, patient liver tissue and computer simulations. RESULTS Expression of LTGF-β1 activators, including thrombospondins (TSPs), ADAMTS proteases and matrix metalloproteinases (MMPs), increased along with profibrotic gene expression in liver tissue of Ecm1-KO mice. In HSCs, overexpression of ECM1 prevented LTGF-β1 activation mediated by TSP-1, ADAMTS1, and MMP-2/9. In vitro interaction assays demonstrated that ECM1 inhibited LTGF-β1 activation by interacting with TSP-1 and ADAMTS1 via their respective, intrinsic KRFK or KTFR amino acid sequences and by suppressing MMP-2/9 proteolytic activity. In mice, ECM1 overexpression attenuated KRFK-induced LTGF-β1 activation while KTFR treatment reversed Ecm1-KO-mediated and Fxr-KO-mediated liver injury. In patients with CLD, ECM1 expression was inversely correlated with TSP-1, ADAMTS1, MMP-2/9 expression and LTGF-β1 activation. And, these results were complemented by a computational compartment model representing the key network of cellular phenotypes and predicted interactions in liver fibrogenesis. CONCLUSION Our findings underscore the hepatoprotective effect of ECM1, which interferes with mediators of LTGF-β1 activation, suggesting ECM1 or its representative peptide as potential antifibrotic therapies in CLD.
Collapse
Affiliation(s)
- Frederik Link
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yujia Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jieling Zhao
- INRIA de Saclay, Palaiseau, France
- IfADo, Dortmund, Germany
| | - Stefan Munker
- Department of Medicine II, University Hospital, LMU, Munich, Germany
- Liver Center Munich, University Hospital, LMU, Munich, Germany
| | - Weiguo Fan
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zeribe C Nwosu
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Ye Yao
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shanshan Wang
- Beijing Institute of Hepatology, Beijing You'an Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Chenjun Huang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Roman Liebe
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Seddik Hammad
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hui Liu
- Department of Pathology, Beijing You'an Hospital, Affiliated with Capital Medical University, Beijing, People's Republic of China
| | - Chen Shao
- Department of Pathology, Beijing You'an Hospital, Affiliated with Capital Medical University, Beijing, People's Republic of China
| | - Chunfang Gao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Bing Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Natalie J Török
- Gastroenterology and Hepatology, Stanford University, Palo Alto, CA, USA
| | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing You'an Hospital, Affiliated with Capital Medical University, Beijing, People's Republic of China
| | - Matthias Pa Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center, Mannheim, Germany
| | - Honglei Weng
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Drasdo
- INRIA de Saclay, Palaiseau, France
- IfADo, Dortmund, Germany
| | - Steven Dooley
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sai Wang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
5
|
Aubert A, Goeres J, Liu A, Kao M, Richardson KC, Jung K, Hinz B, Crawford RI, Granville DJ. Potential implications of granzyme B in keloids and hypertrophic scars through extracellular matrix remodeling and latent TGF-β activation. Front Immunol 2025; 15:1484462. [PMID: 39885984 PMCID: PMC11779620 DOI: 10.3389/fimmu.2024.1484462] [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: 08/21/2024] [Accepted: 12/20/2024] [Indexed: 02/01/2025] Open
Abstract
Keloid scars (KS) and hypertrophic scars (HS) are fibroproliferative wound healing defects characterized by excessive accumulation of extracellular matrix (ECM) in the dermis of affected individuals. Although transforming growth factor (TGF)-β is known to be involved in the formation of KS and HS, the molecular mechanisms responsible for its activation remain unclear. In this study we investigated Granzyme B (GzmB), a serine protease with established roles in fibrosis and scarring through the cleavage of ECM proteins, as a potential new mediator of TGF-β activation in KS and HS. Increased GzmB-positive mast cells were identified in the dermis of KS and HS but not healthy skin controls. Elevated levels of substance P, a neuropeptide involved in mast cell degranulation, suggest that GzmB is released extracellularly, as confirmed by the significant reduction of the established extracellular GzmB substrate decorin in KS and HS. Similarly, presence of latent TGF-β binding protein 1 (LTBP1), a protein involved in the extracellular tethering of latent TGF-β, was disrupted proximal to the dermal-epidermal junction (DEJ) of GzmBhigh KS and HS lesions. Using LTBP1-enriched medium as well as purified LTBP1, its cleavage by GzmB was confirmed in vitro. Increased TGF-β/Smad signaling pathway was observed in keratinocytes treated with GzmB-digested LTBP1 and was abolished by the addition of a pan-TGF-β inhibitor, suggesting that GzmB cleavage of LTBP1 contributes to TGF-β activation. In dermal fibroblasts, GzmB also cleaved cell-derived LTBP1 and induced TGF-β activation through the cleavage of one or more unidentified fibroblast-secreted proteins. Altogether, the present results suggest that GzmB contributes to KS and HS through ECM remodeling and TGF-β activation.
Collapse
Affiliation(s)
- Alexandre Aubert
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
| | - Jenna Goeres
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
| | - Amy Liu
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
| | - Martin Kao
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
| | - Katlyn C. Richardson
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Keenan Research Institute for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Richard I. Crawford
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Dermatology and Skin Science, University of British Columbia (UBC), Vancouver, BC, Canada
| | - David J. Granville
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- British Columbia Professional Firefighters’ Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
| |
Collapse
|
6
|
Meng Q, Bao D, Liu S, Huang J, Guo M, Dai B, Ding L, Xie S, Meng M, Lv C, He W, Luo H, Zhu H. ADAM Metallopeptidase domain 19 promotes skin fibrosis in systemic sclerosis via neuregulin-1. Mol Med 2024; 30:269. [PMID: 39716051 DOI: 10.1186/s10020-024-01047-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND ADAM19 (ADAM Metallopeptidase Domain 19) is known to be involved in extracellular matrix (ECM) remodeling, yet its specific function in systemic sclerosis (SSc) fibrosis remains unclear. OBJECTIVES This study sought to clarify the role and underlying mechanism of ADAM19 in SSc skin fibrosis. METHODS The expression of ADAM19 was assessed in skin tissues of SSc and wound healing using publicly available transcriptome datasets. This analysis was further validated through real-time PCR, western blot, and immunostaining in our SSc cohort, as well as in a mouse model of hypochlorite (HOCl)-induced fibrosis. To downregulate the expression of ADAM19, ADAM19 siRNA was employed. The influence of ADAM19 on fibroblast transcriptomics was examined using bulk RNA-seq. Data analysis and visualization were conducted using R packages, including edgeR, limma, clusterProfiler, ggplot2, gseaplot2, and complexheatmap. RESULTS ADAM19 exhibited a significant upregulation in skin tissues of SSc patients, as well as in wound healing and a HOCl-induced fibrosis mouse model. Additionally, there was a notable positive correlation between ADAM19 and fibrosis-related genes, local skin score, Modified Rodnan skin score, skin thickness progression rate, and the presence of ARA antibodies in SSc patients. Furthermore, ADAM19 levels were markedly elevated in SSc primary dermal fibroblasts and TGF-β-stimulated healthy controls primary dermal fibroblasts. The downregulation of ADAM19 resulted in the repression of TGF-β-induced ECM deposition and fibroblast activation. ADAM19 was identified as a mediator for the shedding of neuregulin-1 (NRG1) in fibroblasts, a pro-fibrotic cytokine that must be cleaved to exert its function. CONCLUSION ADAM19 plays a role in TGF-β-induced ECM deposition and fibroblast activation by mediating the shedding of NRG1, ultimately contributing to the development of skin fibrosis in SSc.
Collapse
Affiliation(s)
- Qiming Meng
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Ding Bao
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Sijia Liu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jing Huang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Muyao Guo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Bingying Dai
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Liqing Ding
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Shasha Xie
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Meng Meng
- Department of Pathology, Xiangya Hospital, Changsha, 410008, P.R. China
| | - Chunliu Lv
- Department of Breast Tumor Plastic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, Changsha, Hunan, 410013, P.R. China
| | - Weijia He
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| | - Honglin Zhu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| |
Collapse
|
7
|
Toba H, Takai S. Exploring the roles of SPARC as a proinflammatory factor and its potential as a novel therapeutic target against cardiovascular disease. Am J Physiol Heart Circ Physiol 2024; 327:H1174-H1186. [PMID: 39269452 DOI: 10.1152/ajpheart.00565.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Cardiovascular disease (CVD) is a leading cause of death worldwide, and the number of patients with CVD continues to increase despite extensive research and developments in this field. Chronic inflammation is a pivotal pathological component of CVD, and unveiling new proinflammatory factors will help devise novel preventive and therapeutic strategies. The extracellular matrix (ECM) not only provides structural support between cells but also contributes to cellular functions. Secreted protein acidic and rich in cysteine (SPARC) is a collagen-binding matricellular protein that is particularly induced during development and tissue remodeling. A proinflammatory role for SPARC has been demonstrated in various animal models, such as in the lipopolysaccharide-induced footpad model and dextran sodium sulfate-induced colitis model. Recent clinical studies reported a positive correlation between elevated plasma SPARC levels and hypertension, obesity, and the inflammatory marker high-sensitive C-reactive protein. In addition, SPARC gene deletion attenuates the cardiac injury induced by aging, myocardial infarction, and pressure load, suggesting that SPARC has deleterious effects on CVD. This review summarizes the regulatory and proinflammatory mechanisms of SPARC on CVD, chronic kidney disease (CKD), and cerebrovascular disease and discusses the rationale behind measuring SPARC as a biomarker of CVD and the effects of inhibition of SPARC in the prevention and treatment of CVD.
Collapse
Affiliation(s)
- Hiroe Toba
- Division of Pathological Sciences, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
- Department of Pharmacology, Educational Foundation of Osaka Medical and Pharmacological University, Takatsuki, Japan
| | - Shinji Takai
- Department of Pharmacology, Educational Foundation of Osaka Medical and Pharmacological University, Takatsuki, Japan
| |
Collapse
|
8
|
Zhao J, Tian T, Huang J, Zha H, Shi L, Yao Y. A novel role of ADAMTS16 in renal fibrosis through activating TGF-β/Smad signaling. Cell Signal 2024; 122:111347. [PMID: 39147297 DOI: 10.1016/j.cellsig.2024.111347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Chronic Kidney Disease (CKD) has emerged as a global public health concern, with its primary pathological basis being Renal Fibrosis (RF), crucial to halt its progression to End-Stage Renal Disease (ESRD). However, effective treatment options are currently lacking. Therefore, exploring the mechanisms of RF, identifying drug targets and diagnostic biomarkers are important. In this study, we identified ADAMTS16 as a newly expressed regulatory factor highly expressed in renal fibrosis tissue. ADAMTS16 interacts with latency-associated peptide (LAP)-transforming growth factor (TGF)-β, leading to the activation of TGF-β. Loss of ADAMTS16 expression effectively reduces TGF-β-dependent transcription activity. Furthermore, the use of RRFR tetrapeptide derived from ADAMTS16 can activate the TGF-β/Smad signaling axis, promoting RF. In summary, ADAMTS16 is induced in the progression of CKD, interacting with LAP-TGF-β and potentially activating SMAD2/3. Therefore, targeting ADAMTS16 may serve as a crucial new strategy to alleviate RF and treat CKD patients.
Collapse
Affiliation(s)
- Juan Zhao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China; Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tongtong Tian
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China
| | - Jing Huang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hongchu Zha
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Yichang, Hubei 443000, China
| | - Lang Shi
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China.; Department of Nephrology, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, China.
| |
Collapse
|
9
|
Bouguéon M, Legagneux V, Hazard O, Bomo J, Siegel A, Feret J, Théret N. A rule-based multiscale model of hepatic stellate cell plasticity: Critical role of the inactivation loop in fibrosis progression. PLoS Comput Biol 2024; 20:e1011858. [PMID: 39074160 PMCID: PMC11309422 DOI: 10.1371/journal.pcbi.1011858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 08/08/2024] [Accepted: 07/05/2024] [Indexed: 07/31/2024] Open
Abstract
Hepatic stellate cells (HSC) are the source of extracellular matrix (ECM) whose overproduction leads to fibrosis, a condition that impairs liver functions in chronic liver diseases. Understanding the dynamics of HSCs will provide insights needed to develop new therapeutic approaches. Few models of hepatic fibrosis have been proposed, and none of them include the heterogeneity of HSC phenotypes recently highlighted by single-cell RNA sequencing analyses. Here, we developed rule-based models to study HSC dynamics during fibrosis progression and reversion. We used the Kappa graph rewriting language, for which we used tokens and counters to overcome temporal explosion. HSCs are modeled as agents that present seven physiological cellular states and that interact with (TGFβ1) molecules which regulate HSC activation and the secretion of type I collagen, the main component of the ECM. Simulation studies revealed the critical role of the HSC inactivation process during fibrosis progression and reversion. While inactivation allows elimination of activated HSCs during reversion steps, reactivation loops of inactivated HSCs (iHSCs) are required to sustain fibrosis. Furthermore, we demonstrated the model's sensitivity to (TGFβ1) parameters, suggesting its adaptability to a variety of pathophysiological conditions for which levels of (TGFβ1) production associated with the inflammatory response differ. Using new experimental data from a mouse model of CCl4-induced liver fibrosis, we validated the predicted ECM dynamics. Our model also predicts the accumulation of iHSCs during chronic liver disease. By analyzing RNA sequencing data from patients with non-alcoholic steatohepatitis (NASH) associated with liver fibrosis, we confirmed this accumulation, identifying iHSCs as novel markers of fibrosis progression. Overall, our study provides the first model of HSC dynamics in chronic liver disease that can be used to explore the regulatory role of iHSCs in liver homeostasis. Moreover, our model can also be generalized to fibroblasts during repair and fibrosis in other tissues.
Collapse
Affiliation(s)
- Matthieu Bouguéon
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France
| | | | - Octave Hazard
- École Polytechnique, Palaiseau, France
- DI-ENS (Inria, ÉNS, CNRS, PSL University), École normale supérieure, Paris, France
| | - Jérémy Bomo
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France
| | - Anne Siegel
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
| | - Jérôme Feret
- DI-ENS (Inria, ÉNS, CNRS, PSL University), École normale supérieure, Paris, France
- Team Antique, Inria, Paris, France
| | - Nathalie Théret
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France
| |
Collapse
|
10
|
Sommerauer C, Gallardo-Dodd CJ, Savva C, Hases L, Birgersson M, Indukuri R, Shen JX, Carravilla P, Geng K, Nørskov Søndergaard J, Ferrer-Aumatell C, Mercier G, Sezgin E, Korach-André M, Petersson C, Hagström H, Lauschke VM, Archer A, Williams C, Kutter C. Estrogen receptor activation remodels TEAD1 gene expression to alleviate hepatic steatosis. Mol Syst Biol 2024; 20:374-402. [PMID: 38459198 PMCID: PMC10987545 DOI: 10.1038/s44320-024-00024-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024] Open
Abstract
Sex-based differences in obesity-related hepatic malignancies suggest the protective roles of estrogen. Using a preclinical model, we dissected estrogen receptor (ER) isoform-driven molecular responses in high-fat diet (HFD)-induced liver diseases of male and female mice treated with or without an estrogen agonist by integrating liver multi-omics data. We found that selective ER activation recovers HFD-induced molecular and physiological liver phenotypes. HFD and systemic ER activation altered core liver pathways, beyond lipid metabolism, that are consistent between mice and primates. By including patient cohort data, we uncovered that ER-regulated enhancers govern central regulatory and metabolic genes with clinical significance in metabolic dysfunction-associated steatotic liver disease (MASLD) patients, including the transcription factor TEAD1. TEAD1 expression increased in MASLD patients, and its downregulation by short interfering RNA reduced intracellular lipid content. Subsequent TEAD small molecule inhibition improved steatosis in primary human hepatocyte spheroids by suppressing lipogenic pathways. Thus, TEAD1 emerged as a new therapeutic candidate whose inhibition ameliorates hepatic steatosis.
Collapse
Affiliation(s)
- Christian Sommerauer
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Carlos J Gallardo-Dodd
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Christina Savva
- Department of Medicine, Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden
| | - Linnea Hases
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Madeleine Birgersson
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Rajitha Indukuri
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Pablo Carravilla
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
- Department of Women's and Children's Health, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Keyi Geng
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Jonas Nørskov Søndergaard
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Clàudia Ferrer-Aumatell
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Grégoire Mercier
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
| | - Erdinc Sezgin
- Department of Women's and Children's Health, Karolinska Institute, Science for Life Laboratory, Solna, Sweden
| | - Marion Korach-André
- Department of Medicine, Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden
| | - Carl Petersson
- Department of Drug Metabolism and Pharmacokinetics, The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | - Hannes Hagström
- Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, Sweden
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Amena Archer
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Cecilia Williams
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Science for Life Laboratory, Solna, Sweden.
| |
Collapse
|
11
|
Xiao Y, Vazquez-Padron RI, Martinez L, Singer HA, Woltmann D, Salman LH. Role of platelet factor 4 in arteriovenous fistula maturation failure: What do we know so far? J Vasc Access 2024; 25:390-406. [PMID: 35751379 PMCID: PMC9974241 DOI: 10.1177/11297298221085458] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The rate of arteriovenous fistula (AVF) maturation failure remains unacceptably high despite continuous efforts on technique improvement and careful pre-surgery planning. In fact, half of all newly created AVFs are unable to be used for hemodialysis (HD) without a salvage procedure. While vascular stenosis in the venous limb of the access is the culprit, the underlying factors leading to vascular narrowing and AVF maturation failure are yet to be determined. We have recently demonstrated that AVF non-maturation is associated with post-operative medial fibrosis and fibrotic stenosis, and post-operative intimal hyperplasia (IH) exacerbates the situation. Multiple pathological processes and signaling pathways are underlying the stenotic remodeling of the AVF. Our group has recently indicated that a pro-inflammatory cytokine platelet factor 4 (PF4/CXCL4) is upregulated in veins that fail to mature after AVF creation. Platelet factor 4 is a fibrosis marker and can be detected in vascular stenosis tissue, suggesting that it may contribute to AVF maturation failure through stimulation of fibrosis and development of fibrotic stenosis. Here, we present an overview of the how PF4-mediated fibrosis determines AVF maturation failure.
Collapse
Affiliation(s)
- Yuxuan Xiao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Roberto I Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Laisel Martinez
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Daniel Woltmann
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Loay H Salman
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY, USA
| |
Collapse
|
12
|
Chien MH, Yang YC, Ho KH, Ding YF, Chen LH, Chiu WK, Chen JQ, Tung MC, Hsiao M, Lee WJ. Cyclic increase in the ADAMTS1-L1CAM-EGFR axis promotes the EMT and cervical lymph node metastasis of oral squamous cell carcinoma. Cell Death Dis 2024; 15:82. [PMID: 38263290 PMCID: PMC10805752 DOI: 10.1038/s41419-024-06452-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/25/2024]
Abstract
The matrix metalloprotease A disintegrin and metalloprotease with thrombospondin motifs 1 (ADAMTS1) was reported to be involved in tumor progression in several cancer types, but its contributions appear discrepant. At present, the role of ADAMTS1 in oral squamous cell carcinoma (SCC; OSCC) remains unclear. Herein, The Cancer Genome Atlas (TCGA) database showed that ADAMTS1 transcripts were downregulated in head and neck SCC (HNSCC) tissues compared to normal tissues, but ADAMTS1 levels were correlated with poorer prognoses of HNSCC patients. In vitro, we observed that ADAMTS1 expression levels were correlated with the invasive abilities of four OSCC cell lines, HSC-3, SCC9, HSC-3M, and SAS. Knockdown of ADAMTS1 in OSCC cells led to a decrease and its overexpression led to an increase in cell-invasive abilities in vitro as well as tumor growth and lymph node (LN) metastasis in OSCC xenografts. Mechanistic investigations showed that the cyclic increase in ADAMTS1-L1 cell adhesion molecule (L1CAM) axis-mediated epidermal growth factor receptor (EGFR) activation led to exacerbation of the invasive abilities of OSCC cells via inducing epithelial-mesenchymal transition (EMT) progression. Clinical analyses revealed that ADAMTS1, L1CAM, and EGFR levels were all correlated with worse prognoses of HNSCC patients, and patients with ADAMTS1high/L1CAMhigh or EGFRhigh tumors had the shortest overall and disease-specific survival times. As to therapeutic aspects, we discovered that an edible plant-derived flavonoid, apigenin (API), drastically inhibited expression of the ADAMTS1-L1CAM-EGFR axis and reduced the ADAMTS1-triggered invasion and LN metastasis of OSCC cells in vitro and in vivo. Most importantly, API treatment significantly prolonged survival rates of xenograft mice with OSCC. In summary, ADAMTS1 may be a useful biomarker for predicting OSCC progression, and API potentially retarded OSCC progression by targeting the ADAMTS1-L1CAM-EGFR signaling pathway.
Collapse
Affiliation(s)
- Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital Taipei, Taipei, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Kuo-Hao Ho
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Fang Ding
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Otolaryngology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Li-Hsin Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Kuan Chiu
- Division of Plastic Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Surgery, Taipei Medical University, Taipei, Taiwan
| | - Ji-Qing Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Cancer Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Min-Che Tung
- Department of Surgery, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
13
|
Chen D, Mao P, Sun C, Fan X, Zhu Q, Chen Z, He Z, Lou Y, Sun H. Prognostic Value of Combined Neutrophil-to-Lymphocyte Ratio and Imaging Tumor Capsule in Solitary Hepatocellular Carcinoma Patients after Narrow-Margin Hepatectomy. J Clin Med 2024; 13:351. [PMID: 38256485 PMCID: PMC10816149 DOI: 10.3390/jcm13020351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND The study aimed to investigate the clinical value and prognostic patterns of the neutrophil-to-lymphocyte ratio (NLR) and imaging tumor capsule (ITC) in solitary hepatocellular carcinoma (HCC) patients undergoing narrow-margin hepatectomy. METHODS Data for solitary HCC patients treated with narrow-margin surgery were extracted from Shanghai General Hospital. Clinical features of recurrence-free survival (RFS), overall survival (OS), and early recurrence were investigated by Cox/logistic regression. The significant variables were subsequently incorporated into the nomogram pattern. Survival analysis stratified by NLR and ITC was also performed. RESULTS The study included a cohort of 222 patients, with median RFS and OS of 24.083 and 32.283 months, respectively. Both an NLR ≥ 2.80 and incomplete ITC had a significant impact on prognosis. NLR and ITC independently affected RFS and OS, whereas alpha-fetoprotein (AFP) and ITC were identified as independent factors for early relapse. The RFS and OS nomogram, generated based on the Cox model, demonstrated good performance in validation. The combination of NLR and ITC showed greater predictive accuracy for 5-year RFS and OS. Subgroups with an NLR ≥ 2.80 and incomplete ITC had the worst prognosis. CONCLUSIONS Both NLR and ITC significantly affected RFS, OS, and early recurrence among solitary HCC patients who underwent narrow-margin hepatectomy. The combination of NLR and ITC has the potential to guide rational clinical treatment and determine the prognosis.
Collapse
Affiliation(s)
- Desheng Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (D.C.); (Q.Z.); (Z.C.); (Z.H.); (Y.L.)
| | - Pengjuan Mao
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China;
| | - Chen Sun
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China;
| | - Xuhui Fan
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China;
| | - Qi Zhu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (D.C.); (Q.Z.); (Z.C.); (Z.H.); (Y.L.)
| | - Zeping Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (D.C.); (Q.Z.); (Z.C.); (Z.H.); (Y.L.)
| | - Zeping He
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (D.C.); (Q.Z.); (Z.C.); (Z.H.); (Y.L.)
| | - Yichao Lou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (D.C.); (Q.Z.); (Z.C.); (Z.H.); (Y.L.)
| | - Hongcheng Sun
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (D.C.); (Q.Z.); (Z.C.); (Z.H.); (Y.L.)
| |
Collapse
|
14
|
Frangogiannis NG. TGF-β as a therapeutic target in the infarcted and failing heart: cellular mechanisms, challenges, and opportunities. Expert Opin Ther Targets 2024; 28:45-56. [PMID: 38329809 DOI: 10.1080/14728222.2024.2316735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/06/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Myocardial fibrosis accompanies most cardiac conditions and can be reparative or maladaptive. Transforming Growth Factor (TGF)-β is a potent fibrogenic mediator, involved in repair, remodeling, and fibrosis of the injured heart. AREAS COVERED This review manuscript discusses the role of TGF-β in heart failure focusing on cellular mechanisms and therapeutic implications. TGF-β is activated in infarcted, remodeling and failing hearts. In addition to its fibrogenic actions, TGF-β has a broad range of effects on cardiomyocytes, immune, and vascular cells that may have both protective and detrimental consequences. TGF-β-mediated effects on macrophages promote anti-inflammatory transition, whereas actions on fibroblasts mediate reparative scar formation and effects on pericytes are involved in maturation of infarct neovessels. On the other hand, TGF-β actions on cardiomyocytes promote adverse remodeling, and prolonged activation of TGF-β signaling in fibroblasts stimulates progression of fibrosis and heart failure. EXPERT OPINION Understanding of the cell-specific actions of TGF-β is necessary to design therapeutic strategies in patients with myocardial disease. Moreover, to implement therapeutic interventions in the heterogeneous population of heart failure patients, mechanism-driven classification of both HFrEF and HFpEF patients is needed. Heart failure patients with prolonged or overactive fibrogenic TGF-β responses may benefit from cautious TGF-β inhibition.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
15
|
Dekky B, Azar F, Bonnier D, Monseur C, Kalebić C, Arpigny E, Colige A, Legagneux V, Théret N. ADAMTS12 is a stromal modulator in chronic liver disease. FASEB J 2023; 37:e23237. [PMID: 37819632 DOI: 10.1096/fj.202200692rrrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023]
Abstract
Adamalysins, a family of metalloproteinases containing a disintegrin and metalloproteinases (ADAMs) and ADAM with thrombospondin motifs (ADAMTSs), belong to the matrisome and play important roles in various biological and pathological processes, such as development, immunity and cancer. Using a liver cancer dataset from the International Cancer Genome Consortium, we developed an extensive in silico screening that identified a cluster of adamalysins co-expressed in livers from patients with hepatocellular carcinoma (HCC). Within this cluster, ADAMTS12 expression was highly associated with recurrence risk and poorly differentiated HCC signatures. We showed that ADAMTS12 was expressed in the stromal cells of the tumor and adjacent fibrotic tissues of HCC patients, and more specifically in activated stellate cells. Using a mouse model of carbon tetrachloride-induced liver injury, we showed that Adamts12 was strongly and transiently expressed after a 24 h acute treatment, and that fibrosis was exacerbated in Adamts12-null mice submitted to carbon tetrachloride-induced chronic liver injury. Using the HSC-derived LX-2 cell line, we showed that silencing of ADAMTS12 resulted in profound changes of the gene expression program. In particular, genes previously reported to be induced upon HSC activation, such as PAI-1, were mostly down-regulated following ADAMTS12 knock-down. The phenotype of these cells was changed to a less differentiated state, showing an altered actin network and decreased nuclear spreading. These phenotypic changes, together with the down-regulation of PAI-1, were offset by TGF-β treatment. The present study thus identifies ADAMTS12 as a modulator of HSC differentiation, and a new player in chronic liver disease.
Collapse
Affiliation(s)
- Bassil Dekky
- University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Fida Azar
- University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Dominique Bonnier
- University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Christine Monseur
- Laboratory of Connective Tissues Biology, GIGA-R, University of Liege, Liege, Belgium
| | - Chiara Kalebić
- University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Esther Arpigny
- Laboratory of Connective Tissues Biology, GIGA-R, University of Liege, Liege, Belgium
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-R, University of Liege, Liege, Belgium
| | - Vincent Legagneux
- University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| | - Nathalie Théret
- University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
| |
Collapse
|
16
|
Hanson I, Pitman KE, Edin NFJ. The Role of TGF-β3 in Radiation Response. Int J Mol Sci 2023; 24:ijms24087614. [PMID: 37108775 PMCID: PMC10141893 DOI: 10.3390/ijms24087614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Transforming growth factor-beta 3 (TGF-β3) is a ubiquitously expressed multifunctional cytokine involved in a range of physiological and pathological conditions, including embryogenesis, cell cycle regulation, immunoregulation, and fibrogenesis. The cytotoxic effects of ionizing radiation are employed in cancer radiotherapy, but its actions also influence cellular signaling pathways, including that of TGF-β3. Furthermore, the cell cycle regulating and anti-fibrotic effects of TGF-β3 have identified it as a potential mitigator of radiation- and chemotherapy-induced toxicity in healthy tissue. This review discusses the radiobiology of TGF-β3, its induction in tissue by ionizing radiation, and its potential radioprotective and anti-fibrotic effects.
Collapse
Affiliation(s)
- Ingunn Hanson
- Department of Physics, University of Oslo, 0371 Oslo, Norway
| | | | - Nina F J Edin
- Department of Physics, University of Oslo, 0371 Oslo, Norway
| |
Collapse
|
17
|
Hu X, Jiang C, Hu N, Hong S. ADAMTS1 induces epithelial-mesenchymal transition pathway in non-small cell lung cancer by regulating TGF-β. Aging (Albany NY) 2023; 15:2097-2114. [PMID: 36947712 PMCID: PMC10085599 DOI: 10.18632/aging.204594] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/16/2023] [Indexed: 03/24/2023]
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 80% of all lung cancers. Identifying key molecular targets related to the initiation, development, and metastasis of lung cancer is important for its diagnosis and target therapy. The ADAMTS families of multidomain extracellular protease enzymes have been reported to be involved in many physiological processes. In this study, we found that ADAMTS1 was highly expressed in NSCLC tissues, which promoted cell proliferation, migration, invasion, and epithelial to mesenchymal transition (EMT) of NSCLC cells. In the NSCLC tumor metastasis model involving nude mice, overexpression of ADAMTS1 promoted EMT and lung metastasis of tumor cells. Moreover, ADAMTS1 positively regulated TGF-β expression, and TGF-β was highly expressed in NSCLC tumor tissues. si-TGF-β or inhibition of TGF-β expression through the short peptide KTFR on ADAMTS1 protein could reverse the oncogenic effects of ADAMTS1 on lung cancer cells. Taken together, ADAMTS1 functioned as an oncogene in NSCLC cells by promoting TGF-β expression, indicating that ADAMTS1 has important regulatory roles in the progression of NSCLC.
Collapse
Affiliation(s)
- Xueqian Hu
- Department of Oncology, Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| | - Chunqi Jiang
- Department of Oncology, Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| | - Ning Hu
- Department of Cardiovascular Division, Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| | - Shanyi Hong
- Department of Internal Medicine, Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo, China
| |
Collapse
|
18
|
Analysis of the Genetic Relationship between Atherosclerosis and Non-Alcoholic Fatty Liver Disease through Biological Interaction Networks. Int J Mol Sci 2023; 24:ijms24044124. [PMID: 36835545 PMCID: PMC9966194 DOI: 10.3390/ijms24044124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) seems to have some molecular links with atherosclerosis (ATH); however, the molecular pathways which connect both pathologies remain unexplored to date. The identification of common factors is of great interest to explore some therapeutic strategies to improve the outcomes for those affected patients. Differentially expressed genes (DEGs) for NAFLD and ATH were extracted from the GSE89632 and GSE100927 datasets, and common up- and downregulated DEGs were identified. Subsequently, a protein-protein interaction (PPI) network based on the common DEGs was performed. Functional modules were identified, and the hub genes were extracted. Then, a Gene Ontology (GO) and pathway analysis of common DEGs was performed. DEGs analysis in NAFLD and ATH showed 21 genes that were regulated similarly in both pathologies. The common DEGs with high centrality scores were ADAMTS1 and CEBPA which appeared to be down- and up-regulated in both disorders, respectively. For the analysis of functional modules, two modules were identified. The first one was oriented to post-translational protein modification, where ADAMTS1 and ADAMTS4 were identified, and the second one mainly related to the immune response, where CSF3 was identified. These factors could be key proteins with an important role in the NAFLD/ATH axis.
Collapse
|
19
|
Wu XJ, Xie Y, Gu XX, Zhu HY, Huang LX. LncRNA XIST promotes mitochondrial dysfunction of hepatocytes to aggravate hepatic fibrogenesis via miR-539-3p/ADAMTS5 axis. Mol Cell Biochem 2023; 478:291-303. [PMID: 35794289 DOI: 10.1007/s11010-022-04506-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/15/2022] [Indexed: 02/02/2023]
Abstract
A previous study indicated that long non-coding RNA X-inactive-specific transcript (XIST) promoted ethanol-induced HSCs autophagy and activation. Considering the critical role of HSC activation in hepatic fibrosis, the aim of the present study was to reveal the exact role of XIST in liver fibrosis and its underlying mechanism. The expression of XIST in the liver from CCL4-induced mice and control mice as well as human fibrotic liver tissue and healthy liver tissue was examined. The mitochondrial reactive oxygen species (mtROS), mitochondrial membrane potential (MMP), and mitochondrial morphology were measured to assess the mitochondrial damage. The relationship between XIST and miR-539-3p as well as between miR-539-3p and ADAMTS5 was verified by a dual-luciferase reporter assay. The expression levels of HSCs activation markers were examined by Western blot. The results showed that the XIST was upregulated in fibrotic liver tissue, and overexpression of XIST induced mitochondrial dysfunction in hepatocytes. miR-539-3p directly targeted XIST, and ADAMTS5 mRNA was a downstream target of miR-539-3p. Knockdown of miR-539-3p led to an increased mitochondrial damage in hepatocytes in terms of reduced mitochondrial length, decreased MMP, and increased ROS production. However, the depletion of ADAMTS5 reversed the regulatory effect of XIST on mitochondrial damage in hepatocytes and the activation of HSCs. Our study revealed the critical role of the XIST/miR-539-3p/ADAMTS5 axis in regulating mitochondrial damage in hepatocytes and the activation of HSCs. This study may provide a potential therapeutic strategy for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Xiong-Jian Wu
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, No. 23 Youth Road, Zhanggong District, Ganzhou, 341000, Jiangxi, People's Republic of China.
| | - Yuan Xie
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, No. 23 Youth Road, Zhanggong District, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Xiao-Xiang Gu
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, No. 23 Youth Road, Zhanggong District, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Hai-Yan Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, No. 23 Youth Road, Zhanggong District, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Li-Xing Huang
- Department of Gastroenterology, The First Affiliated Hospital of Gannan Medical University, No. 23 Youth Road, Zhanggong District, Ganzhou, 341000, Jiangxi, People's Republic of China
| |
Collapse
|
20
|
Humeres C, Venugopal H, Frangogiannis NG. The Role of Mechanosensitive Signaling Cascades in Repair and Fibrotic Remodeling of the Infarcted Heart. CARDIAC AND VASCULAR BIOLOGY 2023:61-100. [DOI: 10.1007/978-3-031-23965-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Rapamycin suppresses inflammation and increases the interaction between p65 and IκBα in rapamycin-induced fatty livers. PLoS One 2023; 18:e0281888. [PMID: 36867603 PMCID: PMC9983852 DOI: 10.1371/journal.pone.0281888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/01/2023] [Indexed: 03/04/2023] Open
Abstract
Rapamycin treatment significantly increases lifespan and ameliorates several aging-related diseases in mice, making it a potential anti-aging drug. However, there are several obvious side effects of rapamycin, which may limit the broad applications of this drug. Lipid metabolism disorders such as fatty liver and hyperlipidemia are some of those unwanted side effects. Fatty liver is characterized as ectopic lipid accumulation in livers, which is usually accompanied by increased inflammation levels. Rapamycin is also a well-known anti-inflammation chemical. How rapamycin affects the inflammation level in rapamycin-induced fatty liver remains poorly understood. Here, we show that eight-day rapamycin treatment induced fatty liver and increased liver free fatty acid levels in mice, while the expression levels of inflammatory markers are even lower than those in the control mice. Mechanistically, the upstream of the pro-inflammatory pathway was activated in rapamycin-induced fatty livers, however, there is no increased NFκB nuclear translocation probably because the interaction between p65 and IκBα was enhanced by rapamycin treatment. The lipolysis pathway in the liver is also suppressed by rapamycin. Liver cirrhosis is an adverse consequence of fatty liver, while prolonged rapamycin treatment did not increase liver cirrhosis markers. Our results indicate that although fatty livers are induced by rapamycin, the fatty livers are not accompanied by increased inflammation levels, implying that rapamycin-induced fatty livers might not be as harmful as other types of fatty livers, such as high-fat diet and alcohol-induced fatty livers.
Collapse
|
22
|
Wu HJ, Kuchtey RW, Kuchtey J. Optic neuropathy associated with TGFβ dysregulation in mice with a glaucoma-causative mutation of ADAMTS10. Matrix Biol 2022; 113:83-99. [PMID: 36216203 PMCID: PMC10001177 DOI: 10.1016/j.matbio.2022.10.001] [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: 09/28/2021] [Revised: 10/06/2022] [Accepted: 10/06/2022] [Indexed: 12/11/2022]
Abstract
Glaucoma is a neurodegenerative disease that causes irreversible blindness due to loss of retinal ganglion cells (RGCs) and their axons. We previously identified a G661R mutation of ADAMTS10 (A Disintegrin And Metalloproteinase with ThromboSpondin type 1 motif 10) as the disease-causing mutation in a beagle model of glaucoma. ADAMTS10 is a secreted matrix metalloproteinase that belongs to the ADAMTS family which is involved in extracellular matrix (ECM) turnover. Previous studies have shown that ADAMTS10 binds fibrillin microfibrils, promotes their formation, and influences their fibrillin isoform composition. Here, we established a mouse model carrying the G661R mutation of ADAMTS10 (ADAMTS10G661R/G661R) to investigate its ocular phenotypes related to glaucoma and to explore possible functions of ADAMTS10. We found that ADAMTS10 was expressed in the inner retina and along RGC axons in the optic nerve. However, ADAMTS10 was not colocalized with fibrillin microfibrils in these tissues, suggesting fibrillin-independent function for ADAMTS10. In electroretinogram experiments, we found that ADAMTS10G661R/G661R mice had reduced amplitude of retinal responses to dim light stimulus, indicating RGC dysfunction. The reduced RGC function coincided with RGC axon structural changes manifested as smaller optic nerves and fewer optic nerve axons, which may contribute to glaucoma. The reduced number of optic nerve axons found for ADAMTS10G661R/G661R mice occurred early, suggesting developmental deficits. Subsequent experiments found increased apoptosis in the retina of ADAMTS10G661R/G661R mice during postnatal development, which could result in fewer RGCs produced, accounting for fewer optic nerve axons in adulthood. Consistent with a protective effect of transforming growth factor β (TGFβ) signaling against apoptosis during retinal development as shown previously by others, we found increased apoptosis accompanied by decreased TGFβ signaling in the developing retina of ADAMTS10G661R/G661R mice, suggesting a novel role for ADAMTS10 in regulating TGFβ signaling which could involve direct interaction between ADAMTS10 and latent TGFβ.
Collapse
Affiliation(s)
- Hang-Jing Wu
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, 1161 21st Ave S, AA7100 MCN, Nashville, TN 37232-8808, USA
| | - Rachel W Kuchtey
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, 1161 21st Ave S, AA7100 MCN, Nashville, TN 37232-8808, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232-0022, USA
| | - John Kuchtey
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, 1161 21st Ave S, AA7100 MCN, Nashville, TN 37232-8808, USA.
| |
Collapse
|
23
|
Pedersen RS, Nissen NI, Jensen C, Thorlacius-Ussing J, Manon-Jensen T, Olesen ML, Langholm LL, Diab HMH, Jorgensen LN, Hansen CP, Chen IM, Johansen JS, Karsdal MA, Willumsen N. Plasma Kallikrein-Activated TGF-β Is Prognostic for Poor Overall Survival in Patients with Pancreatic Ductal Adenocarcinoma and Associates with Increased Fibrogenesis. Biomolecules 2022; 12:biom12091315. [PMID: 36139154 PMCID: PMC9496221 DOI: 10.3390/biom12091315] [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: 08/19/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a hard-to-treat cancer due to the collagen-rich (fibrotic) and immune-suppressed microenvironment. A major driver of this phenomenon is transforming growth factor beta (TGF-β). TGF-β is produced in an inactive complex with a latency-associated protein (LAP) that can be cleaved by plasma kallikrein (PLK), hereby releasing active TGF-β. The aim of this study was to evaluate LAP cleaved by PLK as a non-invasive biomarker for PDAC and tumor fibrosis. An ELISA was developed for the quantification of PLK-cleaved LAP-TGF-β in the serum of 34 patients with PDAC (stage 1−4) and 20 healthy individuals. Biomarker levels were correlated with overall survival (OS) and compared to serum type III collagen (PRO-C3) and type VI collagen (PRO-C6) pro-peptides. PLK-cleaved LAP-TGF-β was higher in patients with PDAC compared to healthy individuals (p < 0.0001). High levels (>median) of PLK-cleaved LAP-TGF-β were associated with poor OS in patients with PDAC independent of age and stage (HR 2.57, 95% CI: 1.22−5.44, p = 0.0135). High levels of PLK-cleaved LAP-TGF-β were associated with high PRO-C3 and PRO-C6, indicating a relationship between the PLK-cleaved LAP-TGF-β fragment, TGF-β activity, and tumor fibrosis. If these preliminary results are validated, circulating PLK-cleaved LAP-TGF-β may be a biomarker for future clinical trials.
Collapse
Affiliation(s)
- Rasmus S. Pedersen
- Nordic Bioscience, 2730 Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence:
| | | | | | | | | | | | | | - Hadi M. H. Diab
- Digestive Disease Center, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, 2400 Copenhagen, Denmark
| | - Lars N. Jorgensen
- Digestive Disease Center, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, 2400 Copenhagen, Denmark
| | - Carsten P. Hansen
- Department of Surgery, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Inna M. Chen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark
| | - Julia S. Johansen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark
- Department of Medicine, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | | |
Collapse
|
24
|
Hanson I, Pitman KE, Altanerova U, Altaner Č, Malinen E, Edin NFJ. Low-Dose-Rate Radiation-Induced Secretion of TGF-β3 Together with an Activator in Small Extracellular Vesicles Modifies Low-Dose Hyper-Radiosensitivity through ALK1 Binding. Int J Mol Sci 2022; 23:ijms23158147. [PMID: 35897723 PMCID: PMC9332371 DOI: 10.3390/ijms23158147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/14/2022] [Accepted: 07/21/2022] [Indexed: 02/05/2023] Open
Abstract
Hyper-radiosensitivity (HRS) is the increased sensitivity to low doses of ionizing radiation observed in most cell lines. We previously demonstrated that HRS is permanently abolished in cells irradiated at a low dose rate (LDR), in a mechanism dependent on transforming growth factor β3 (TGF-β3). In this study, we aimed to elucidate the activation and receptor binding of TGF-β3 in this mechanism. T-47D cells were pretreated with inhibitors of potential receptors and activators of TGF-β3, along with addition of small extracellular vesicles (sEVs) from LDR primed cells, before their radiosensitivity was assessed by the clonogenic assay. The protein content of sEVs from LDR primed cells was analyzed with mass spectrometry. Our results show that sEVs contain TGF-β3 regardless of priming status, but only sEVs from LDR primed cells remove HRS in reporter cells. Inhibition of the matrix metalloproteinase (MMP) family prevents removal of HRS, suggesting an MMP-dependent activation of TGF-β3 in the LDR primed cells. We demonstrate a functional interaction between TGF-β3 and activin receptor like kinase 1 (ALK1) by showing that TGF-β3 removes HRS through ALK1 binding, independent of ALK5 and TGF-βRII. These results are an important contribution to a more comprehensive understanding of the mechanism behind TGF-β3 mediated removal of HRS.
Collapse
Affiliation(s)
- Ingunn Hanson
- Department of Physics, University of Oslo, 0371 Oslo, Norway; (K.E.P.); (E.M.); (N.F.J.E.)
- Correspondence:
| | - Kathinka E. Pitman
- Department of Physics, University of Oslo, 0371 Oslo, Norway; (K.E.P.); (E.M.); (N.F.J.E.)
| | - Ursula Altanerova
- Department of Stem Cell Preparation, St. Elisabeth Cancer Institute, 84505 Bratislava, Slovakia; (U.A.); (Č.A.)
| | - Čestmír Altaner
- Department of Stem Cell Preparation, St. Elisabeth Cancer Institute, 84505 Bratislava, Slovakia; (U.A.); (Č.A.)
- Cancer Research Institute, Slovak Academy of Sciences, Bratislava, 94505 Bratislava, Slovakia
| | - Eirik Malinen
- Department of Physics, University of Oslo, 0371 Oslo, Norway; (K.E.P.); (E.M.); (N.F.J.E.)
- Department of Medical Physics, Oslo University Hospital, 0379 Oslo, Norway
| | - Nina F. J. Edin
- Department of Physics, University of Oslo, 0371 Oslo, Norway; (K.E.P.); (E.M.); (N.F.J.E.)
| |
Collapse
|
25
|
Smad-dependent pathways in the infarcted and failing heart. Curr Opin Pharmacol 2022; 64:102207. [DOI: 10.1016/j.coph.2022.102207] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/10/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023]
|
26
|
Li Y, Fan W, Link F, Wang S, Dooley S. Transforming growth factor β latency: A mechanism of cytokine storage and signalling regulation in liver homeostasis and disease. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 4:100397. [PMID: 35059619 PMCID: PMC8760520 DOI: 10.1016/j.jhepr.2021.100397] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) is a potent effector in the liver, which is involved in a plethora of processes initiated upon liver injury. TGF-β affects parenchymal, non-parenchymal, and inflammatory cells in a highly context-dependent manner. Its bioavailability is critical for a fast response to various insults. In the liver – and probably in other organs – this is made possible by the deposition of a large portion of TGF-β in the extracellular matrix as an inactivated precursor form termed latent TGF-β (L-TGF-β). Several matrisomal proteins participate in matrix deposition, latent complex stabilisation, and activation of L-TGF-β. Extracellular matrix protein 1 (ECM1) was recently identified as a critical factor in maintaining the latency of deposited L-TGF-β in the healthy liver. Indeed, its depletion causes spontaneous TGF-β signalling activation with deleterious effects on liver architecture and function. This review article presents the current knowledge on intracellular L-TGF-β complex formation, secretion, matrix deposition, and activation and describes the proteins and processes involved. Further, we emphasise the therapeutic potential of toning down L-TGF-β activation in liver fibrosis and liver cancer.
Collapse
Affiliation(s)
- Yujia Li
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Weiguo Fan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford CA, USA
| | - Frederik Link
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sai Wang
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Tel.: 06213835595.
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Corresponding authors. Addresses: Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Tel.: 06213833768;
| |
Collapse
|
27
|
Duwaerts CC, Maiers JL. ER Disposal Pathways in Chronic Liver Disease: Protective, Pathogenic, and Potential Therapeutic Targets. Front Mol Biosci 2022; 8:804097. [PMID: 35174209 PMCID: PMC8841999 DOI: 10.3389/fmolb.2021.804097] [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: 10/28/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum is a central player in liver pathophysiology. Chronic injury to the ER through increased lipid content, alcohol metabolism, or accumulation of misfolded proteins causes ER stress, dysregulated hepatocyte function, inflammation, and worsened disease pathogenesis. A key adaptation of the ER to resolve stress is the removal of excess or misfolded proteins. Degradation of intra-luminal or ER membrane proteins occurs through distinct mechanisms that include ER-associated Degradation (ERAD) and ER-to-lysosome-associated degradation (ERLAD), which includes macro-ER-phagy, micro-ER-phagy, and Atg8/LC-3-dependent vesicular delivery. All three of these processes are critical for removing misfolded or unfolded protein aggregates, and re-establishing ER homeostasis following expansion/stress, which is critical for liver function and adaptation to injury. Despite playing a key role in resolving ER stress, the contribution of these degradative processes to liver physiology and pathophysiology is understudied. Analysis of publicly available datasets from diseased livers revealed that numerous genes involved in ER-related degradative pathways are dysregulated; however, their roles and regulation in disease progression are not well defined. Here we discuss the dynamic regulation of ER-related protein disposal pathways in chronic liver disease and cell-type specific roles, as well as potentially targetable mechanisms for treatment of chronic liver disease.
Collapse
Affiliation(s)
- Caroline C. Duwaerts
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jessica L. Maiers
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
28
|
Toba H, Ikemoto MJ, Kobara M, Nakata T. Secreted protein acidic and rich in cysteine (SPARC) and a disintegrin and metalloproteinase with thrombospondin type 1 motif (ADAMTS1) increments by the renin-angiotensin system induce renal fibrosis in deoxycorticosterone acetate-salt hypertensive rats. Eur J Pharmacol 2022; 914:174681. [PMID: 34871556 DOI: 10.1016/j.ejphar.2021.174681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023]
Abstract
Secreted protein acidic and rich in cysteine (SPARC), an extracellular matrix (ECM) protein, was recently shown to induce collagen deposition through the production of a disintegrin and metalloproteinase with thrombospondin type 1 motif (ADAMTS1) in the aging heart. ADAMTS1 regulates ECM turnover by degrading ECM components, and its excessive activation contributes to various pathological states, including fibrosis. The present study investigated the pathophysiological regulation and role of SPARC and ADAMTS1 in renal fibrosis using uninephrectomized rats treated with deoxycorticosterone acetate (DOCA, 40 mg/kg/week, subcutaneously) and salt (1% in drinking water). The administration of DOCA and salt gradually and significantly elevated systolic blood pressure during the 3-week treatment period, induced proteinuria, decreased creatinine clearance, and increased NADPH oxidase-derived superoxide production, malondialdehyde concentrations, and monocyte chemoattractant protein-1 and osteopontin expression in the kidneys. Glomerulosclerosis, fibrillar collagen deposition, and transforming growth factor-β expression increased in a time-dependent manner, and SPARC and ADAMTS1 expression showed a similar pattern to these changes. The angiotensin II type-1 receptor blocker losartan suppressed the overexpression of SPARC and ADAMTS1, and an in vitro exposure to angiotensin II induced the production of both SPARC and ADAMTS1 in renal fibroblast NRK-49F cells. Knockdown of the SPARC gene with small interfering RNA reduced all forms (the 110-kDa latent and 87- and 65-kDa bioactive forms) of ADAMTS1 expression as well as collagen production. These results suggest that SPARC is induced by the renin-angiotensin system and may be a fibrogenic factor, at least in part, by producing ADAMTS1 in hypertensive renal disease.
Collapse
Affiliation(s)
- Hiroe Toba
- Department of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan.
| | - Mitsushi J Ikemoto
- Molecular Composite Physiology Research Group, Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Miyuki Kobara
- Department of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tetsuo Nakata
- Department of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
29
|
Abstract
Transforming growth factor-β (TGFβ) isoforms are upregulated and activated in myocardial diseases and have an important role in cardiac repair and remodelling, regulating the phenotype and function of cardiomyocytes, fibroblasts, immune cells and vascular cells. Cardiac injury triggers the generation of bioactive TGFβ from latent stores, through mechanisms involving proteases, integrins and specialized extracellular matrix (ECM) proteins. Activated TGFβ signals through the SMAD intracellular effectors or through non-SMAD cascades. In the infarcted heart, the anti-inflammatory and fibroblast-activating actions of TGFβ have an important role in repair; however, excessive or prolonged TGFβ signalling accentuates adverse remodelling, contributing to cardiac dysfunction. Cardiac pressure overload also activates TGFβ cascades, which initially can have a protective role, promoting an ECM-preserving phenotype in fibroblasts and preventing the generation of injurious, pro-inflammatory ECM fragments. However, prolonged and overactive TGFβ signalling in pressure-overloaded cardiomyocytes and fibroblasts can promote cardiac fibrosis and dysfunction. In the atria, TGFβ-mediated fibrosis can contribute to the pathogenic substrate for atrial fibrillation. Overactive or dysregulated TGFβ responses have also been implicated in cardiac ageing and in the pathogenesis of diabetic, genetic and inflammatory cardiomyopathies. This Review summarizes the current evidence on the role of TGFβ signalling in myocardial diseases, focusing on cellular targets and molecular mechanisms, and discussing challenges and opportunities for therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
30
|
TGF-β1 signaling can worsen NAFLD with liver fibrosis backdrop. Exp Mol Pathol 2021; 124:104733. [PMID: 34914973 DOI: 10.1016/j.yexmp.2021.104733] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 11/08/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is characterized by the accumulation of fats in the liver. Relatively benign NAFLD often progresses to fibrosis, cirrhosis, and liver malignancies. Although NAFLD precedes fibrosis, continuous lipid overload keeps fueling fibrosis and the process of disease progression remains unhindered. It is well known that TGF-β1 plays its part in liver fibrosis, yet its effects on liver lipid overload remain unknown. As TGF-β1 signaling has been increasingly attempted to manage liver fibrosis, its actions on the primary suspect (NAFLD) are easily ignored. The complex interaction of inflammatory stress and lipid accumulation aided by mediators scuh as pro-inflammatory interleukins and TGF-β1 forms the basis of NAFLD progression. Anticipatorily, the inhibition of TGF-β1 signaling during anti-fibrotic treatment should reverse the NAFLD though the data remain scattered on this subject to date. TGF-β1 signaling pathway is an important drug target in liver fibrosis and abundant literature is available on it, but its direct effects on NAFLD are rarely studied. This review aims to cover the pathogenesis of NAFLD focusing on the role of the TGF-β1 in the disease progression, especially in the backdrop of liver fibrosis.
Collapse
|
31
|
Zhang H, Ma Y, Cheng X, Wu D, Huang X, Chen B, Ren Y, Jiang W, Tang X, Bai T, Chen Y, Zhao Y, Zhang C, Xiao X, Liu J, Deng Y, Ye T, Chen L, Liu HM, Friedman SL, Chen L, Ding BS, Cao Z. Targeting epigenetically maladapted vascular niche alleviates liver fibrosis in nonalcoholic steatohepatitis. Sci Transl Med 2021; 13:eabd1206. [PMID: 34613814 DOI: 10.1126/scitranslmed.abd1206] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hua Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yongyuan Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Xinying Cheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingming Huang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Chen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yafeng Ren
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Jiang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Bai
- Department of Cardiology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yutian Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yilin Zhao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Chunxue Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Xia Xiao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Deng
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Tinghong Ye
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Han-Min Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Scott L Friedman
- Fibrosis Research Program, Division of Pulmonary and Critical Care Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Liping Chen
- Department of Biliary Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Fibrosis Research Program, Division of Pulmonary and Critical Care Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Division of Regenerative Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Zhongwei Cao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Fibrosis Research Program, Division of Pulmonary and Critical Care Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
32
|
Savchenko RR, Murashkina AA, Fishman VS, Sukhikh ES, Vertinsky AV, Sukhikh LG, Serov OL, Lebedev IN, Vasilyev SA. Effect of ADAMTS1 Differential Expression on the Radiation-Induced Response of HеLа Cell Line. RUSS J GENET+ 2021. [DOI: 10.1134/s1022795421070127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
33
|
Wang Y, Xiao Y, Zheng Y, Yang L, Wang D. An anti-ADAMTS1 treatment relieved muscle dysfunction and fibrosis in dystrophic mice. Life Sci 2021; 281:119756. [PMID: 34175316 DOI: 10.1016/j.lfs.2021.119756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 11/27/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is caused by mutations in the dystrophin gene, accompanied by aberrant extracellular matrix synthesis and muscle damage. ADAMTS1 metalloproteinase was reported increased in dystrophin-deficient mdx mouse. The aim of this study was to explore the role of ADAMTS1 in muscle function, fibrosis and damage, and respiratory function of mdx mice. 102 DMD patients and their mothers were included in this study. Multiplex ligation dependent probe amplification (MLPA) assay and Next-generation sequencing (NGS) were adopted to do genetic diagnosis. Dystrophin-deficient mdx mice were treated with anti-ADAMTS1 antibody (anti-ADAMTS1) for three weeks. The results showed that ADAMTS1 was increased in gastrocnemius muscle of mdx mice and serum of DMD patients. Anti-ADAMTS1 treatment increased Versican transcription but suppressed versican protein expression. Besides, we found anti-ADAMTS1 improved muscle strength, diaphragm and extensor digitorum longus muscles functions in mdx mice. Meanwhile, muscle fibrosis and damage were attenuated in anti-ADAMTS1 treated dystrophic mice. In summary, anti-ADAMTS1 antibody relieved muscle dysfunction and fibrosis in dystrophic mice. It is suggested that ADAMTS1 is a potential target for developing new biological therapies for DMD.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi 'an 710004, Shaanxi Province, China; Department of Neurology, Xi'an Children's Hospital, Xi'an 710000, Shaanxi Province, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi 'an 710004, Shaanxi Province, China.
| | - Yanyan Zheng
- Department of Neurology, Xi'an Children's Hospital, Xi'an 710000, Shaanxi Province, China
| | - Le Yang
- Department of Neurology, Xi'an Children's Hospital, Xi'an 710000, Shaanxi Province, China
| | - Dong Wang
- Department of Neurology, Xi'an Children's Hospital, Xi'an 710000, Shaanxi Province, China
| |
Collapse
|
34
|
Théret N, Bouezzeddine F, Azar F, Diab-Assaf M, Legagneux V. ADAM and ADAMTS Proteins, New Players in the Regulation of Hepatocellular Carcinoma Microenvironment. Cancers (Basel) 2021; 13:cancers13071563. [PMID: 33805340 PMCID: PMC8037375 DOI: 10.3390/cancers13071563] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Members of the adamalysin family are multi-domain proteins involved in many cancer-related functions. In this review, we will examine the literature on the involvement of adamalysins in hepatocellular carcinoma progression and their importance in the tumor microenvironment where they regulate the inflammatory response and the epithelial–mesenchymal transition. We complete this review with an analysis of adamalysin expression in a large cohort of patients with hepatocellular carcinoma from The Cancer Genome Atlas (TCGA) database. These original results give a new insight into the involvement of all adamalysins in the primary liver cancer. Abstract The tumor microenvironment plays a major role in tumor growth, invasion and resistance to chemotherapy, however understanding how all actors from microenvironment interact together remains a complex issue. The tumor microenvironment is classically represented as three closely connected components including the stromal cells such as immune cells, fibroblasts, adipocytes and endothelial cells, the extracellular matrix (ECM) and the cytokine/growth factors. Within this space, proteins of the adamalysin family (ADAM for a disintegrin and metalloproteinase; ADAMTS for ADAM with thrombospondin motifs; ADAMTSL for ADAMTS-like) play critical roles by modulating cell–cell and cell–ECM communication. During last decade, the implication of adamalysins in the development of hepatocellular carcinoma (HCC) has been supported by numerous studies however the functional characterization of most of them remain unsettled. In the present review we propose both an overview of the literature and a meta-analysis of adamalysins expression in HCC using data generated by The Cancer Genome Atlas (TCGA) Research Network.
Collapse
Affiliation(s)
- Nathalie Théret
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
- Correspondence:
| | - Fidaa Bouezzeddine
- Molecular Cancer and Pharmaceutical Biology Laboratory, Faculty of Sciences II, Lebanese University Fanar, 1500 Beirut, Lebanon; (F.B.); (M.D.-A.)
| | - Fida Azar
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
| | - Mona Diab-Assaf
- Molecular Cancer and Pharmaceutical Biology Laboratory, Faculty of Sciences II, Lebanese University Fanar, 1500 Beirut, Lebanon; (F.B.); (M.D.-A.)
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en santé, Environnement et Travail)-UMR_S1085, University of Rennes 1, 35000 Rennes, France; (F.A.); (V.L.)
| |
Collapse
|
35
|
de Assis Lima M, da Silva SV, Serrano-Garrido O, Hülsemann M, Santos-Neres L, Rodríguez-Manzaneque JC, Hodgson L, Freitas VM. Metalloprotease ADAMTS-1 decreases cell migration and invasion modulating the spatiotemporal dynamics of Cdc42 activity. Cell Signal 2021; 77:109827. [PMID: 33161094 PMCID: PMC7723338 DOI: 10.1016/j.cellsig.2020.109827] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 01/06/2023]
Abstract
ADAMTSs (A Disintegrin And Metalloproteinase with ThromboSpondin motifs) are secreted proteases dependent on Zn2+/Ca2+, involved in physiological and pathological processes and are part of the extracellular matrix (ECM). Here, we investigated if ADAMTS-1 is required for invasion and migration of cells and the possible mechanism involved. In order to test ADAMTS-1's role in ovarian cancer cells (CHO, NIH-OVCAR-3 and ES2) and NIH-3 T3 fibroblasts, we modified the levels of ADAMTS-1 and compared those to parental. Cells exposed to ADAMTS-1-enriched medium exhibited a decline in cell migration and invasion when compared to controls with or without a functional metalloproteinase domain. The opposite was observed in cells when ADAMTS-1 was deleted via the CRISPR/Cas9 approach. The decline in ADAMTS-1 levels enhanced the phosphorylated form of Src and FAK. We also evaluated the activities of cellular Rho GTPases from cell lysates using the GLISA® kit. The Cdc42-GTP signal was significantly increased in the CRISPR ADAMTS-1 ES-2 cells. By a Förster resonance energy transfer (FRET) biosensor for Cdc42 activity in ES-2 cells we demonstrated that Cdc42 activity was strongly polarized at the leading edge of migrating cells with ADAMTS-1 deletion, compared to the wild type cells. As conclusion, ADAMTS-1 inhibits proliferation, polarization and migration.
Collapse
Affiliation(s)
- Maíra de Assis Lima
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes 1524, Ed Biomédicas 1 sala 428, São Paulo, SP 05508-000, Brazil.
| | - Suély Vieira da Silva
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes 1524, Ed Biomédicas 1 sala 428, São Paulo, SP 05508-000, Brazil.
| | - Orlando Serrano-Garrido
- GENYO, Centre for Genomics and Oncological Research, Avenida de la Ilustración, 114, Granada 18016, Spain.
| | - Maren Hülsemann
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America, Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America.
| | - Luana Santos-Neres
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes 1524, Ed Biomédicas 1 sala 428, São Paulo, SP 05508-000, Brazil.
| | | | - Louis Hodgson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America, Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, 10461, United States of America.
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes 1524, Ed Biomédicas 1 sala 428, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
36
|
Zhou S, Zhu Y, Li Z, Zhu Y, He Z, Zhang C. Exosome-derived long non-coding RNA ADAMTS9-AS2 suppresses progression of oral submucous fibrosis via AKT signalling pathway. J Cell Mol Med 2020; 25:2262-2273. [PMID: 33345447 PMCID: PMC7882956 DOI: 10.1111/jcmm.16219] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/23/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Oral submucosal fibrosis (OSF) is one of the pre‐cancerous lesions of oral squamous cell carcinoma (OSCC). Its malignant rate is increasing, but the mechanism of malignancy is not clear. We previously have elucidated the long non‐coding RNA (lncRNA) expression profile during OSF progression at the genome‐wide level. However, the role of lncRNA ADAMTS9‐AS2 in OSF progression via extracellular communication remains unclear. lncRNA ADAMTS9‐AS2 is down‐regulated in OSCC tissues compared with OSF and normal mucous tissues. Low ADAMTS9‐AS2 expression is associated with poor overall survival. ADAMTS9‐AS2 is frequently methylated in OSCC tissues, but not in normal oral mucous and OSF tissues, suggesting tumour‐specific methylation. Functional studies reveal that exosomal ADAMTS9‐AS2 suppresses OSCC cell growth, migration and invasion in vitro. Mechanistically, exosomal ADAMTS9‐AS2 inhibits AKT signalling pathway and regulates epithelial‐mesenchymal transition markers. Through profiling miRNA expression profile regulated by exosomal ADAMTS9‐AS2, significantly enriched pathways include metabolic pathway, PI3K‐Akt signalling pathway and pathways in cancer, indicating that exosomal ADAMTS9‐AS2 exerts its functions through interacting with miRNAs during OSF progression. Thus, our findings highlight the crucial role of ADAMTS9‐AS2 in the cell microenvironment during OSF carcinogenesis, which is expected to become a marker for early diagnosis of OSCC.
Collapse
Affiliation(s)
- Shanghui Zhou
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yun Zhu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhenming Li
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yonggan Zhu
- Department of Nursing, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijing He
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenping Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
37
|
Ricard-Blum S, Miele AE. Omic approaches to decipher the molecular mechanisms of fibrosis, and design new anti-fibrotic strategies. Semin Cell Dev Biol 2020; 101:161-169. [DOI: 10.1016/j.semcdb.2019.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
|
38
|
Lambert J, Makin K, Akbareian S, Johnson R, Alghamdi AAA, Robinson SD, Edwards DR. ADAMTS-1 and syndecan-4 intersect in the regulation of cell migration and angiogenesis. J Cell Sci 2020; 133:jcs.235762. [PMID: 32269093 PMCID: PMC7157938 DOI: 10.1242/jcs.235762] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
ADAMTS-1 is an extracellular protease with critical roles in organogenesis and angiogenesis. Here we demonstrate a functional convergence of ADAMTS-1 and the transmembrane heparan sulfate proteoglycan syndecan-4 in influencing adhesion, migration and angiogenesis. Knockdown of ADAMTS-1 in endothelial cells resulted in a parallel reduction in cell surface syndecan-4, attributable to increased matrix metalloproteinase-9 (MMP9) activity. Knockdown of either ADAMTS-1 or syndecan-4 increased cellular responses to vascular endothelial growth factor A isoform VEGFA164, and increased ex vivo aortic ring microvessel sprouting. On fibronectin, knockdown of either protein enhanced migration and promoted formation of long α5 integrin-containing fibrillar adhesions. However, integrin α5 null cells still showed increased migration in response to ADAMTS-1 and syndecan-4 siRNA treatment. Plating of naïve endothelial cells on cell-conditioned matrix from ADAMTS-1 and syndecan-4 knockdown cells demonstrated that the altered adhesive behaviour was matrix dependent, and this correlated with a lack of expression of fibulin-1: an extracellular matrix co-factor for ADAMTS-1 that is known to inhibit migration. These findings support the notion that ADAMTS-1 and syndecan-4 are functionally interconnected in regulating cell migration and angiogenesis, via collaboration with MMP9 and fibulin-1. This article has an associated First Person interview with the first author of the paper. Summary: ADAMTS-1 and syndecan-4 collaborate to regulate cell adhesion, migration and integrin α5 trafficking, and to sequester VEGFA164, inhibiting angiogenesis.
Collapse
Affiliation(s)
- Jordi Lambert
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Kate Makin
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Sophia Akbareian
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Robert Johnson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Abdullah A A Alghamdi
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.,Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Stephen D Robinson
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.,Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Dylan R Edwards
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
39
|
Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020; 217:e20190103. [PMID: 32997468 PMCID: PMC7062524 DOI: 10.1084/jem.20190103] [Citation(s) in RCA: 713] [Impact Index Per Article: 142.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022] Open
Abstract
TGF-β is extensively implicated in the pathogenesis of fibrosis. In fibrotic lesions, spatially restricted generation of bioactive TGF-β from latent stores requires the cooperation of proteases, integrins, and specialized extracellular matrix molecules. Although fibroblasts are major targets of TGF-β, some fibrogenic actions may reflect activation of other cell types, including macrophages, epithelial cells, and vascular cells. TGF-β–driven fibrosis is mediated through Smad-dependent or non-Smad pathways and is modulated by coreceptors and by interacting networks. This review discusses the role of TGF-β in fibrosis, highlighting mechanisms of TGF-β activation and signaling, the cellular targets of TGF-β actions, and the challenges of therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
40
|
Tanwar S, Rhodes F, Srivastava A, Trembling PM, Rosenberg WM. Inflammation and fibrosis in chronic liver diseases including non-alcoholic fatty liver disease and hepatitis C. World J Gastroenterol 2020; 26:109-133. [PMID: 31969775 PMCID: PMC6962431 DOI: 10.3748/wjg.v26.i2.109] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/18/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023] Open
Abstract
At present chronic liver disease (CLD), the third commonest cause of premature death in the United Kingdom is detected late, when interventions are ineffective, resulting in considerable morbidity and mortality. Injury to the liver, the largest solid organ in the body, leads to a cascade of inflammatory events. Chronic inflammation leads to the activation of hepatic stellate cells that undergo trans-differentiation to become myofibroblasts, the main extra-cellular matrix producing cells in the liver; over time increased extra-cellular matrix production results in the formation of liver fibrosis. Although fibrogenesis may be viewed as having evolved as a “wound healing” process that preserves tissue integrity, sustained chronic fibrosis can become pathogenic culminating in CLD, cirrhosis and its associated complications. As the reference standard for detecting liver fibrosis, liver biopsy, is invasive and has an associated morbidity, the diagnostic assessment of CLD by non-invasive testing is attractive. Accordingly, in this review the mechanisms by which liver inflammation and fibrosis develop in chronic liver diseases are explored to identify appropriate and meaningful diagnostic targets for clinical practice. Due to differing disease prevalence and treatment efficacy, disease specific diagnostic targets are required to optimally manage individual CLDs such as non-alcoholic fatty liver disease and chronic hepatitis C infection. To facilitate this, a review of the pathogenesis of both conditions is also conducted. Finally, the evidence for hepatic fibrosis regression and the mechanisms by which this occurs are discussed, including the current use of antifibrotic therapy.
Collapse
Affiliation(s)
- Sudeep Tanwar
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
- Department of Gastroenterology, Whipps Cross University Hospital, Barts Health NHS Trust, Leytonstone, London E11 1NR, United Kingdom
| | - Freya Rhodes
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| | - Ankur Srivastava
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| | - Paul M Trembling
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| | - William M Rosenberg
- UCL Institute for Liver and Digestive Health, Division of Medicine, University College London, Royal Free Campus, Hampstead, London NW3 2PF United Kingdom
| |
Collapse
|
41
|
Roy R, Morad G, Jedinak A, Moses MA. Metalloproteinases and their roles in human cancer. Anat Rec (Hoboken) 2019; 303:1557-1572. [PMID: 31168956 DOI: 10.1002/ar.24188] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/27/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
It is now widely appreciated that members of the matrix metalloproteinase (MMP) family of enzymes play a key role in cancer development and progression along with many of the hallmarks associated with them. The activity of these enzymes has been directly implicated in extracellular matrix remodeling, the processing of growth factors and receptors, the modulation of cell migration, proliferation, and invasion, the epithelial to mesenchymal transition, the regulation of immune responses, and the control of angiogenesis. Certain MMP family members have been validated as biomarkers of a variety of human cancers including those of the breast, brain, pancreas, prostate, ovary, and others. The related metalloproteinases, the A disintegrin and metalloproteinases (ADAMs), share a number of these functions as well. Here, we explore these essential metalloproteinases and some of their disease-associated activities in detail as well as some of their complementary translational potential. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roopali Roy
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Golnaz Morad
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andrej Jedinak
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marsha A Moses
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Armutcu F, Demircan K, Yildirim U, Namuslu M, Yagmurca M, Celik HT. Hypoxia causes important changes of extracellular matrix biomarkers and ADAMTS proteinases in the adriamycin-induced renal fibrosis model. Nephrology (Carlton) 2019; 24:863-875. [PMID: 30719800 DOI: 10.1111/nep.13572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2019] [Indexed: 12/12/2022]
Abstract
AIM Renal fibrosis is a common cause of renal dysfunction with chronic kidney diseases. This process is characterized by excessive production of extracellular matrix (ECM) or inhibition of ECM degradation. A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) proteinases, which are widely presented in mammals, have very critical roles in ECM remodelling. We aimed to study the role of ADAMTS proteinases and some of the ECM markers in the pathogenesis of renal fibrosis and to investigate the effects of hypoxia on these biomarkers. METHODS In addition to the control group, Adriamycin (ADR) treated rats were divided into four groups as ADR, sham and two hypoxia groups. Renal nephropathy was assessed biochemical assays, pathological and immunohistochemical staining methods. The expression of ADAMTSs and mRNA were determined using Western blotting and real-time PCR, respectively. RESULTS Renal dysfuntion and tissue damage in favour of ECM accumulation and renal fibrosis were observed in the ADR group. This was approved by remarkable changes in the expression of ADAMTS such as increased ADAMTS-1, -12 and -15. In addition, it was found that hypoxia and duration of hypoxia enhanced markers of tubulointerstitial fibrosis in the rat kidney tissues. Also, expression differences especially in ADAMTS-1, -6 and -15 were observed in the hypoxia groups. The variable and different expression patterns of ADAMTS proteinases in the ADR-induced renal fibrosis suggest that ADAMTS family members are involved in the development and progression of fibrosis. CONCLUSION The expression changes of ADAMTS proteinases in kidney and association with hypoxia have potential clues to contribute to the early diagnosis and treatment options of renal fibrosis.
Collapse
Affiliation(s)
- Ferah Armutcu
- Department of Biochemistry, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Kadir Demircan
- Department of Medical Biology, Turgut Ozal University, Faculty of Medicine, Ankara, Turkey
| | - Umran Yildirim
- Department of Pathology, Turgut Ozal University, Faculty of Medicine, Ankara, Turkey
| | - Mehmet Namuslu
- Department of Biochemistry, Turgut Ozal University, Faculty of Medicine, Ankara, Turkey
| | - Murat Yagmurca
- Health Sciences University, Higher Specialization Training and Research Hospital, Histology and Embryology Clinic, Bursa, Turkey
| | - Hüseyin T Celik
- Department of Biochemistry, Turgut Ozal University, Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
43
|
Synthesis and biological evaluation of quercetin and resveratrol peptidyl derivatives as potential anticancer and antioxidant agents. Amino Acids 2018; 51:319-329. [PMID: 30392096 DOI: 10.1007/s00726-018-2668-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 10/16/2018] [Indexed: 12/26/2022]
Abstract
Quercetin and resveratrol are polyphenolic compounds, members of the flavonoid and the stilbene family, respectively, both medicinally important as dietary anticancer and antioxidant agents. They are present in a variety of foods-including fruits, vegetables, tea, wine, as well as other dietary supplements-and are responsible for various health benefits. Different quercetin and resveratrol esters of Leu/Met-enkephalin and tetrapeptide Leu-Ser-Lys-Leu (LSKL) were synthesized as model systems for monitoring the influence of the peptides on biological activity of resveratrol and quercetin. General formula of the main peptidyl-quercetin derivatives is 2-[3-(aa)n-4-hydroxyphenyl]-3,5,7-tri-hydroxy-4H-1-benzopyran-4-on, and the general formula of the main peptidyl-resveratrol derivatives is (E)-5-[4-(aa)n)styryl]benzene-1,3-diol. The antioxidant and anticancer activities of prepared compounds were investigated. Significant anticancer activity was obtained for the LSKL-based both quercetin and resveratrol derivatives. All prepared compounds exhibit antioxidant activity, in particular quercetin derivative containing Met-enkephalin.
Collapse
|
44
|
Role of hepatic stellate cell (HSC)-derived cytokines in hepatic inflammation and immunity. Cytokine 2018; 124:154542. [PMID: 30241896 DOI: 10.1016/j.cyto.2018.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/01/2018] [Accepted: 09/07/2018] [Indexed: 12/15/2022]
Abstract
In their quiescent state, Hepatic stellate cells (HSCs), are present in the sub-endothelial space of Disse and have minimal interaction with immune cells. However, upon activation following injury, HSCs directly or indirectly interact with various immune cells that enter the space of Disse and thereby regulate diverse hepatic function and immune physiology. Other than the normal physiological functions of HSCs such as hepatic homeostasis, maturation and differentiation, they also participate in hepatic inflammation by releasing a battery of inflammatory cytokines and chemokines and interacting with other liver cells. Here, we have reviewed the role of HSC in the pathogenesis of liver inflammation and some infectious diseases in order to understand how the interplay between immune cells and HSCs regulates the overall outcome and disease pathology.
Collapse
|
45
|
ADAMTS1 protease is required for a balanced immune cell repertoire and tumour inflammatory response. Sci Rep 2018; 8:13103. [PMID: 30166561 PMCID: PMC6117274 DOI: 10.1038/s41598-018-31288-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
Recent advances have emphasized the relevance of studying the extracellular microenvironment given its main contribution to tissue homeostasis and disease. Within this complex scenario, we have studied the extracellular protease ADAMTS1 (a disintegrin and metalloprotease with thrombospondin motif 1), implicated in vascularization and development, with reported anti- and pro-tumorigenic activities. In this work we performed a detailed study of the vasculature and substrates in adult organs of wild type and Adamts1-deficient mice. In addition to the expected alterations of organs like kidney, heart and aorta, we found that the lack of ADAMTS1 differently affects lymphocyte and myeloid populations in the spleen and bone marrow. The study of the substrate versican also revealed its alteration in the absence of the protease. With such premises, we challenged our mice with subcutaneous B16F1 syngeneic tumours and closely evaluated the immune repertoire in the tumours but also in the distant spleen and bone marrow. Our results confirmed a pro-inflammatory landscape in the absence of ADAMTS1, correlating with tumour blockade, supporting its novel role as a modulator of the immune cell response.
Collapse
|
46
|
Xiong Y, Wu S, Yu H, Wu J, Wang Y, Li H, Huang H, Zhang H. miR-190 promotes HCC proliferation and metastasis by targeting PHLPP1. Exp Cell Res 2018; 371:185-195. [PMID: 30092222 DOI: 10.1016/j.yexcr.2018.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 08/03/2018] [Accepted: 08/05/2018] [Indexed: 01/10/2023]
Abstract
miRNAs regulate gene expression and enable clinicians to distinguish between benign and malignant tissues in cancers. PH domain leucine-rich repeat-containing protein phosphatase 1 (PHLPP1) is known to be a tumour suppressor. A lentiviral overexpression system was used to stably express miR-190, leading to the enhancement of hepatocellular carcinoma (HCC) proliferation and metastasis as a result of inhibited PHLPP1 expression. The results showed that stable miR-190 expression increased the expression of EMT-related proteins (Snail and TCF8/ZEB1) as well as the phosphorylation of Akt at Ser473 and the expression of a disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1). However, restoring PHLPP1 expression counteracted the effects of miR-190 on HCC proliferation, migration and invasion. The results of the animal experiments showed that miR-190 improved the HepG2 cell tumour formation and lung metastasis ability. Stable miR-190 overexpression leads to the downregulation of PHLPP1 protein expression. miR-190 has potential as a target in the treatment and diagnosis of HCC.
Collapse
Affiliation(s)
- Yuzhen Xiong
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Shang Wu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Huajun Yu
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Jun Wu
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Yajun Wang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, China; Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Huimin Li
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Hui Huang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| | - Haitao Zhang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
47
|
Ricard-Blum S, Baffet G, Théret N. Molecular and tissue alterations of collagens in fibrosis. Matrix Biol 2018; 68-69:122-149. [DOI: 10.1016/j.matbio.2018.02.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 02/07/2023]
|
48
|
Signaling Crosstalk of TGF-β/ALK5 and PAR2/PAR1: A Complex Regulatory Network Controlling Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19061568. [PMID: 29795022 PMCID: PMC6032192 DOI: 10.3390/ijms19061568] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Both signaling by transforming growth factor-β (TGF-β) and agonists of the G Protein-coupled receptors proteinase-activated receptor-1 (PAR1) and -2 (PAR2) have been linked to tissue fibrosis and cancer. Intriguingly, TGF-β and PAR signaling either converge on the regulation of certain matrix genes overexpressed in these pathologies or display mutual regulation of their signaling components, which is mediated in part through sphingosine kinases and sphingosine-1-phosphate and indicative of an intimate signaling crosstalk between the two pathways. In the first part of this review, we summarize the various regulatory interactions that have been discovered so far according to the organ/tissue in which they were described. In the second part, we highlight the types of signaling crosstalk between TGF-β on the one hand and PAR2/PAR1 on the other hand. Both ligand–receptor systems interact at various levels and by several mechanisms including mutual regulation of ligand–ligand, ligand–receptor, and receptor–receptor at the transcriptional, post-transcriptional, and receptor transactivation levels. These mutual interactions between PAR2/PAR1 and TGF-β signaling components eventually result in feed-forward loops/vicious cycles of matrix deposition and malignant traits that exacerbate fibrosis and oncogenesis, respectively. Given the crucial role of PAR2 and PAR1 in controlling TGF-β receptor activation, signaling, TGF-β synthesis and bioactivation, combining PAR inhibitors with TGF-β blocking agents may turn out to be more efficient than targeting TGF-β alone in alleviating unwanted TGF-β-dependent responses but retaining the beneficial ones.
Collapse
|
49
|
Fava M, Barallobre-Barreiro J, Mayr U, Lu R, Didangelos A, Baig F, Lynch M, Catibog N, Joshi A, Barwari T, Yin X, Jahangiri M, Mayr M. Role of ADAMTS-5 in Aortic Dilatation and Extracellular Matrix Remodeling. Arterioscler Thromb Vasc Biol 2018; 38:1537-1548. [PMID: 29622560 PMCID: PMC6026471 DOI: 10.1161/atvbaha.117.310562] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 03/19/2018] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Thoracic aortic aneurysm (TAA), a degenerative disease of the aortic wall, is accompanied by changes in the structure and composition of the aortic ECM (extracellular matrix). The ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of proteases has recently been implicated in TAA formation. This study aimed to investigate the contribution of ADAMTS-5 to TAA development. Approach and Results— A model of aortic dilatation by AngII (angiotensin II) infusion was adopted in mice lacking the catalytic domain of ADAMTS-5 (Adamts5Δcat). Adamts5Δcat mice showed an attenuated rise in blood pressure while displaying increased dilatation of the ascending aorta (AsAo). Interestingly, a proteomic comparison of the aortic ECM from AngII-treated wild-type and Adamts5Δcat mice revealed versican as the most upregulated ECM protein in Adamts5Δcat mice. This was accompanied by a marked reduction of ADAMTS-specific versican cleavage products (versikine) and a decrease of LRP1 (low-density lipoprotein-related protein 1). Silencing LRP1 expression in human aortic smooth muscle cells reduced the expression of ADAMTS5, attenuated the generation of versikine, but increased soluble ADAMTS-1. A similar increase in ADAMTS-1 was observed in aortas of AngII-treated Adamts5Δcat mice but was not sufficient to maintain versican processing and prevent aortic dilatation. Conclusions— Our results support the emerging role of ADAMTS proteases in TAA. ADAMTS-5 rather than ADAMTS-1 is the key protease for versican regulation in murine aortas. Further studies are needed to define the ECM substrates of the different ADAMTS proteases and their contribution to TAA formation.
Collapse
MESH Headings
- ADAMTS1 Protein/metabolism
- ADAMTS5 Protein/deficiency
- ADAMTS5 Protein/genetics
- ADAMTS5 Protein/metabolism
- Angiotensin II
- Animals
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Cells, Cultured
- Dilatation, Pathologic
- Disease Models, Animal
- Extracellular Matrix/enzymology
- Extracellular Matrix/pathology
- Humans
- Low Density Lipoprotein Receptor-Related Protein-1/genetics
- Low Density Lipoprotein Receptor-Related Protein-1/metabolism
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle
- Receptors, LDL/metabolism
- Tumor Suppressor Proteins/metabolism
- Vascular Remodeling
- Versicans/metabolism
Collapse
Affiliation(s)
- Marika Fava
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
- St George's University of London, NHS Trust, United Kingdom (M.F., M.J.)
- Cardiovascular Institute, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (M.F., M.M.)
| | - Javier Barallobre-Barreiro
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ursula Mayr
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ruifang Lu
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Athanasios Didangelos
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ferheen Baig
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Marc Lynch
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Norman Catibog
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Abhishek Joshi
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Temo Barwari
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Xiaoke Yin
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Marjan Jahangiri
- St George's University of London, NHS Trust, United Kingdom (M.F., M.J.)
| | - Manuel Mayr
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
- Cardiovascular Institute, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (M.F., M.M.)
| |
Collapse
|
50
|
Reed KM, Mendoza KM, Abrahante JE, Coulombe RA. Comparative Response of the Hepatic Transcriptomes of Domesticated and Wild Turkey to Aflatoxin B₁. Toxins (Basel) 2018; 10:toxins10010042. [PMID: 29342849 PMCID: PMC5793129 DOI: 10.3390/toxins10010042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/15/2022] Open
Abstract
The food-borne mycotoxin aflatoxin B1 (AFB1) poses a significant risk to poultry, which are highly susceptible to its hepatotoxic effects. Domesticated turkeys (Meleagris gallopavo) are especially sensitive, whereas wild turkeys (M. g. silvestris) are more resistant. AFB1 toxicity entails bioactivation by hepatic cytochrome P450s to the electrophilic exo-AFB1-8,9-epoxide (AFBO). Domesticated turkeys lack functional hepatic GST-mediated detoxification of AFBO, and this is largely responsible for the differences in resistance between turkey types. This study was designed to characterize transcriptional changes induced in turkey livers by AFB1, and to contrast the response of domesticated (susceptible) and wild (more resistant) birds. Gene expression responses to AFB1 were examined using RNA-sequencing. Statistically significant differences in gene expression were observed among treatment groups and between turkey types. Expression analysis identified 4621 genes with significant differential expression (DE) in AFB1-treated birds compared to controls. Characterization of DE transcripts revealed genes dis-regulated in response to toxic insult with significant association of Phase I and Phase II genes and others important in cellular regulation, modulation of apoptosis, and inflammatory responses. Constitutive expression of GSTA3 was significantly higher in wild birds and was significantly higher in AFB1-treated birds when compared to controls for both genetic groups. This pattern was also observed by qRT-PCR in other wild and domesticated turkey strains. Results of this study emphasize the differential response of these genetically distinct birds, and identify genes and pathways that are differentially altered in aflatoxicosis.
Collapse
Affiliation(s)
- Kent M Reed
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA.
| | - Kristelle M Mendoza
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA.
| | - Juan E Abrahante
- University of Minnesota Informatics Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Roger A Coulombe
- Department of Animal, Dairy and Veterinary Sciences, College of Agriculture, Utah State University, Logan, UT 84322, USA.
| |
Collapse
|