1
|
Rani P, Koulmane Laxminarayana SL, Swaminathan SM, Nagaraju SP, Bhojaraja MV, Shetty S, Kanakalakshmi ST. TGF-β: elusive target in diabetic kidney disease. Ren Fail 2025; 47:2483990. [PMID: 40180324 PMCID: PMC11980245 DOI: 10.1080/0886022x.2025.2483990] [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: 08/21/2024] [Revised: 02/17/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Transforming growth factor-beta (TGF-β), a cytokine with near omnipresence, is an integral part of many vital cellular processes across the human body. The family includes three isoforms: Transforming growth factor-beta 1, 2, and 3. These cytokines play a significant role in the fibrosis cascade. Diabetic kidney disease (DKD), a major complication of diabetes, is increasing in prevalence daily, and the classical diagnosis of diabetes is based on the presence of albuminuria. The occurrence of nonalbuminuric DKD has provided new insight into the pathogenesis of this disease. The emphasis on multifactorial pathways involved in developing DKD has highlighted some markers associated with tissue fibrosis. In diabetic nephropathy, TGF-β is significantly involved in its pathology. Its presence in serum and urine means that it could be a diagnostic tool while its regulation provides potential therapeutic targets. Completely blocking TGF-β signaling could reach untargeted regions and cause unanticipated effects. This paper reviews the basic details of TGF-β as a cytokine, its role in DKD, and updates on research carried out to validate its candidacy.
Collapse
Affiliation(s)
- Priya Rani
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Shilna Muttickal Swaminathan
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Shankar Prasad Nagaraju
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Sahana Shetty
- Department of Endocrinology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | |
Collapse
|
2
|
Irqsusi M, Rodepeter FR, Günther M, Kirschbaum A, Vogt S. Matrix metalloproteinases and their tissue inhibitors as indicators of aortic aneurysm and dissection development in extracellular matrix remodeling. World J Exp Med 2025; 15:100166. [DOI: 10.5493/wjem.v15.i2.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 04/16/2025] Open
Abstract
Aneurysms and dissections represent some of the most serious cardiovascular diseases. The prevailing theory posits that mechanical overloading of the vessel wall is the underlying cause. Inspired by Barkhordarian et al, the authors present matrix metalloproteinases (MMPs) and their inhibitors in immunohistological analyses as contributing factors in the pathophysiology of aortic aneurysms (AA). Data analysis of MMP-1, MMP-9, tissue inhibitors of metalloproteinases (TIMPs), including TIMP-1 and TIMP-2 expression reveals a varied distribution between the adventitia and media and a non-uniform expression of the investigated markers. These elements, as key components of the extracellular matrix (ECM), indicate that the formation of AA is not solely driven by endoluminal pressure loading of the aortic wall. Instead, degenerative processes within ECM elements contribute significantly. Importantly, AA do not necessarily imply dissection. Tissue destruction, allowing blood flow entry, arises from reduced oxygen supply to the media, primarily due to incomplete capillarization or neocapillarization.
Collapse
Affiliation(s)
- Marc Irqsusi
- Department of Heart Surgery, Universitätsklinikum Marburg and Gießen GmbH, Marburg 35043, Hesse, Germany
| | - Fiona R Rodepeter
- Institute of Pathology, Philipps-University Marburg, Marburg 35043, Hesse, Germany
| | - Madeline Günther
- Department of Heart Surgery, Cardiovascular Research Laboratory, Philipps-University Marburg, Marburg 35043, Hesse, Germany
| | - Andreas Kirschbaum
- Department of Visceral Surgery, University Hospital Giessen and Marburg GmbH, Marburg 35043, Hesse, Germany
| | - Sebastian Vogt
- Department of Heart Surgery, Philipps-University Marburg, Marburg 35043, Hesse, Germany
| |
Collapse
|
3
|
Rocans RP, Zarins J, Bine E, Mahauri I, Deksnis R, Citovica M, Donina S, Gravelsina S, Vilmane A, Rasa-Dzelzkaleja S, Sabelnikovs O, Mamaja B. Early Postoperative Increase in Transforming Growth Factor Beta-1 Predicts Microvascular Flap Loss in Reconstructive Surgery: A Prospective Cohort Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:863. [PMID: 40428821 PMCID: PMC12113236 DOI: 10.3390/medicina61050863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/21/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Microvascular flap surgery is a widely used reconstructive technique for the repair of various defects. Biomarkers have become an essential tool for monitoring flap viability, early detection of complications, and prediction of surgical outcomes. Studies focusing on immunomodulatory cytokines in the early prediction of microvascular flap complications are lacking. We aimed to investigate the predictive value of postoperative changes in transforming growth factor beta-1 (TGF-β1) for microvascular flap complications. Materials and Methods: This prospective observational study comprised 44 adults scheduled for elective microvascular flap surgery. Preoperative blood samples for analysis were obtained before surgery, prior to the administration of intravenous fluids. Postoperative blood draws were collected after surgery, before leaving the operating room. Preoperative and postoperative serum concentrations of TGF-β1, as well as preoperative plasma albumin, total protein, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, full blood count, albumin, interleukin-6, C-reactive protein, and fibrinogen, were determined. Results: Postoperative changes in TGF-β1 were higher in cases with flap loss compared to patients with healthy recovery or patients with minor flap complications (0.403 log10 of ng/mL [0.024-0.782] vs. 0.157 [0.029-0.285] vs. -0.089 [-0.233-0.056], p = 0.002). Increased postoperative TGF-β1 was positively linked to preoperative C-reactive protein (p = 0.021), fibrinogen (p = 0.020), hematocrit (p = 0.039), and hemoglobin (p = 0.009). Conclusions: The postoperative increase in circulating TGF-β1 was associated with microvascular flap complications. Assessment of the postoperative changes in circulating TGF-β1 may be valuable for the early postoperative prediction of true flap loss.
Collapse
Affiliation(s)
- Rihards Peteris Rocans
- Intensive Care Clinic, Riga East Clinical University Hospital, Hipokrata Street 2, LV-1079 Riga, Latvia;
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| | - Janis Zarins
- Department of Hand and Plastic Surgery, Microsurgery Centre of Latvia, Brivibas Street 410, LV-1024 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Riga Technical University, Pulka Street 3, LV-1007 Riga, Latvia
| | - Evita Bine
- Intensive Care Clinic, Riga East Clinical University Hospital, Hipokrata Street 2, LV-1079 Riga, Latvia;
| | - Insana Mahauri
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| | - Renars Deksnis
- Surgical Oncology Clinic, Riga East Clinical University Hospital, Hipokrata Street 4, LV-1079 Riga, Latvia;
| | - Margarita Citovica
- Laboratory Department, Riga East Clinical University Hospital, Hipokrata Street 2, LV-1079 Riga, Latvia;
| | - Simona Donina
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
- Outpatient Department, Riga East Clinical University Hospital, Hipokrata Street 4, LV-1079 Riga, Latvia
| | - Sabine Gravelsina
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
| | - Anda Vilmane
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
| | - Santa Rasa-Dzelzkaleja
- Institute of Microbiology and Virology, Rīga Stradiņš University, Ratsupites Street 5, LV-1067 Riga, Latvia; (S.D.); (S.G.); (S.R.-D.)
| | - Olegs Sabelnikovs
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| | - Biruta Mamaja
- Department of Anaesthesia and Intensive Care, Riga Stradiņš University, Dzirciema Street 16, LV-1007 Riga, Latvia; (I.M.); (O.S.); (B.M.)
| |
Collapse
|
4
|
Ungefroren H, Randeva H, Lehnert H, Schrader J, Marquardt JU, Konukiewitz B, Hass R. Crosstalk of TGF-β and somatostatin signaling in adenocarcinoma and neuroendocrine tumors of the pancreas: a brief review. Front Endocrinol (Lausanne) 2025; 16:1511348. [PMID: 40134804 PMCID: PMC11934628 DOI: 10.3389/fendo.2025.1511348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/03/2025] [Indexed: 03/27/2025] Open
Abstract
Although the vast majority of cancers affecting the human pancreas are pancreatic ductal adenocarcinomas (PDAC), there are several other cancer types originating from non-exocrine cells of this organ, i.e., pancreatic neuroendocrine tumors (panNET). Genomic analyses of PDAC and panNET revealed that certain signaling pathways such as those triggered by transforming growth factor-β (TGF-β) are frequently altered, highlighting their crucial role in pancreatic tumor development. In PDAC, TGF-β plays a dual role acting as a tumor suppressor in healthy tissue and early stages of tumor development but as a promoter of tumor progression in later stages. This peptide growth factor acts as a potent inducer of epithelial-to-mesenchymal transition (EMT), a developmental program that transforms otherwise stationary epithelial cells to invasive mesenchymal cells with enhanced metastatic potential. TGF-β signals through both the canonical Smad pathway involving the receptor-regulated Smad proteins, SMAD2 and SMAD3, and the common-mediator Smad, SMAD4, as well as Smad-independent pathways, i.e., ERK1/2, PI3K/AKT, and somatostatin (SST). Accumulating evidence indicates an intimate crosstalk between TGF-β and SST signaling, not only in PDAC but, more recently, also in panNET. In this work, we review the available evidence on signaling interactions between both pathways, which we believe are of potential but as yet insufficiently appreciated importance for pancreatic cancer development and/or progression as well as novel therapeutic approaches.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- Institute of Pathology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
| | - Harpal Randeva
- University Hospital of Coventry and Warwickshire (UHCW) and Warwick Medical School, Coventry, United Kingdom
| | - Hendrik Lehnert
- University Hospital of Coventry and Warwickshire (UHCW) and Warwick Medical School, Coventry, United Kingdom
| | - Jörg Schrader
- First Department of Medicine, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jens-Uwe Marquardt
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, Lübeck, Germany
| | - Björn Konukiewitz
- Institute of Pathology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Colemon A, Romney CV, Jones AD, Bagsby C, Jackson R, Ramanathan S. Interplay Between TGFβ1 Signaling and Cancer-Testis Antigen MAGEB2: A New Thorn in Cancer's Side? Int J Mol Sci 2025; 26:2448. [PMID: 40141091 PMCID: PMC11942090 DOI: 10.3390/ijms26062448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
The Melanoma Antigen Gene (MAGE) family of proteins is the largest family of cancer-testis antigens (CTAs) and shares a MAGE homology domain (MHD). MAGE proteins are divided into Type I and Type II MAGEs depending on their chromosomal location and expression patterns. Type I MAGEs are true CTAs. MAGEB2 is a Type I MAGE, belonging to the MAGEB subfamily, and unlike some MAGE proteins, has not been found to bind to and enhance E3 ligase activity. MAGEB2 has been discovered to be an RNA-binding protein that serves to protect spermatogonial cells in the testis from extraneous stressors. We have discovered that MAGEB2 is necessary and sufficient for the proliferation of cells and is expressed by the differential DNA methylation of its gene promoter. Furthermore, we identified JunD as the transcription factor that regulates MAGEB2 expression. When expressed, MAGEB2 suppresses transforming grown factor-β1 (TGFβ1) signaling by decreasing mRNA levels of Thrombospondin-1 (TSP-1). TSP-1 is an anti-angiogenic protein that activates TGFβ1. Restoring levels of TSP-1 or TGFβ1 results in the inability of MAGEB2 to drive proliferation, suggesting that MAGEB2-expressing tumors might be more susceptible to therapies that induce or activate TSP-1 or TGFβ1 signaling.
Collapse
Affiliation(s)
- Ashley Colemon
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37208, USA
- Fisk-Vanderbilt Master’s-to-Ph.D. Bridge Program, Nashville, TN 37208, USA
| | - Carlan V. Romney
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37208, USA
- Fisk-Vanderbilt Master’s-to-Ph.D. Bridge Program, Nashville, TN 37208, USA
| | - Angelle D. Jones
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Clarke Bagsby
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Richala Jackson
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37208, USA
| | - Saumya Ramanathan
- Department of Life and Physical Sciences, Fisk University, Nashville, TN 37208, USA
- Fisk-Vanderbilt Master’s-to-Ph.D. Bridge Program, Nashville, TN 37208, USA
| |
Collapse
|
6
|
Wang T, Maldonado CC, Huang BL, Budbazar E, Martin A, Layne MD, Murphy-Ullrich JE, Grinstaff MW, Albro MB. A Bio-inspired Latent TGF-β Conjugated Scaffold Improves Neocartilage Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636279. [PMID: 39975171 PMCID: PMC11838498 DOI: 10.1101/2025.02.03.636279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
In cartilage tissue engineering, active TGF-β is conventionally supplemented in culture medium at highly supraphysiologic doses to accelerate neocartilage development. While this approach enhances cartilage extracellular matrix (ECM) biosynthesis, it further promotes tissue features detrimental to hyaline cartilage function, including the induction of tissue swelling, hyperplasia, hypertrophy, and ECM heterogeneities. In contrast, during native cartilage development, chondrocytes are surrounded by TGF-β configured in a latent complex (LTGF-β), which undergoes cell-mediated activation, giving rise to moderated, physiologic dosing regimens that enhance ECM biosynthesis while avoiding detrimental features associated with TGF-β excesses. Here, we explore a bio-inspired strategy, consisting of LTGF-β-conjugated scaffolds, providing TGF-β exposure regimens that are moderated and uniformly administered throughout the construct. Specifically, we evaluate the performance of LTGF-β scaffolds to improve neocartilage development with bovine chondrocyte-seeded agarose constructs compared to outcomes from active TGF-β media supplementation (MS) at a physiologic 0.3 ng/mL dose (MS-0.3), supraphysiologic 10 ng/mL dose (MS-10), or TGF-β free. For small-size constructs (∅3×2 mm), LTGF-β scaffolds yield neocartilage that achieves native-matched mechanical properties (800-925 kPa) and sGAG content (6.6%-7.1%), while providing a cell morphology and collagen distribution more reminiscent of hyaline cartilage. LTGF-β scaffolds further afford an optimal chondrogenic phenotype, marked by a 12-to 28-fold reduction of COL-I expression relative to TGF-β-free and a 7-to 17-fold reduction of COL-X expression relative to MS-10. Further, for large-size constructs, which approach the dimensions needed for clinical cartilage repair, LTGF-β scaffolds significantly reduce mechanical and biochemical heterogeneities relative to MS-0.3 and MS-10. Overall, the use of LTGF-β scaffolds improves the composition, structure, material properties, and cell phenotype of neocartilage.
Collapse
|
7
|
Xiao Y, Wang Y, Zhang M, Zhang Y, Ju Z, Wang J, Zhang Y, Yang C, Wang X, Jiang Q, Gao Y, Wei X, Liu W, Gao Y, Hu P, Huang J. Tankyrase inhibitor IWR-1 modulates HIPPO and Transforming Growth Factor β signaling in primed bovine embryonic stem cells cultured on mouse embryonic fibroblasts. Theriogenology 2025; 233:100-111. [PMID: 39613494 DOI: 10.1016/j.theriogenology.2024.11.010] [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: 08/25/2024] [Revised: 11/17/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
The use of tankyrase inhibitors is essential for capturing livestock embryonic stem cells (ESC), yet their mechanisms of action remain largely uncharacterized. Previous studies indicate that their roles extend beyond the suppression of canonical WNT signaling. This study investigates the effects of the tankyrase inhibitor IWR-1 on maintaining the pluripotency of bovine embryonic stem cells (bESC) cultured on mitotically inactivated mouse embryonic fibroblasts (MEF). Notably, bESC exhibited significant differentiation after one month of IWR-1 withdrawal, without a clear bias toward any specific germ layer. IWR-1 effectively inhibited TNKS2 activity in bESC, whereas it suppressed TNKS1 protein level in growth-arrested MEF. Early differentiation upon IWR-1 removal induced more substantial transcriptomic changes in MEF than in bESC. Furthermore, cell communication analysis predicted that IWR-1 influenced several paracrine and autocrine signals within the culture system. We also observed that IWR-1 repressed protein abundance of the HIPPO pathway components including TEAD4 and YAP1 in bESC and decreased transcription of HIPPO targeted genes CYR61. Protein analysis in growth-arrested MEF suggested that IWR-1 modulated MEF function by impeding TGF-β1 activation and activin A secretion which mitigated nuclear localization of SMAD2/3 in the bESC. This study underscores the role of tankyrase inhibitors in ESC self-renewal by modulating key signaling pathways and orchestrating cell-cell interactions, which may be meaningful in understanding the delicate signaling control of pluripotency in livestock and improving the culture system.
Collapse
Affiliation(s)
- Yao Xiao
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China
| | - Yujie Wang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; College of Life Sciences, Shandong Normal University, Jinan, 250358, China
| | - Minghao Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yan Zhang
- Key Laboratory of Efficient Dairy Cattle Propagation and Germplasm Innovation of Ministry of Agriculture and Rural Affairs, Shandong OX Livestock Breeding Co., Ltd, Jinan, 250100, China
| | - Zhihua Ju
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Jinpeng Wang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Yaran Zhang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Chunhong Yang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Xiuge Wang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Qiang Jiang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Yaping Gao
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Xiaochao Wei
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Wenhao Liu
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China
| | - Yundong Gao
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China; Key Laboratory of Efficient Dairy Cattle Propagation and Germplasm Innovation of Ministry of Agriculture and Rural Affairs, Shandong OX Livestock Breeding Co., Ltd, Jinan, 250100, China
| | - Peng Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jinming Huang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China; College of Life Sciences, Shandong Normal University, Jinan, 250358, China.
| |
Collapse
|
8
|
Niu L, Liu J, Xu H, Liu B, Song M, Hu C, Jiang R, Sun X, Lei Y. Exposure to particulate matter (PM 2.5) weakens corneal defense by downregulating thrombospondin-1 and tight junction proteins. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117276. [PMID: 39536561 DOI: 10.1016/j.ecoenv.2024.117276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Fine particulate matter (PM2.5) induces ocular surface toxicity through pyroptosis, oxidative stress, autophagy, and inflammatory responses. However, the precise molecular pathways through which PM2.5 causes corneal damage remain unclear. This study aims to investigate the underlying mechanisms by exposing human corneal epithelial cells (HCECs) to PM2.5. METHODS After the morphology and chemical composition analysis of the PM samples, we conducted both in vivo and in vitro experiments to investigate PM2.5-induced corneal epithelial damage. We assessed corneal barrier function in HCECs using transepithelial electrical resistance (TEER) assays. To explore the molecular mechanisms of PM2.5-induced corneal epithelial damage, we performed whole-transcriptome resequencing, quantitative RT-PCR, and western blotting in vitro. In addition, we analyzed mouse corneas exposed to concentrated ambient PM2.5 through immunofluorescence staining to observe the resulting changes in corneal epithelial protein expression in vivo. RESULTS Our results showed significant impairment of corneal epithelial barrier function in PM2.5-treated HCECs, as indicated by decreased TEER values. The expression of thrombospondin-1 (THBS1) and claudin-1, both key factors for maintaining corneal epithelial barrier integrity, was markedly reduced at the gene and protein levels in both in vitro and in vivo PM2.5 exposure models. Moreover, the levels of tight junction-associated proteins, including occludin, zonula occludens-1 (ZO-1) and ZO-2, essential components of the corneal epithelial barrier, were significantly diminished in PM2.5-treated HCECs. CONCLUSION PM2.5 exposure leads to corneal epithelium damage by disrupting tight junction proteins and THBS1 expression. These findings provide insight into potential pathways for PM2.5-induced ocular toxicity and underscore the need for protective strategies against such environmental pollutants.
Collapse
Affiliation(s)
- Liangliang Niu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China; Ocular Trauma Center, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Jiamin Liu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Huan Xu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China; Ocular Trauma Center, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Binghui Liu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Maomao Song
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
| | - Chunchun Hu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China; Ocular Trauma Center, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Rui Jiang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China; Ocular Trauma Center, Eye & ENT Hospital, Fudan University, Shanghai 200031, China.
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Yuan Lei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China.
| |
Collapse
|
9
|
Han DH, Shin MK, Sung JS, Kim M. miR-335-3p attenuates transforming growth factor beta 1-induced fibrosis by suppressing Thrombospondin 1. PLoS One 2024; 19:e0311594. [PMID: 39374214 PMCID: PMC11457990 DOI: 10.1371/journal.pone.0311594] [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: 05/21/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
Pulmonary fibrosis is characterized by excessive extracellular matrix (ECM) accumulation caused by detrimental stimuli. The progressive impairment in lung functions is chronic and highly fatal, presenting itself as a global health challenge. Because of the lack of efficacious treatments, the underlying mechanism should be investigated. The progression of fibrosis involves transforming growth factor-beta 1 (TGF-β1), which accelerates ECM production via epithelial-mesenchymal transition and cell invasion. As microRNAs (miRNAs) serve as regulators of disease development and progression, this study aimed to investigate the interaction of miRNAs and target genes that could contribute to pulmonary fibrosis when exposed to TGF-β1. Differentially expressed mRNA and miRNA were identified in respiratory epithelial cells via transcriptome analysis by using the constructed TGF-β1-induced fibrosis model. Our results revealed a significant increase in the expression of thrombospondin 1 (THBS1), which participates in TGF-β1 activation, where THBS1 was identified as a core gene in protein interactions analyzed through bioinformatics. The expression of miR-335-3p, which targets 3'-UTR of THBS1, substantially decreased upon TGF-β1 treatment. The TGF-β1 downstream signal was suppressed by inhibiting the interaction between TGF-β1 and THBS1, consequently alleviating fibrosis. When the miR-335-3p mimic was transfected in TGF-β1-treated respiratory epithelial cells, THBS1 and fibrosis markers were downregulated, while the introduction of miR-335-3p inhibitor exhibited a reverse phenomenon. Our findings demonstrated that TGF-β1 exposure to respiratory epithelial cells led to a decrease in miR-335-3p expression, resulting in the upregulation of THBS1 and ultimately exacerbating fibrosis. This study provides insights into TGF-β1-induced pulmonary fibrosis, suggesting new therapeutic targets and mechanisms.
Collapse
Affiliation(s)
- Dong-Hee Han
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, Korea
| | - Min Kyoung Shin
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, Korea
| | - Jung-Suk Sung
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, Korea
| | - Min Kim
- Department of Life Science, Biomedi Campus, Dongguk University-Seoul, Goyang-si, Gyeonggi-do, Korea
| |
Collapse
|
10
|
Hanč P, von Andrian UH. No pain, no gain - how nociceptors orchestrate tissue repair. Cell Res 2024; 34:673-674. [PMID: 38783113 PMCID: PMC11442612 DOI: 10.1038/s41422-024-00979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Affiliation(s)
- Pavel Hanč
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
- The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
11
|
Zelisko N, Lesyk R, Stoika R. Structure, unique biological properties, and mechanisms of action of transforming growth factor β. Bioorg Chem 2024; 150:107611. [PMID: 38964148 DOI: 10.1016/j.bioorg.2024.107611] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/07/2024] [Accepted: 06/30/2024] [Indexed: 07/06/2024]
Abstract
Transforming growth factor β (TGF-β) is a ubiquitous molecule that is extremely conserved structurally and plays a systemic role in human organism. TGF-β is a homodimeric molecule consisting of two subunits joined through a disulphide bond. In mammals, three genes code for TGF-β1, TGF-β2, and TGF-β3 isoforms of this cytokine with a dominating expression of TGF-β1. Virtually, all normal cells contain TGF-β and its specific receptors. Considering the exceptional role of fine balance played by the TGF-β in anumber of physiological and pathological processes in human body, this cytokine may be proposed for use in medicine as an immunosuppressant in transplantology, wound healing and bone repair. TGFb itself is an important target in oncology. Strategies for blocking members of TGF-β signaling pathway as therapeutic targets have been considered. In this review, signalling mechanisms of TGF-β1 action are addressed, and their role in physiology and pathology with main focus on carcinogenesis are described.
Collapse
Affiliation(s)
- Nataliya Zelisko
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine.
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine
| |
Collapse
|
12
|
Zhou T, Yang H, Assa C, DeRoo E, Bontekoe J, Burkel B, Ponik S, Lu HS, Daugherty A, Liu B. Myeloid-Specific Thrombospondin-1 Deficiency Exacerbates Aortic Rupture via Broad Suppression of Extracellular Matrix Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605216. [PMID: 39211130 PMCID: PMC11361016 DOI: 10.1101/2024.07.30.605216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Rationale Rupture of abdominal aortic aneurysms (AAA) is associated with high mortality. However, the precise molecular and cellular drivers of AAA rupture remain elusive. Our prior study showed that global and myeloid-specific deletion of matricellular protein thrombospondin-1 (TSP1) protects mice from aneurysm formation primarily by inhibiting vascular inflammation. Objective To investigate the cellular and molecular mechanisms that drive AAA rupture by testing how TSP1 deficiency in different cell populations affects the rupture event. Methods and Results We deleted TSP1 in endothelial cells and macrophages --- the major TSP1-expressing cells in aneurysmal tissues ---- by crossbreeding Thbs1 flox/flox mice with VE-cadherin Cre and Lyz2-cre mice, respectively. Aortic aneurysm and rupture were induced by angiotensin II in mice with hypercholesterolemia. Myeloid-specific Thbs1 knockout, but not endothelial-specific knockout, increased the rate of lethal aortic rupture by more than 2 folds. Combined analyses of single-cell RNA sequencing and histology showed a unique cellular and molecular signature of the rupture-prone aorta that was characterized by a broad suppression in inflammation and extracellular matrix production. Visium spatial transcriptomic analysis on human AAA tissues showed a correlation between low TSP1 expression and aortic dissection. Conclusions TSP1 expression by myeloid cells negatively regulates aneurysm rupture, likely through promoting the matrix repair phenotypes of vascular smooth muscle cells thereby increasing the strength of the vascular wall.
Collapse
|
13
|
Kareem DS, Al-Ameri LMH, Alalawi AS. Impact of Photobiomodulation on IL1β and TGFβ-1 concentrations in patients with aphthous stomatitis. Lasers Med Sci 2024; 39:176. [PMID: 38976032 DOI: 10.1007/s10103-024-04130-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Laser therapy has shown effectiveness in promoting wound healing by influencing various physiological factors such as blood flow, cytokines, histamine, nerve signals, lymphocyte function, tissue oxygenation, and cell growth. This study aims to evaluate the therapeutic efficacy of Photobiomodulation (PBM) treatment, by using diode laser, in modifying the levels of interleukin-1 beta (IL1β) and transforming growth factor beta-1 (TGFβ-1) in patients diagnosed with aphthous stomatitis. A before-after interventional design was conducted over 10 months with 20 subjects. Data on demographic details and serum concentrations of IL1β and TGFβ-1 were collected pre-treatment and on Days 3 and 7 post-treatments. The intervention involved a single session of four 30-second applications of a QuickLase dual-wavelength laser operating at 980 nm. Results show significant reductions in IL1β and TGFβ-1 levels after 7 days of treatment, indicating a time-dependent effect of PBM therapy on these inflammatory markers. The findings suggest that PBM therapy holds promise as an intervention for reducing inflammation associated with aphthous stomatitis.
Collapse
Affiliation(s)
- Dalia Saleem Kareem
- Medical and Biological applications Branch Institute of Laser for Postgraduate Studies, University of Baghdad, Baghdad, Iraq.
| | - Layla M H Al-Ameri
- Medical and Biological applications Branch Institute of Laser for Postgraduate Studies, University of Baghdad, Baghdad, Iraq
| | - Ammar Saleh Alalawi
- Laser/Dentistry (oral surgery), Al-Emamein Al-Kadhimein Madical City, Baghdad, Iraq
| |
Collapse
|
14
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
15
|
Dressler FF, Diedrichs F, Sabtan D, Hinrichs S, Krisp C, Gemoll T, Hennig M, Mackedanz P, Schlotfeldt M, Voß H, Offermann A, Kirfel J, Roesch MC, Struck JP, Kramer MW, Merseburger AS, Gratzke C, Schoeb DS, Miernik A, Schlüter H, Wetterauer U, Zubarev R, Perner S, Wolf P, Végvári Á. Proteomic analysis of the urothelial cancer landscape. Nat Commun 2024; 15:4513. [PMID: 38802361 PMCID: PMC11130393 DOI: 10.1038/s41467-024-48096-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Urothelial bladder cancer (UC) has a wide tumor biological spectrum with challenging prognostic stratification and relevant therapy-associated morbidity. Most molecular classifications relate only indirectly to the therapeutically relevant protein level. We improve the pre-analytics of clinical samples for proteome analyses and characterize a cohort of 434 samples with 242 tumors and 192 paired normal mucosae covering the full range of UC. We evaluate sample-wise tumor specificity and rank biomarkers by target relevance. We identify robust proteomic subtypes with prognostic information independent from histopathological groups. In silico drug prediction suggests efficacy of several compounds hitherto not in clinical use. Both in silico and in vitro data indicate predictive value of the proteomic clusters for these drugs. We underline that proteomics is relevant for personalized oncology and provide abundance and tumor specificity data for a large part of the UC proteome ( www.cancerproteins.org ).
Collapse
Affiliation(s)
- Franz F Dressler
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| | - Falk Diedrichs
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Deema Sabtan
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sofie Hinrichs
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Christoph Krisp
- Section Mass Spectrometry and Proteomics, Campus Forschung N27 00.008, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Gemoll
- Section for Translational Surgical Oncology and Biobanking, Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Martin Hennig
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Paulina Mackedanz
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Mareile Schlotfeldt
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Hannah Voß
- Section Mass Spectrometry and Proteomics, Campus Forschung N27 00.008, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Offermann
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Jutta Kirfel
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Marie C Roesch
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Julian P Struck
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Department of Urology, Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Brandenburg, Germany
| | - Mario W Kramer
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dominik S Schoeb
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Arkadiusz Miernik
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, Campus Forschung N27 00.008, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Wetterauer
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, 3500, Krems, Austria
| | - Roman Zubarev
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- The National Medical Research Center for Endocrinology, Moscow, Russia
- Department of Pharmacological & Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sven Perner
- Institute of Pathology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Institute of Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- Center for Precision Oncology, Tuebingen, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Julovi SM, Trinh K, Robertson H, Xu C, Minhas N, Viswanathan S, Patrick E, Horowitz JD, Meijles DN, Rogers NM. Thrombospondin-1 Drives Cardiac Remodeling in Chronic Kidney Disease. JACC Basic Transl Sci 2024; 9:607-627. [PMID: 38984053 PMCID: PMC11228122 DOI: 10.1016/j.jacbts.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 07/11/2024]
Abstract
Patients with chronic kidney disease (CKD) face a high risk of cardiovascular disease. Previous studies reported that endogenous thrombospondin 1 (TSP1) involves right ventricular remodeling and dysfunction. Here we show that a murine model of CKD increased myocardial TSP1 expression and produced left ventricular hypertrophy, fibrosis, and dysfunction. TSP1 knockout mice were protected from these features. In vitro, indoxyl sulfate is driving deleterious changes in cardiomyocyte through the TSP1. In patients with CKD, TSP1 and aryl hydrocarbon receptor were both differentially expressed in the myocardium. Our findings summon large clinical studies to confirm the translational role of TSP1 in patients with CKD.
Collapse
Affiliation(s)
- Sohel M Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Harry Robertson
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
| | - Cuicui Xu
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Nikita Minhas
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Seethalakshmi Viswanathan
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Ellis Patrick
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
- School of Mathematics, University of Sydney, New South Wales, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong Special Administrative Region, China
| | - John D Horowitz
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
- Cardiovascular Pathophysiology and Therapeutics Research Group, Basil Hetzel Institute, Woodville, South Australia, Australia
- Department of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Natasha M Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Renal and Transplantation Unit, Westmead Hospital, New South Wales, Australia
| |
Collapse
|
17
|
Wawrzykowski J, Jamioł M, Kankofer M. A pilot study on the relationship between thrombospondin-1 (THBS1) and transforming growth factor beta1 (TGFβ1) in the bovine placenta during early mid-pregnancy as well as parturition with normally released and retained placenta. Mol Reprod Dev 2024; 91:e23710. [PMID: 37811864 DOI: 10.1002/mrd.23710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023]
Abstract
During pregnancy, it is necessary to create appropriate conditions for the development of the placenta and the fetus. However, during parturition, the placenta must be separated and subsequently removed as soon as possible to not expose the female to the possibility of infection. In this study, the relationship between thrombospondin-1 (THBS1) and transforming growth factor beta1 (TGFβ1) concentrations was described during bovine pregnancy (second, fourth, and sixth months; n = 3/each month), at normal parturition (NR) and parturition with fetal membrane retention (R). The presence of THBS1 and TGFβ1 was confirmed in bovine placental tissues of both maternal and fetal parts. Enzyme-linked immunosorbent assay showed statistically significant differences (p < 0.05) in THBS1 concentrations (pg/mg protein) between examined parturient samples (maternal part: 5.76 ± 1.61 in R vs. 2.26 ± 1.58 in NR; fetal part: 2.62 ± 1.94 in R vs. 1.70 ± 0.23 in NR). TGFβ1 concentrations (pg/mg protein) were significantly lower (p < 0.05) in the retained fetal membranes compared to the released fetal membranes in the maternal part of the placenta (26.22 ± 7.53 in NR vs. 17.80 ± 5.01 in R). The participation of THBS1 in the activation of TGFβ1 in parturient bovine placental tissues leading to the normal release of fetal membranes may be suggested.
Collapse
Affiliation(s)
- Jacek Wawrzykowski
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Science in Lublin, Lublin, Poland
| | - Monika Jamioł
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Science in Lublin, Lublin, Poland
| | - Marta Kankofer
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Science in Lublin, Lublin, Poland
| |
Collapse
|
18
|
Huang D, Liang J, Yang J, Yang C, Wang X, Dai T, Steinberg T, Li C, Wang F. Current Status of Tissue Regenerative Engineering for the Treatment of Uterine Infertility. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:558-573. [PMID: 37335062 DOI: 10.1089/ten.teb.2022.0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
With the recent developments in tissue engineering, scientists have attempted to establish seed cells from different sources, create cell sheets through various technologies, implant them on scaffolds with various spatial structures, or load scaffolds with cytokines. These research results are very optimistic, bringing hope to the treatment of patients with uterine infertility. In this article, we reviewed articles related to the treatment of uterine infertility from the aspects of experimental treatment strategy, seed cells, scaffold application, and repair criteria so as to provide a basis for future research.
Collapse
Affiliation(s)
- Di Huang
- Shandong First Medical University, Jinan, China
| | - Junhui Liang
- Departments of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Yang
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Chunrun Yang
- Departments of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Ultrasonography, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tianyu Dai
- Shandong First Medical University, Jinan, China
| | - Thorsten Steinberg
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Changzhong Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Wang
- Departments of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
19
|
Burioli EAV, Hammel M, Vignal E, Vidal-Dupiol J, Mitta G, Thomas F, Bierne N, Destoumieux-Garzón D, Charrière GM. Transcriptomics of mussel transmissible cancer MtrBTN2 suggests accumulation of multiple cancer traits and oncogenic pathways shared among bilaterians. Open Biol 2023; 13:230259. [PMID: 37816387 PMCID: PMC10564563 DOI: 10.1098/rsob.230259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Transmissible cancer cell lines are rare biological entities giving rise to diseases at the crossroads of cancer and parasitic diseases. These malignant cells have acquired the amazing capacity to spread from host to host. They have been described only in dogs, Tasmanian devils and marine bivalves. The Mytilus trossulus bivalve transmissible neoplasia 2 (MtrBTN2) lineage has even acquired the capacity to spread inter-specifically between marine mussels of the Mytilus edulis complex worldwide. To identify the oncogenic processes underpinning the biology of these atypical cancers we performed transcriptomics of MtrBTN2 cells. Differential expression, enrichment, protein-protein interaction network, and targeted analyses were used. Overall, our results suggest the accumulation of multiple cancerous traits that may be linked to the long-term evolution of MtrBTN2. We also highlight that vertebrate and lophotrochozoan cancers could share a large panel of common drivers, which supports the hypothesis of an ancient origin of oncogenic processes in bilaterians.
Collapse
Affiliation(s)
- E A V Burioli
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - M Hammel
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
- ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - E Vignal
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - J Vidal-Dupiol
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - G Mitta
- IFREMER, UMR 241 Écosystèmes Insulaires Océaniens, Labex Corail, Centre Ifremer du Pacifique, Tahiti, Polynésie française
| | - F Thomas
- CREEC/CANECEV (CREES), MIVEGEC, Unité Mixte de Recherches, IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France
| | - N Bierne
- ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - D Destoumieux-Garzón
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| | - G M Charrière
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Montpellier, France
| |
Collapse
|
20
|
Agarwal R, Iezhitsa I. Advances in targeting the extracellular matrix for glaucoma therapy: current updates. Expert Opin Ther Targets 2023; 27:1217-1229. [PMID: 38069479 DOI: 10.1080/14728222.2023.2293748] [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: 10/11/2023] [Accepted: 12/07/2023] [Indexed: 12/31/2023]
Abstract
INTRODUCTION Elevated intraocular pressure (IOP) is a well-recognized risk factor for development of primary open angle glaucoma (POAG), a leading cause of irreversible blindness. Ocular hypertension is associated with excessive extracellular matrix (ECM) deposition in trabecular meshwork (TM) resulting in increased aqueous outflow resistance and elevated IOP. Hence, therapeutic options targeting ECM remodeling in TM to lower IOP in glaucomatous eyes are of considerable importance. AREAS COVERED This paper discusses the complex process of ECM regulation in TM and explores promising therapeutic targets. The role of Transforming Growth Factor-β as a central player in ECM deposition in TM is discussed. We elaborate the key regulatory processes involved in its activation, release, signaling, and cross talk with other signaling pathways including Rho GTPase, Wnt, integrin, cytokines, and renin-angiotensin-aldosterone. Further, we summarize the therapeutic agents that have been explored to target ECM dysregulation in TM. EXPERT OPINION Targeting molecular pathways to reduce ECM deposition and/or enhance its degradation are of considerable significance for IOP lowering. Challenges lie in pinpointing specific targets and designing drug delivery systems to precisely interact with pathologically active/inactive signaling. Recent advances in monoclonal antibodies, fusion molecules, and vectored nanotechnology offer potential solutions.
Collapse
Affiliation(s)
- Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Igor Iezhitsa
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Pirkkanen J, Tharmalingam S, Thome C, Sinex HC, Benjamin LV, Losch AC, Borgmann AJ, Dhaemers RM, Gordon C, Boreham DR, Mendonca MS. Genomic Loss and Epigenetic Silencing of the FOSL1 Tumor Suppressor Gene in Radiation-induced Neoplastic Transformation of Human CGL1 Cells Alters the Tumorigenic Phenotype In Vitro and In Vivo. Radiat Res 2023; 200:48-64. [PMID: 37141110 PMCID: PMC10409446 DOI: 10.1667/rade-22-00216.1] [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: 12/06/2022] [Accepted: 03/24/2023] [Indexed: 05/05/2023]
Abstract
The CGL1 human hybrid cell system has been utilized for many decades as an excellent cellular tool for investigating neoplastic transformation. Substantial work has been done previously implicating genetic factors related to chromosome 11 to the alteration of tumorigenic phenotype in CGL1 cells. This includes candidate tumor suppressor gene FOSL1, a member of the AP-1 transcription factor complex which encodes for protein FRA1. Here we present novel evidence supporting the role of FOSL1 in the suppression of tumorigenicity in segregants of the CGL1 system. Gamma-induced mutant (GIM) and control (CON) cells were isolated from 7 Gy gamma-irradiated CGL1s. Western, Southern and Northern blot analysis were utilized to assess FOSL1/FRA1 expression as well as methylation studies. GIMs were transfected to re-express FRA1 and in vivo tumorigenicity studies were conducted. Global transcriptomic microarray and RT-qPCR analysis were used to further characterize these unique cell segregants. GIMs were found to be tumorigenic in vivo when injected into nude mice whereas CON cells were not. GIMs show loss of Fosl/FRA1 expression as confirmed by Western blot. Southern and Northern blot analysis further reveals that FRA1 reduction in tumorigenic CGL1 segregants is likely due to transcriptional suppression. Results suggest that radiation-induced neoplastic transformation of CGL1 is in part due to silencing of the FOSL1 tumor suppressor gene promoter by methylation. The radiation-induced tumorigenic GIMs transfected to re-express FRA1 resulted in suppression of subcutaneous tumor growth in nude mice in vivo. Global microarray analysis and RT-qPCR validation elucidated several hundred differentially expressed genes. Downstream analysis reveals a significant number of altered pathways and enriched Gene Ontology terms genes related to cellular adhesion, proliferation, and migration. Together these findings provide strong evidence that FRA1 is a tumor suppressor gene deleted and epigenetically silenced after ionizing radiation-induced neoplastic transformation in the CGL1 human hybrid cell system.
Collapse
Affiliation(s)
- Jake Pirkkanen
- Department of Biology, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
| | - Sujeenthar Tharmalingam
- Department of Biology, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, Ontario, P3E 2C6, Canada
| | - Christopher Thome
- Department of Biology, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
- Medical Sciences Division, NOSM University, Sudbury, Ontario, P3E 2C6, Canada
| | - Helen Chin Sinex
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Laura V. Benjamin
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Adam C. Losch
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Anthony J. Borgmann
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Ryan M. Dhaemers
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Christopher Gordon
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Douglas R. Boreham
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, P3E 2C6, Canada
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Bruce Power, PO Box 1540, 177 Tie Rd, R.R. 2, Tiverton, Ontario, N0G 2T0, Canada
- Nuclear Innovation Institute, Port Elgin, Ontario, N0H 2C0, Canada
| | - Marc S. Mendonca
- Department of Radiation Oncology, Radiation and Cancer Biology Laboratories, and Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
22
|
Hayashi KG, Sakumoto R. Differential expression of pro- and anti-angiogenic factors in the endometrium between repeat breeder and normally fertile cows. Anim Reprod Sci 2023; 254:107265. [PMID: 37270879 DOI: 10.1016/j.anireprosci.2023.107265] [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: 02/16/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/06/2023]
Abstract
In cattle, the establishment of appropriate endometrial vasculature during the estrous cycle is required for preparing a receptive endometrium. This study aimed to investigate 1) mRNA expression of potent pro- and anti-angiogenic factors, 2) protein localization of the anti-angiogenic factor thrombospondin (TSP), and 3) vascularity in the endometrium of repeat breeder (RB) and normally fertile (non-RB) cows. Caruncular and intercaruncular endometrium was collected from RB and non-RB cows during the luteal phase of the estrous cycle. RB cows had greater mRNA expression levels of TSP ligands (TSP1 and TSP2) and receptors (CD36 and CD47) than non-RB cows. Although the mRNA expression levels of most angiogenic factors did not change by repeat breeding, RB cows had greater mRNA expression of fibroblast growth factor receptor 1 (FGFR1), angiopoietin 1 (ANGPT1), and ANGPT2 and a less mRNA expression of vascular endothelial growth factor B (VEGFB) than non-RB cows. By immunohistochemistry, TSP1, TSP2, CD36, and CD47 were detected in the luminal epithelium, glandular epithelium, stromal cells, and blood vessels of the endometrium. Two indexes of vascularity, the number of blood vessels and the percentage of area stained positive for the von Willebrand factor, were lower in the endometrium of RB than in that of non-RB cows. These results demonstrate that RB cows have a greater expression of both ligands and receptors for the anti-angiogenic factor TSP and a reduced vascular distribution in the endometrium compared with non-RB cows, suggesting suppressed endometrial angiogenesis.
Collapse
Affiliation(s)
- Ken-Go Hayashi
- Division of Advanced Feeding Technology Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba 305-0901, Japan.
| | - Ryosuke Sakumoto
- Division of Advanced Feeding Technology Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba 305-0901, Japan.
| |
Collapse
|
23
|
Nishitsuji K, Ikezaki M, Manabe S, Ihara Y. Functions of Protein <i>C</i>-Mannosylation in Physiology and Pathology. TRENDS GLYCOSCI GLYC 2023. [DOI: 10.4052/tigg.2218.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
24
|
Nishitsuji K, Ikezaki M, Manabe S, Ihara Y. Functions of Protein <i>C</i>-Mannosylation in Physiology and Pathology. TRENDS GLYCOSCI GLYC 2023. [DOI: 10.4052/tigg.2218.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
25
|
Alex L, Tuleta I, Hanna A, Frangogiannis NG. Diabetes Induces Cardiac Fibroblast Activation, Promoting a Matrix-Preserving Nonmyofibroblast Phenotype, Without Stimulating Pericyte to Fibroblast Conversion. J Am Heart Assoc 2023; 12:e027463. [PMID: 36892073 PMCID: PMC10111546 DOI: 10.1161/jaha.122.027463] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/27/2023] [Indexed: 03/10/2023]
Abstract
Background Interstitial and perivascular fibrosis may contribute to diabetes-associated heart failure. Pericytes can convert to fibroblasts under conditions of stress and have been implicated in the pathogenesis of fibrotic diseases. We hypothesized that in diabetic hearts, pericytes may convert to fibroblasts, contributing to fibrosis and to the development of diastolic dysfunction. Methods and Results Using pericyte:fibroblast dual reporter (NG2Dsred [neuron-glial antigen 2 red fluorescent protein variant]; PDGFRαEGFP [platelet-derived growth factor receptor alpha enhanced green fluorescent protein]) mice in a type 2 diabetic db/db background, we found that diabetes does not significantly affect pericyte density but reduces the myocardial pericyte:fibroblast ratio. Lineage tracing using the inducible NG2CreER driver, along with reliable labeling of fibroblasts with the PDGFRα reporter system, showed no significant pericyte to fibroblast conversion in lean and db/db hearts. In addition, db/db mouse cardiac fibroblasts did not undergo myofibroblast conversion and had no significant induction of structural collagens but exhibited a matrix-preserving phenotype, associated with increased expression of antiproteases, matricellular genes, matrix cross-linking enzymes, and the fibrogenic transcription factor cMyc. In contrast, db/db mouse cardiac pericytes had increased expression of Timp3, without any changes in expression of other fibrosis-associated genes. The matrix-preserving phenotype of diabetic fibroblasts was associated with induction of genes encoding oxidative (Ptgs2/cycloxygenase-2, and Fmo2) and antioxidant proteins (Hmox1, Sod1). In vitro, high glucose partially recapitulated the in vivo changes in diabetic fibroblasts. Conclusions Diabetic fibrosis is not mediated through pericyte to fibroblast conversion but involves acquisition of a matrix-preserving fibroblast program, which is independent of myofibroblast conversion and is only partially explained by the effects of the hyperglycemic environment.
Collapse
Affiliation(s)
- Linda Alex
- The Wilf Family Cardiovascular Research InstituteDepartment of Medicine (Cardiology), Albert Einstein College of MedicineBronxNY
| | - Izabela Tuleta
- The Wilf Family Cardiovascular Research InstituteDepartment of Medicine (Cardiology), Albert Einstein College of MedicineBronxNY
| | - Anis Hanna
- The Wilf Family Cardiovascular Research InstituteDepartment of Medicine (Cardiology), Albert Einstein College of MedicineBronxNY
| | - Nikolaos G. Frangogiannis
- The Wilf Family Cardiovascular Research InstituteDepartment of Medicine (Cardiology), Albert Einstein College of MedicineBronxNY
| |
Collapse
|
26
|
Coatti GC, Paranjapye A, Harris A. Dual SMAD inhibition enhances the longevity of human epididymis epithelial cells. Cell Tissue Res 2023; 391:409-417. [PMID: 36454271 PMCID: PMC9713186 DOI: 10.1007/s00441-022-03712-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022]
Abstract
Primary human epididymis epithelial (HEE) cells are valuable reagents for functional studies on the human epididymis. We used them previously to determine the transcriptional networks that establish cell identity along the length of the epididymis from caput, corpus, and cauda. These studies on HEE cells and organoids derived from them revealed important cellular properties. However, similar to other primary cells, HEE cells undergo replicative senescence and de-differentiation in culture. A cocktail of small molecules was shown elsewhere to extend longevity of epithelial basal cells. The components included transforming growth factor β (TGF-β)/bone morphogenetic protein (BMP) antagonists, WNT agonist, and Rho-associated and coiled-coil containing protein kinase (ROCK) inhibitor (ROCKi), which together prevented the senescence-related upregulation of TGF-β signaling pathway members. Here, we treat HEE cells with the same cocktail and observed enhanced replicative potential and prolonged expression of markers of HEE differentiation. This treatment expands the differentiated HEE cell population available from individual epididymis tissue samples that can be used for molecular, cellular, and functional studies.
Collapse
Affiliation(s)
- Giuliana C Coatti
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
27
|
Novitskaya T, Nishat S, Covarrubias R, Wheeler DG, Chepurko E, Bermeo-Blanco O, Xu Z, Baer B, He H, Moore SN, Dwyer KM, Cowan PJ, Su YR, Absi TS, Schoenecker J, Bellan LM, Koch WJ, Bansal S, Feoktistov I, Robson SC, Gao E, Gumina RJ. Ectonucleoside triphosphate diphosphohydrolase-1 (CD39) impacts TGF-β1 responses: insights into cardiac fibrosis and function following myocardial infarction. Am J Physiol Heart Circ Physiol 2022; 323:H1244-H1261. [PMID: 36240436 PMCID: PMC9722260 DOI: 10.1152/ajpheart.00138.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
Extracellular purine nucleotides and nucleosides released from activated or injured cells influence multiple aspects of cardiac physiology and pathophysiology. Ectonucleoside triphosphate diphosphohydrolase-1 (ENTPD1; CD39) hydrolyzes released nucleotides and thereby regulates the magnitude and duration of purinergic signaling. However, the impact of CD39 activity on post-myocardial infarction (MI) remodeling is incompletely understood. We measured the levels and activity of ectonucleotidases in human left ventricular samples from control and ischemic cardiomyopathy (ICM) hearts and examined the impact of ablation of Cd39 expression on post-myocardial infarction remodeling in mice. We found that human CD39 levels and activity are significantly decreased in ICM hearts (n = 5) compared with control hearts (n = 5). In mice null for Cd39, cardiac function and remodeling are significantly compromised in Cd39-/- mice following myocardial infarction. Fibrotic markers including plasminogen activator inhibitor-1 (PAI-1) expression, fibrin deposition, α-smooth muscle actin (αSMA), and collagen expression are increased in Cd39-/- hearts. Importantly, we found that transforming growth factor β1 (TGF-β1) stimulates ATP release and induces Cd39 expression and activity on cardiac fibroblasts, constituting an autocrine regulatory pathway not previously appreciated. Absence of CD39 activity on cardiac fibroblasts exacerbates TGF-β1 profibrotic responses. Treatment with exogenous ectonucleotidase rescues this profibrotic response in Cd39-/- fibroblasts. Together, these data demonstrate that CD39 has important interactions with TGF-β1-stimulated autocrine purinergic signaling in cardiac fibroblasts and dictates outcomes of cardiac remodeling following myocardial infarction. Our results reveal that ENTPD1 (CD39) regulates TGF-β1-mediated fibroblast activation and limits adverse cardiac remodeling following myocardial infarction.NEW & NOTEWORTHY We show that CD39 is a critical modulator of TGF-β1-mediated fibroblast activation and cardiac remodeling following myocardial infarction via modulation of nucleotide signaling. TGF-β1-induced CD39 expression generates a negative feedback loop that attenuates cardiac fibroblast activation. In the absence of CD39 activity, collagen deposition is increased, elastin expression is decreased, and diastolic dysfunction is worsened. Treatment with ecto-apyrase attenuates the TGF-β1-induced profibrotic cardiac fibroblast phenotype, revealing a novel approach to combat post-myocardial infarction cardiac fibrosis.
Collapse
Affiliation(s)
- Tatiana Novitskaya
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shamama Nishat
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Roman Covarrubias
- Division of Cardiac Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Debra G Wheeler
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Elena Chepurko
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Oscar Bermeo-Blanco
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Zhaobin Xu
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Bradly Baer
- Department of Mechanical Engineering, Vanderbilt University School of Engineering, Nashville, Tennessee
| | - Heng He
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Stephanie N Moore
- Division of Orthopedic Surgery, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Karen M Dwyer
- Immunology Research Center, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter J Cowan
- Immunology Research Center, St. Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Yan Ru Su
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tarek S Absi
- Division of Cardiac Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan Schoenecker
- Division of Orthopedic Surgery, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leon M Bellan
- Department of Mechanical Engineering, Vanderbilt University School of Engineering, Nashville, Tennessee
| | | | - Shyam Bansal
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Igor Feoktistov
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Simon C Robson
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Erhe Gao
- Temple University, Philadelphia, Pennsylvania
| | - Richard J Gumina
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
28
|
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
|
29
|
BMP3 inhibits TGFβ2-mediated myofibroblast differentiation during wound healing of the embryonic cornea. NPJ Regen Med 2022; 7:36. [PMID: 35879352 PMCID: PMC9314337 DOI: 10.1038/s41536-022-00232-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
Often acute damage to the cornea initiates drastic tissue remodeling, resulting in fibrotic scarring that disrupts light transmission and precedes vision impairment. Very little is known about the factors that can mitigate fibrosis and promote scar-free cornea wound healing. We previously described transient myofibroblast differentiation during non-fibrotic repair in an embryonic cornea injury model. Here, we sought to elucidate the mechanistic regulation of myofibroblast differentiation during embryonic cornea wound healing. We found that alpha-smooth muscle actin (αSMA)-positive myofibroblasts are superficial and their presence inversely correlates with wound closure. Expression of TGFβ2 and nuclear localization of pSMAD2 were elevated during myofibroblast induction. BMP3 and BMP7 were localized in the corneal epithelium and corresponded with pSMAD1/5/8 activation and absence of myofibroblasts in the healing stroma. In vitro analyses with corneal fibroblasts revealed that BMP3 inhibits the persistence of TGFβ2-induced myofibroblasts by promoting disassembly of focal adhesions and αSMA fibers. This was confirmed by the expression of vinculin and pFAK. Together, these data highlight a mechanism to inhibit myofibroblast persistence during cornea wound repair.
Collapse
|
30
|
Song X, Shi J, Liu J, Liu Y, Yu Y, Qiu Y, Cao Z, Pan Y, Yuan X, Chu Y, Wu D. Recombinant truncated latency-associated peptide alleviates liver fibrosis in vitro and in vivo via inhibition of TGF-β/Smad pathway. Mol Med 2022; 28:80. [PMID: 35842576 PMCID: PMC9288003 DOI: 10.1186/s10020-022-00508-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Background Liver fibrosis is a progressive liver injury response. Transforming growth factor β1 (TGF-β1) is oversecreted during liver fibrosis and promotes the development of liver fibrosis. Therapeutic approaches targeting TGF-β1 and its downstream pathways are essential to inhibit liver fibrosis. The N-terminal latency-associated peptide (LAP) blocks the binding of TGF-β1 to its receptor. Removal of LAP is critical for the activation of TGF-β1. Therefore, inhibition of TGF-β1 and its downstream pathways by LAP may be a potential approach to affect liver fibrosis. Methods Truncated LAP (tLAP) plasmids were constructed. Recombinant proteins were purified by Ni affinity chromatography. The effects of LAP and tLAP on liver fibrosis were investigated in TGF-β1-induced HSC-T6 cells, AML12 cells and CCl4-induced liver fibrosis mice by real time cellular analysis (RTCA), western blot, real-time quantitative PCR (RT-qPCR), immunofluorescence and pathological staining. Results LAP and tLAP could inhibit TGF-β1-induced AML12 cells inflammation, apoptosis and EMT, and could inhibit TGF-β1-induced HSC-T6 cells proliferation and fibrosis. LAP and tLAP could attenuate the pathological changes of liver fibrosis and inhibit the expression of fibrosis-related proteins and mRNAs in CCl4-induced liver fibrosis mice. Conclusion LAP and tLAP could alleviate liver fibrosis in vitro and in vivo via inhibition of TGF-β/Smad pathway. TLAP has higher expression level and more effective anti-fibrosis activity compared to LAP. This study may provide new ideas for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Xudong Song
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Jiayi Shi
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Jieting Liu
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Yong Liu
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Yang Yu
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Yufei Qiu
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Zhiqin Cao
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Yu Pan
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Xiaohuan Yuan
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China.,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China
| | - Yanhui Chu
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China. .,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China.
| | - Dan Wu
- Heilongjiang Province Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, No. 3, Tongxiang Street, Aimin District, Mudanjiang, 157011, Heilongjiang, China. .,College of Life Sciences, Mudanjiang Medical University, Mudanjiang, 157011, Heilongjiang, China.
| |
Collapse
|
31
|
Unravelling Novel Roles of Salivary Exosomes in the Regulation of Human Corneal Stromal Cell Migration and Wound Healing. Int J Mol Sci 2022; 23:ijms23084330. [PMID: 35457149 PMCID: PMC9024472 DOI: 10.3390/ijms23084330] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 02/01/2023] Open
Abstract
Salivary exosomes have demonstrated vast therapeutic and diagnostic potential in numerous diseases. This study pioneers previously unexplored roles of SE in the context of corneal wound healing by utilizing primary corneal stromal cells from healthy (HCFs), type I diabetes mellitus (T1DMs), type II DM (T2DMs), and keratoconus (HKCs) subjects. Purified, healthy human SEs carrying tetraspanins CD9+, CD63+, and CD81+ were utilized. Scratch and cell migration assays were performed after 0, 6, 12, 24, and 48 h following SE stimulation (5 and 25 µg/mL). Significantly slower wound closure was observed at 6 and 12 h in HCFs with 5 μg/mL SE and T1DMs with 5 and 25 μg/mL SE. All wounds were closed by 24-hour, post-wounding. HKCs, T1DMs, and T2DMs with 25µg/mL SE exhibited a significant upregulation of cleaved vimentin compared to controls. Thrombospondin 1 was significantly upregulated in HCFs, HKCs, and T2DMs with 25 µg/mL SE. Lastly, HKCs, T1DMs, and T2DMs exhibited a significant downregulation of fibronectin with 25 μg/mL SE. Whether SEs can be utilized to clinical settings in restoring corneal defects is unknown. This is the first-ever study exploring the role of SEs in corneal wound healing. While the sample size was small, results are highly novel and provide a strong foundation for future studies.
Collapse
|
32
|
Czaja AJ. Immune Inhibitory Properties and Therapeutic Prospects of Transforming Growth Factor-Beta and Interleukin 10 in Autoimmune Hepatitis. Dig Dis Sci 2022; 67:1163-1186. [PMID: 33835375 DOI: 10.1007/s10620-021-06968-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-beta and interleukin 10 have diverse immune inhibitory properties that have restored homeostatic defense mechanisms in experimental models of autoimmune disease. The goals of this review are to describe the actions of each cytokine, review their investigational use in animal models and patients, and indicate their prospects as interventions in autoimmune hepatitis. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Transforming growth factor-beta expands the natural and inducible populations of regulatory T cells, limits the proliferation of natural killer cells, suppresses the activation of naïve CD8+ T cells, decreases the production of interferon-gamma, and stimulates fibrotic repair. Interleukin 10 selectively inhibits the CD28 co-stimulatory signal for antigen recognition and impairs antigen-specific activation of uncommitted CD4+ and CD8+ T cells. It also inhibits maturation of dendritic cells, suppresses Th17 cells, supports regulatory T cells, and limits production of diverse pro-inflammatory cytokines. Contradictory immune stimulatory effects have been associated with each cytokine and may relate to the dose and accompanying cytokine milieu. Experimental findings have not translated into successful early clinical trials. The recombinant preparation of each agent in low dosage has been safe in human studies. In conclusion, transforming growth factor-beta and interleukin 10 have powerful immune inhibitory actions of potential therapeutic value in autoimmune hepatitis. The keys to their therapeutic application will be to match their predominant non-redundant function with the pivotal pathogenic mechanism or cytokine deficiency and to avoid contradictory immune stimulatory actions.
Collapse
Affiliation(s)
- Albert J Czaja
- Professor Emeritus of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
33
|
Chen P, Liu C, Zhang J, Chen X, Liu X, He S, He A, Chen S, Qiu J, Li Y, Jiang Z, Yu K, Zhuang J. Tsp-1 is involved in DNA stability through Tgf-β1 activation domain in cone photoreceptor 661 W cells. Cell Tissue Res 2022; 388:259-271. [PMID: 35260935 DOI: 10.1007/s00441-022-03606-z] [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: 06/01/2021] [Accepted: 02/24/2022] [Indexed: 11/02/2022]
Abstract
Thrombospondin-1 (Tsp-1), a matricellular protein, could protect retinal neurons from endogenous or exogenous insults; however, its underlying mechanism remains unclear. Thus, this study aimed to investigate Tsp-1-mediated neuron-protection effect in retinal cells. Our data showed that Tsp-1 downregulation would aggravate UV irradiation-induced DNA damage in 661 W cells and cone photoreceptor cells. The increasing levels of poly (ADP ribose) polymer (PAR) and γ-H2AX in Tsp-1-silenced 661 W cells indicate severe DNA single-strand breaks (SSBs) and double-strand breaks (DSBs). By utilizing an error-prone substrate, Tsp-1 silencing significantly increased deleted DNA end joining in 661 W cells with spontaneous DNA damage (SDD). Moreover, Tsp-1 is indirectly involved in DNA stability in 661 W cells as UV treatment caused a significant Tsp-1 decreasing in cytoplasm, but no obvious Tsp-1 alteration in cell nuclear of 661 W cells. Furthermore, our data indicate that Tgf-β1 activation domain in Tsp-1 plays a critical role in DNA stability in 661 W cells through expressing mutated exogenous Tsp-1 and Tgf-β inhibitor, LSKL. Therefore, this study provides new insights into the mechanism of the neuroprotective action positively mediated by Tsp-1, which might be a therapeutic target for the treatment of retinal pathology.
Collapse
Affiliation(s)
- Pei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Chang Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xuan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shengyu He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Anqi He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shuilian Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jin Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Zihua Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Keming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
34
|
Bandzerewicz A, Gadomska-Gajadhur A. Into the Tissues: Extracellular Matrix and Its Artificial Substitutes: Cell Signalling Mechanisms. Cells 2022; 11:914. [PMID: 35269536 PMCID: PMC8909573 DOI: 10.3390/cells11050914] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
The existence of orderly structures, such as tissues and organs is made possible by cell adhesion, i.e., the process by which cells attach to neighbouring cells and a supporting substance in the form of the extracellular matrix. The extracellular matrix is a three-dimensional structure composed of collagens, elastin, and various proteoglycans and glycoproteins. It is a storehouse for multiple signalling factors. Cells are informed of their correct connection to the matrix via receptors. Tissue disruption often prevents the natural reconstitution of the matrix. The use of appropriate implants is then required. This review is a compilation of crucial information on the structural and functional features of the extracellular matrix and the complex mechanisms of cell-cell connectivity. The possibilities of regenerating damaged tissues using an artificial matrix substitute are described, detailing the host response to the implant. An important issue is the surface properties of such an implant and the possibilities of their modification.
Collapse
|
35
|
Dakshinamurti S. Thrombospondin in the Puzzle of Bronchopulmonary Dysplasia. Am J Respir Crit Care Med 2022; 205:610-612. [PMID: 35120300 DOI: 10.1164/rccm.202201-0101ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Shyamala Dakshinamurti
- University of Manitoba Faculty of Health Sciences, 423134, Pediatrics, Winnipeg, Manitoba, Canada.,University of Manitoba Children's Hospital Research Institute of Manitoba, 423136, Biology of Breathing, Winnipeg, Manitoba, Canada;
| |
Collapse
|
36
|
He L, Wang GP, Guo JY, Chen ZR, Liu K, Gong SS. Epithelial-Mesenchymal Transition Participates in the Formation of Vestibular Flat Epithelium. Front Mol Neurosci 2022; 14:809878. [PMID: 34975404 PMCID: PMC8719593 DOI: 10.3389/fnmol.2021.809878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/02/2021] [Indexed: 12/03/2022] Open
Abstract
The vestibular sensory epithelium of humans and mice may degenerate into a layer of flat cells, known as flat epithelium (FE), after a severe lesion. However, the pathogenesis of vestibular FE remains unclear. To determine whether the epithelial–mesenchymal transition (EMT) participates in the formation of vestibular FE, we used a well-established mouse model in which FE was induced in the utricle by an injection of streptomycin into the inner ear. The mesenchymal and epithelial cell markers and cell proliferation were examined using immunofluorescence staining and quantitative reverse transcription polymerase chain reaction (qRT-PCR). The function of the EMT was assessed through transcriptome microarray analysis. The results demonstrated that mesenchymal cell markers (α-SMA, S100A4, vimentin, and Fn1) were upregulated in vestibular FE compared with the normal utricle. Robust cell proliferation, which was absent in the normal status, was observed in the formation of FE. Microarray analysis identified 1,227 upregulated and 962 downregulated genes in vestibular FE. Gene Ontology (GO) analysis revealed that differentially expressed genes (DEGs) were highly associated with several EMT-related GO terms, such as cell adhesion, cell migration, and extracellular matrix. Pathway enrichment analysis revealed that DEGs were enriched in the EMT-related signaling pathways, including extracellular matrix (ECM)-receptor interaction, focal adhesion, PI3K/Akt signaling pathway and cell adhesion molecule. Protein–protein interaction networks screened 20 hub genes, which were Akt, Casp3, Col1a1, Col1a2, Fn1, Hgf, Igf1,Il1b, Irs1, Itga2, Itga5, Jun, Mapk1, Myc, Nras, Pdgfrb, Tgfb1, Thbs1, Trp53, and Col2a1. Most of these genes are reportedly involved in the EMT process in various tissues. The mRNA expression level of hub genes was validated using qRT-PCR. In conclusion, the present study indicates that EMT plays a significant role in the formation of vestibular FE and provides an overview of transcriptome characteristics in vestibular FE.
Collapse
Affiliation(s)
- Lu He
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guo-Peng Wang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jing-Ying Guo
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhong-Rui Chen
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ke Liu
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shu-Sheng Gong
- Department of Otolaryngology-Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Li X, Wang J, Wu C, Lu X, Huang J. MicroRNAs involved in the TGF-β signaling pathway in atherosclerosis. Biomed Pharmacother 2021; 146:112499. [PMID: 34959122 DOI: 10.1016/j.biopha.2021.112499] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease with a multifactorial pathogenesis. It becomes a global health concern, especially causing an array of fatal consequences among the elderly. However, the mechanisms of AS remain unexplained. The transforming growth factor-β (TGF-β) signaling pathway is widely involved in the inflammation, immune function, proliferation, differentiation,and apoptosis in vivo. Based on previous researches, it has not been confirmed whether the TGF-β pathway promotes or inhibits atherosclerosis. Furthermore, more and more studies have found that microRNAs can regulate atherosclerosis through the TGF-β signaling pathway. In this review, we summarize and discuss the role of microRNAs in the pathogenesis of atherosclerosis via the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinyu Wang
- Department of Cardiology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Wu
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiang Lu
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jingjing Huang
- Department of Geriatrics, the Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
38
|
Mesenchymal stromal cells mitigate liver damage after extended resection in the pig by modulating thrombospondin-1/TGF-β. NPJ Regen Med 2021; 6:84. [PMID: 34862411 PMCID: PMC8642541 DOI: 10.1038/s41536-021-00194-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Post-surgery liver failure is a serious complication for patients after extended partial hepatectomies (ePHx). Previously, we demonstrated in the pig model that transplantation of mesenchymal stromal cells (MSC) improved circulatory maintenance and supported multi-organ functions after 70% liver resection. Mechanisms behind the beneficial MSC effects remained unknown. Here we performed 70% liver resection in pigs with and without MSC treatment, and animals were monitored for 24 h post surgery. Gene expression profiles were determined in the lung and liver. Bioinformatics analysis predicted organ-independent MSC targets, importantly a role for thrombospondin-1 linked to transforming growth factor-β (TGF-β) and downstream signaling towards providing epithelial plasticity and epithelial-mesenchymal transition (EMT). This prediction was supported histologically and mechanistically, the latter with primary hepatocyte cell cultures. MSC attenuated the surgery-induced increase of tissue damage, of thrombospondin-1 and TGF-β, as well as of epithelial plasticity in both the liver and lung. This suggests that MSC ameliorated surgery-induced hepatocellular stress and EMT, thus supporting epithelial integrity and facilitating regeneration. MSC-derived soluble factor(s) did not directly interfere with intracellular TGF-β signaling, but inhibited thrombospondin-1 secretion from thrombocytes and non-parenchymal liver cells, therewith obviously reducing the availability of active TGF-β.
Collapse
|
39
|
Halper J. Basic Components of Connective Tissues and Extracellular Matrix: Fibronectin, Fibrinogen, Laminin, Elastin, Fibrillins, Fibulins, Matrilins, Tenascins and Thrombospondins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1348:105-126. [PMID: 34807416 DOI: 10.1007/978-3-030-80614-9_4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Collagens are the most abundant components of the extracellular matrix (ECM) and many types of soft tissues. Elastin is another major component of certain soft tissues, such as arterial walls and ligaments. It is an insoluble polymer of the monomeric soluble precursor tropoelastin, and the main component of elastic fibers in matrix tissue where it provides elastic recoil and resilience to a variety of connective tissues, e.g., aorta and ligaments. Elastic fibers regulate activity of transforming growth factors β (TGFβ) through their association with fibrillin microfibrils. Elastin also plays a role in cell adhesion, cell migration, and has the ability to participate in cell signaling. Mutations in the elastin gene lead to cutis laxa. Many other molecules, though lower in quantity, function as essential, structural and/or functional components of the extracellular matrix in soft tissues. Some of these are reviewed in this chapter. Besides their basic structure, biochemistry and physiology, their roles in disorders of soft tissues are discussed only briefly as most chapters in this volume deal with relevant individual compounds. Fibronectin with its multidomain structure plays a role of "master organizer" in matrix assembly as it forms a bridge between cell surface receptors, e.g., integrins, and compounds such collagen, proteoglycans and other focal adhesion molecules. It also plays an essential role in the assembly of fibrillin-1 into a structured network. Though the primary role of fibrinogen is in clot formation, after conversion to fibrin by thrombin it also binds to a variety of compounds, particularly to various growth factors, and as such, fibrinogen is a player in cardiovascular and extracellular matrix physiology. Laminins contribute to the structure of the ECM and modulate cellular functions such as adhesion, differentiation, migration, stability of phenotype, and resistance towards apoptosis. Fibrillins represent the predominant core of microfibrils in elastic as well as non-elastic extracellular matrixes, and interact closely with tropoelastin and integrins. Not only do microfibrils provide structural integrity of specific organ systems, but they also provide basis for elastogenesis in elastic tissues. Fibrillin is important for the assembly of elastin into elastic fibers. Mutations in the fibrillin-1 gene are closely associated with Marfan syndrome. Latent TGFβ binding proteins (LTBPs) are included here as their structure is similar to fibrillins. Several categories of ECM components described after fibrillins are sub-classified as matricellular proteins, i.e., they are secreted into ECM, but do not provide structure. Rather they interact with cell membrane receptors, collagens, proteases, hormones and growth factors, communicating and directing cell-ECM traffic. Fibulins are tightly connected with basement membranes, elastic fibers and other components of extracellular matrix and participate in formation of elastic fibers. Matrilins have been emerging as a new group of supporting actors, and their role in connective tissue physiology and pathophysiology has not been fully characterized. Tenascins are ECM polymorphic glycoproteins found in many connective tissues in the body. Their expression is regulated by mechanical stress both during development and in adulthood. Tenascins mediate both inflammatory and fibrotic processes to enable effective tissue repair and play roles in pathogenesis of Ehlers-Danlos, heart disease, and regeneration and recovery of musculo-tendinous tissue. One of the roles of thrombospondin 1 is activation of TGFβ. Increased expression of thrombospondin and TGFβ activity was observed in fibrotic skin disorders such as keloids and scleroderma. Cartilage oligomeric matrix protein (COMP) or thrombospondin-5 is primarily present in the cartilage. High levels of COMP are present in fibrotic scars and systemic sclerosis of the skin, and in tendon, especially with physical activity, loading and post-injury. It plays a role in vascular wall remodeling and has been found in atherosclerotic plaques as well.
Collapse
Affiliation(s)
- Jaroslava Halper
- Department of Pathology, College of Veterinary Medicine, and Department of Basic Sciences, AU/UGA Medical Partnership, The University of Georgia, Athens, GA, USA.
| |
Collapse
|
40
|
Corrigendum. FEBS Lett 2021; 595:2844. [PMID: 34747013 DOI: 10.1002/1873-3468.14217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
41
|
Yu L, Guo Y, Chang Z, Zhang D, Zhang S, Pei H, Pang J, Zhao ZJ, Chen Y. Bidirectional Interaction Between Cancer Cells and Platelets Provides Potential Strategies for Cancer Therapies. Front Oncol 2021; 11:764119. [PMID: 34722319 PMCID: PMC8551800 DOI: 10.3389/fonc.2021.764119] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Platelets are essential components in the tumor microenvironment. For decades, clinical data have demonstrated that cancer patients have a high risk of thrombosis that is associated with adverse prognosis and decreased survival, indicating the involvement of platelets in cancer progression. Increasing evidence confirms that cancer cells are able to induce production and activation of platelets. Once activated, platelets serve as allies of cancer cells in tumor growth and metastasis. They can protect circulating tumor cells (CTCs) against the immune system and detachment-induced apoptosis while facilitating angiogenesis and tumor cell adhesion and invasion. Therefore, antiplatelet agents and platelet-based therapies should be developed for cancer treatment. Here, we discuss the mechanisms underlying the bidirectional cancer-platelet crosstalk and platelet-based therapeutic approaches.
Collapse
Affiliation(s)
- Liuting Yu
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yao Guo
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhiguang Chang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hanzhong Pei
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jun Pang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
42
|
Pacurari M, Mitra A, Turner T. Idiopathic Pulmonary Comorbidities and Mechanisms. Int J Inflam 2021; 2021:3963659. [PMID: 34691383 PMCID: PMC8528608 DOI: 10.1155/2021/3963659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/24/2021] [Accepted: 10/05/2021] [Indexed: 11/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease with an unknown etiology mainly characterized by a progressive decline of lung function due to the scarring of the tissue deep in the lungs. The overall survival after diagnosis remains low between 3 and 5 years. IPF is a heterogeneous disease and much progress has been made in the past decade in understanding the disease mechanisms that contributed to the development of two new drugs, pirfenidone and nintedanib, which improved the therapeutic management of the disease. The understanding of the cofactors and comorbidities of IPF also contributed to improved management of the disease outcome. In the present review, we evaluate scientific evidence which indicates IPF as a risk factor for other diseases based on the complexity of molecular and cellular mechanisms involved in the disease development and of comorbidities. We conclude from the existing literature that while much progress has been made in understating the mechanisms involved in IPF development, further studies are still necessary to fully understand IPF pathogenesis which will contribute to the identification of novel therapeutic targets for IPF management as well as other diseases for which IPF is a major risk factor.
Collapse
Affiliation(s)
- Maricica Pacurari
- Department of Biology, College of Science, Engineering, and Technology, Jackson State University, Jackson, MS 39217, USA
| | - Amal Mitra
- Department of Epidemiology and Biostatistics, School of Public Health, Jackson State University, Jackson, MS 39217, USA
| | - Timothy Turner
- Department of Biology, College of Science, Engineering, and Technology, Jackson State University, Jackson, MS 39217, USA
| |
Collapse
|
43
|
Voronovic E, Skripka A, Jarockyte G, Ger M, Kuciauskas D, Kaupinis A, Valius M, Rotomskis R, Vetrone F, Karabanovas V. Uptake of Upconverting Nanoparticles by Breast Cancer Cells: Surface Coating versus the Protein Corona. ACS APPLIED MATERIALS & INTERFACES 2021; 13:39076-39087. [PMID: 34378375 PMCID: PMC8824430 DOI: 10.1021/acsami.1c10618] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Fluorophores with multifunctional properties known as rare-earth-doped nanoparticles (RENPs) are promising candidates for bioimaging, therapy, and drug delivery. When applied in vivo, these nanoparticles (NPs) have to retain long blood-circulation time, bypass elimination by phagocytic cells, and successfully arrive at the target area. Usually, NPs in a biological medium are exposed to proteins, which form the so-called "protein corona" (PC) around the NPs and influence their targeted delivery and accumulation in cells and tissues. Different surface coatings change the PC size and composition, subsequently deciding the fate of the NPs. Thus, detailed studies on the PC are of utmost importance to determine the most suitable NP surface modification for biomedical use. When it comes to RENPs, these studies are particularly scarce. Here, we investigate the PC composition and its impact on the cellular uptake of citrate-, SiO2-, and phospholipid micelle-coated RENPs (LiYF4:Yb3+,Tm3+). We observed that the PC of citrate- and phospholipid-coated RENPs is relatively stable and similar in the adsorbed protein composition, while the PC of SiO2-coated RENPs is larger and highly dynamic. Moreover, biocompatibility, accumulation, and cytotoxicity of various RENPs in cancer cells have been evaluated. On the basis of the cellular imaging, supported by the inhibition studies, it was revealed that RENPs are internalized by endocytosis and that specific endocytic routes are PC composition dependent. Overall, these results are essential to fill the gaps in the fundamental understanding of the nano-biointeractions of RENPs, pertinent for their envisioned application in biomedicine.
Collapse
Affiliation(s)
- Evelina Voronovic
- Biomedical
Physics Laboratory of National Cancer Institute, Baublio 3B, LT-08406 Vilnius, Lithuania
- Life
Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania
- Department
of Chemistry and Bioengineering, Vilnius
Gediminas Technical University, Sauletekio av. 11, LT-10223 Vilnius, Lithuania
| | - Artiom Skripka
- Centre
Énergie, Matériaux et Télécommunications, Institut National de la Recherche Scientifique, Université
du Québec, 1650 Boul. Lionel-Boulet, Varennes, Quebec J3X 1S2, Canada
| | - Greta Jarockyte
- Biomedical
Physics Laboratory of National Cancer Institute, Baublio 3B, LT-08406 Vilnius, Lithuania
- Life
Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania
| | - Marija Ger
- Institute
of Biochemistry, Life Sciences Center, Vilnius
University, Sauletekio
av. 7, LT-10257 Vilnius, Lithuania
| | - Dalius Kuciauskas
- Institute
of Biochemistry, Life Sciences Center, Vilnius
University, Sauletekio
av. 7, LT-10257 Vilnius, Lithuania
| | - Algirdas Kaupinis
- Institute
of Biochemistry, Life Sciences Center, Vilnius
University, Sauletekio
av. 7, LT-10257 Vilnius, Lithuania
| | - Mindaugas Valius
- Institute
of Biochemistry, Life Sciences Center, Vilnius
University, Sauletekio
av. 7, LT-10257 Vilnius, Lithuania
| | - Ricardas Rotomskis
- Biomedical
Physics Laboratory of National Cancer Institute, Baublio 3B, LT-08406 Vilnius, Lithuania
- Biophotonics
Group of Laser Research Centre, Vilnius
University, Sauletekio
av. 9, LT-10222 Vilnius, Lithuania
| | - Fiorenzo Vetrone
- Centre
Énergie, Matériaux et Télécommunications, Institut National de la Recherche Scientifique, Université
du Québec, 1650 Boul. Lionel-Boulet, Varennes, Quebec J3X 1S2, Canada
| | - Vitalijus Karabanovas
- Biomedical
Physics Laboratory of National Cancer Institute, Baublio 3B, LT-08406 Vilnius, Lithuania
- Department
of Chemistry and Bioengineering, Vilnius
Gediminas Technical University, Sauletekio av. 11, LT-10223 Vilnius, Lithuania
| |
Collapse
|
44
|
Habuddha V, Suwannasing C, Buddawong A, Seenprachawong K, Duangchan T, Sombutkayasith C, Supokawej A, Weerachatyanukul W, Asuvapongpatana S. Characterization of Thrombospondin Type 1 Repeat in Haliotis diversicolor and Its Possible Role in Osteoinduction. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:641-652. [PMID: 34471969 DOI: 10.1007/s10126-021-10054-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/02/2021] [Indexed: 06/13/2023]
Abstract
Thrombospondin repeats (TSR) are important peptide domains present in the sequences of many extracellular and transmembrane proteins with which a variety of ligands interact. In this study, we characterized HdTSR domains in the ADAMTS3 protein of Thai abalone, Haliotis diversicolor, based on the transcriptomic analysis of its mantle tissues. PCR amplification and localization studies demonstrated the existence of HdTSR transcript and protein in H. diversicolor tissues, particularly in both the inner and outer mantle epithelial folds. We, therefore, generated a short recombinant protein, termed HdTSR1/2, based on the existence of the WxxWxxW or WxxxxW motif (which binds to TGF-β, a known signaling in bone formation/repair) in HdTSR1 and HdTSR2 sequences and used it to test the osteoinduction function in the pre-osteoblastic cell line, MC3T3-E1. This recombinant protein demonstrated the ability to induce the differentiation of MC3T3-E1 cells by the concentration- and time-dependent upregulation of many known osteogenic markers, including RUNX2, COL1A1, OCN, and OPN. We also demonstrated the upregulation of the SMAD2 gene after cell treatment with HdTSR1/2 proteinindicating its possible interaction through TGF-β, which thus activates its downstream signaling cascade and triggers the biomineralization process in the differentiated osteoblastic cells. Together, HdTSR domains existed in an extracellular ADAMTS3 protein in the mantle epithelium of H. diversicolor and played a role in osteoinduction as similar to the other nacreous proteins, opening up its possibility to be developed as an inducing agent of bone repair.
Collapse
Affiliation(s)
- Valainipha Habuddha
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
- School of Allied Health Science, Walailak University, Nakhon Si Thammarat, Thailand
| | - Chanyatip Suwannasing
- Department of Radiological Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Aticha Buddawong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
- Chulabhorn International College of Medicine, Thammasat University, Khlong Nueng Pathumthani 12121, Rangsit Campus, Thailand
| | - Kanokwan Seenprachawong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University (Salaya Campus), Salaya, Nakhonpathom, Thailand
| | - Thitinat Duangchan
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University (Salaya Campus), Salaya, Nakhonpathom, Thailand
| | | | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University (Salaya Campus), Salaya, Nakhonpathom, Thailand
| | | | | |
Collapse
|
45
|
Fu Z, Dean JW, Xiong L, Dougherty MW, Oliff KN, Chen ZME, Jobin C, Garrett TJ, Zhou L. Mitochondrial transcription factor A in RORγt + lymphocytes regulate small intestine homeostasis and metabolism. Nat Commun 2021; 12:4462. [PMID: 34294718 PMCID: PMC8298438 DOI: 10.1038/s41467-021-24755-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
RORγt+ lymphocytes, including interleukin 17 (IL-17)-producing gamma delta T (γδT17) cells, T helper 17 (Th17) cells, and group 3 innate lymphoid cells (ILC3s), are important immune regulators. Compared to Th17 cells and ILC3s, γδT17 cell metabolism and its role in tissue homeostasis remains poorly understood. Here, we report that the tissue milieu shapes splenic and intestinal γδT17 cell gene signatures. Conditional deletion of mitochondrial transcription factor A (Tfam) in RORγt+ lymphocytes significantly affects systemic γδT17 cell maintenance and reduces ILC3s without affecting Th17 cells in the gut. In vivo deletion of Tfam in RORγt+ lymphocytes, especially in γδT17 cells, results in small intestine tissue remodeling and increases small intestine length by enhancing the type 2 immune responses in mice. Moreover, these mice show dysregulation of the small intestine transcriptome and metabolism with less body weight but enhanced anti-helminth immunity. IL-22, a cytokine produced by RORγt+ lymphocytes inhibits IL-13-induced tuft cell differentiation in vitro, and suppresses the tuft cell-type 2 immune circuit and small intestine lengthening in vivo, highlighting its key role in gut tissue remodeling.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Joseph W Dean
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Lifeng Xiong
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | | | - Kristen N Oliff
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Zong-Ming E Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Christian Jobin
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32608, USA
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
46
|
Kaur G, Wright K, Mital P, Hibler T, Miranda JM, Thompson LA, Halley K, Dufour JM. Neonatal Pig Sertoli Cells Survive Xenotransplantation by Creating an Immune Modulatory Environment Involving CD4 and CD8 Regulatory T Cells. Cell Transplant 2021; 29:963689720947102. [PMID: 32841048 PMCID: PMC7564626 DOI: 10.1177/0963689720947102] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The acute cell-mediated immune response presents a significant barrier to
xenotransplantation. Immune-privileged Sertoli cells (SC) can prolong the
survival of co-transplanted cells including xenogeneic islets, hepatocytes, and
neurons by protecting them from immune rejection. Additionally, SC survive as
allo- and xenografts without the use of any immunosuppressive drugs suggesting
elucidating the survival mechanism(s) of SC could be used to improve survival of
xenografts. In this study, the survival and immune response generated toward
neonatal pig SC (NPSC) or neonatal pig islets (NPI), nonimmune-privileged
controls, was compared after xenotransplantation into naïve Lewis rats without
immune suppression. The NPSC survived throughout the study, while NPI were
rejected within 9 days. Analysis of the grafts revealed that macrophages and T
cells were the main immune cells infiltrating the NPSC and NPI grafts. Further
characterization of the T cells within the grafts indicated that the NPSC grafts
contained significantly more cluster of differentiation 4 (CD4) and cluster of
differentiation 8 (CD8) regulatory T cells (Tregs) at early time points than the
NPI grafts. Additionally, the presence of increased amounts of interleukin 10
(IL-10) and transforming growth factor (TGF) β and decreased levels of tumor
necrosis factor (TNF) α and apoptosis in the NPSC grafts compared to NPI grafts
suggests the presence of regulatory immune cells in the NPSC grafts. The NPSC
expressed several immunoregulatory factors such as TGFβ, thrombospondin-1
(THBS1), indoleamine-pyrrole 2,3-dioxygenase, and galectin-1, which could
promote the recruitment of these regulatory immune cells to the NPSC grafts. In
contrast, NPI grafts had fewer Tregs and increased apoptosis and inflammation
(increased TNFα, decreased IL-10 and TGFβ) suggestive of cytotoxic immune cells
that contribute to their early rejection. Collectively, our data suggest that a
regulatory graft environment with regulatory immune cells including CD4 and
CD8 Tregs in NPSC grafts could be attributed to the prolonged survival of the
NPSC xenografts.
Collapse
Affiliation(s)
- Gurvinder Kaur
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Medical Education, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kandis Wright
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Payal Mital
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Taylor Hibler
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jonathan M Miranda
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Lea Ann Thompson
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Katelyn Halley
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jannette M Dufour
- Department of Cell Biology and Biochemistry, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Medical Education, 12343Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
47
|
Wang E, Chen H, Sun B, Wang H, Qu H, Liu Y, Sun X, Qu J, Fang Z, Tian L, Zeng Y, Huang S, Hakonarson H, Liu Z. Serum levels of the IgA isotype switch factor TGF-β1 are elevated in patients with COVID-19. FEBS Lett 2021; 595:1819-1824. [PMID: 33961290 PMCID: PMC8209884 DOI: 10.1002/1873-3468.14104] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/25/2021] [Accepted: 04/30/2021] [Indexed: 01/27/2023]
Abstract
We previously observed enhanced immunoglobulin A (IgA) responses in severe COVID-19, which might confer damaging effects. Given the important role of IgA in immune and inflammatory responses, the aim of this study was to investigate the dynamic response of the IgA isotype switch factor TGF-β1 in COVID-19 patients. We observed, in a total of 153 COVID-19 patients, that the serum levels of TGF-β1 were increased significantly at the early and middle stages of COVID-19, and correlated with the levels of SARS-CoV-2-specific IgA, as well as with the APACHE II score in patients with severe disease. In view of the genetic association of the TGF-β1 activator THBS3 with severe COVID-19 identified by the COVID-19 Host Genetics Initiative, this study suggests TGF-β1 may play a key role in COVID-19.
Collapse
Affiliation(s)
- Er‐yi Wang
- Shenzhen Key Laboratory of Allergy & ImmunologyState Key Laboratory of Respiratory Disease for Allergy at Shenzhen UniversityShenzhen University School of MedicineChina
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityChina
| | - Hao Chen
- Department of Allergy, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory DiseaseFirst Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Bao‐qing Sun
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthFirst Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Hui Wang
- Department of Medical LaboratoryThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hui‐Qi Qu
- Center for Applied GenomicsThe Children’s Hospital of PhiladelphiaPAUSA
| | - Yichuan Liu
- Center for Applied GenomicsThe Children’s Hospital of PhiladelphiaPAUSA
| | - Xi‐zhuo Sun
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityChina
| | - Jingchun Qu
- Center for Applied GenomicsThe Children’s Hospital of PhiladelphiaPAUSA
| | - Zhang‐fu Fang
- Shenzhen Key Laboratory of Allergy & ImmunologyState Key Laboratory of Respiratory Disease for Allergy at Shenzhen UniversityShenzhen University School of MedicineChina
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityChina
| | - Lifeng Tian
- Center for Applied GenomicsThe Children’s Hospital of PhiladelphiaPAUSA
| | - Yi‐feng Zeng
- State Key Laboratory of Respiratory DiseaseNational Clinical Research Center for Respiratory DiseaseGuangzhou Institute of Respiratory HealthFirst Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Shau‐Ku Huang
- Shenzhen Key Laboratory of Allergy & ImmunologyState Key Laboratory of Respiratory Disease for Allergy at Shenzhen UniversityShenzhen University School of MedicineChina
- National Health Research InstitutesMiaoliTaiwan
| | - Hakon Hakonarson
- Center for Applied GenomicsThe Children’s Hospital of PhiladelphiaPAUSA
- Division of Human Genetics & Division of Pulmonary MedicineChildren’s Hospital of PhiladelphiaPAUSA
| | - Zhi‐gang Liu
- Shenzhen Key Laboratory of Allergy & ImmunologyState Key Laboratory of Respiratory Disease for Allergy at Shenzhen UniversityShenzhen University School of MedicineChina
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityChina
| |
Collapse
|
48
|
Aubert A, Mercier-Gouy P, Aguero S, Berthier L, Liot S, Prigent L, Alcaraz LB, Verrier B, Terreux R, Moali C, Lambert E, Valcourt U. Latent TGF-β Activation Is a Hallmark of the Tenascin Family. Front Immunol 2021; 12:613438. [PMID: 34054795 PMCID: PMC8155481 DOI: 10.3389/fimmu.2021.613438] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor-β (TGF-β) isoforms are secreted as inactive complexes formed through non-covalent interactions between bioactive TGF-β entities and their N-terminal pro-domains called latency-associated peptides (LAP). Extracellular activation of latent TGF-β within this complex is a crucial step in the regulation of TGF-β activity for tissue homeostasis and immune cell function. We previously showed that the matrix glycoprotein Tenascin-X (TN-X) interacted with the small latent TGF-β complex and triggered the activation of the latent cytokine into a bioactive TGF-β. This activation most likely occurs through a conformational change within the latent TGF-β complex and requires the C-terminal fibrinogen-like (FBG) domain of the glycoprotein. As the FBG-like domain is highly conserved among the Tenascin family members, we hypothesized that Tenascin-C (TN-C), Tenascin-R (TN-R) and Tenascin-W (TN-W) might share with TN-X the ability to regulate TGF-β bioavailability through their C-terminal domain. Here, we demonstrate that purified recombinant full-length Tenascins associate with the small latent TGF-β complex through their FBG-like domains. This association promotes activation of the latent cytokine and subsequent TGF-β cell responses in mammary epithelial cells, such as cytostasis and epithelial-to-mesenchymal transition (EMT). Considering the pleiotropic role of TGF-β in numerous physiological and pathological contexts, our data indicate a novel common function for the Tenascin family in the regulation of tissue homeostasis under healthy and pathological conditions.
Collapse
Affiliation(s)
- Alexandre Aubert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Perrine Mercier-Gouy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Stéphanie Aguero
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laurent Berthier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Sophie Liot
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laura Prigent
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Lindsay B Alcaraz
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut du Cancer de Montpellier (ICM), Montpellier, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Raphaël Terreux
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Catherine Moali
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Ulrich Valcourt
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| |
Collapse
|
49
|
Karolczak K, Watala C. Blood Platelets as an Important but Underrated Circulating Source of TGFβ. Int J Mol Sci 2021; 22:ijms22094492. [PMID: 33925804 PMCID: PMC8123509 DOI: 10.3390/ijms22094492] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/17/2021] [Accepted: 04/24/2021] [Indexed: 12/13/2022] Open
Abstract
When treating diseases related primarily to tissue remodeling and fibrosis, it is desirable to regulate TGFβ concentration and modulate its biological effects. The highest cellular concentrations of TGFβ are found in platelets, with about 40% of all TGFβ found in peripheral blood plasma being secreted by them. Therefore, an understanding of the mechanisms of TGFβ secretion from platelets may be of key importance for medicine. Unfortunately, despite the finding that platelets are an important regulator of TGFβ levels, little research has been carried out into the development of platelet-directed therapies that might modulate the TGFβ-dependent processes. Nevertheless, there are some very encouraging reports suggesting that platelet TGFβ may be specifically involved in cardiovascular diseases, liver fibrosis, tumour metastasis, cerebral malaria and in the regulation of inflammatory cell functions. The purpose of this review is to briefly summarize these few, extremely encouraging reports to indicate the state of current knowledge in this topic. It also attempts to better characterize the influence of TGFβ on platelet activation and reactivity, and its shaping of the roles of blood platelets in haemostasis and thrombosis.
Collapse
|
50
|
Wilson SE. TGF beta -1, -2 and -3 in the modulation of fibrosis in the cornea and other organs. Exp Eye Res 2021; 207:108594. [PMID: 33894227 DOI: 10.1016/j.exer.2021.108594] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/10/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
The TGF beta-1, -2 and -3 isoforms are transcribed from different genes but bind to the same receptors and signal through the same canonical and non-canonical signal transduction pathways. There are numerous regulatory mechanisms controlling the action of each isoform that include the organ-specific cells producing latent TGF beta growth factors, multiple effectors that activate the isoforms, ECM-associated SLRPs and basement membrane components that modulate the activity and localization of the isoforms, other interactive cytokine-growth factor receptor systems, such as PDGF and CTGF, TGF beta receptor expression on target cells, including myofibroblast precursors, receptor binding competition, positive and negative signal transduction effectors, and transcription and translational regulatory mechanisms. While there has long been the view that TGF beta-1and TGF beta-2 are pro-fibrotic, while TGF beta-3 is anti-fibrotic, this review suggests that view is too simplistic, at least in adult tissues, since TGF beta-3 shares far more similarities in its modulation of fibrotic gene expression with TGF beta-1 and TGF beta-2, than it does differences, and often the differences are subtle. Rather, TGF beta-3 should be seen as a fibro-modulatory partner to the other two isoforms that modulates a nuanced and better controlled response to injury. The complex interplay between the three isoforms and numerous interactive proteins, in the context of the cellular milieu, controls regenerative non-fibrotic vs. fibrotic healing in a response to injury in a particular organ, as well as the resolution of fibrosis, when that occurs.
Collapse
Affiliation(s)
- Steven E Wilson
- The Cole Eye Institute, The Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|