1
|
Zhu H, Liu F, Liao Y, Li H, Gao K, Liang X, Jiang H, Chen F, Wu J, Wang Q, Wang Y, Shuai X, Yi X. Biomimetic nanostructural materials based on placental amniotic membrane-derived nanofibers for self-healing and anti-adhesion during cesarean section. Biomaterials 2025; 317:123081. [PMID: 39787897 DOI: 10.1016/j.biomaterials.2024.123081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/05/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
Cesarean section (CS) is highly prevalent surgery among females. However, current absorbable anti-adhesion membranes used clinically can partially prevent postoperative adhesions but show limited efficacy in tissue regeneration, leaving post-cesarean women at risk for severe complications including cesarean scar pregnancy, placenta previa, and uterine rupture. Herein, we designed a fully amniotic membrane (AM)-derived biomimetic nanostructural materials (AM-BNMs) as an anti-adhesion barrier, and validated its therapeutic efficacy in a rat CS model. The biomaterial consisted of AM-extracellular matrix (ECM) nanofibers, enriched with hemostatic proteins (collagen, S100A8, S100A9, etc.), carrying AM mesenchymal stem cells (MSCs)-secretome that exhibited significantly elevated levels of pro-regenerative factors (miR-302a-3p, angiogenin, VEGF, etc.) compared to endogenous secretion. The reconstituted AM-BNMs demonstrated synergistic effects at CS wounds, effectively preventing adhesion formation while promoting hemostasis and tissue regeneration. In summary, this readily accessible human-derived biomaterial shows promising potential in preventing adhesion-related complications and enhancing uterine wound healing, thereby promoting female reproductive health.
Collapse
Affiliation(s)
- Honglei Zhu
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Fenghua Liu
- Center for Reproductive Medicine, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Yuru Liao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Huayan Li
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kunjie Gao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Xiaomei Liang
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Haoyuan Jiang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Feng Chen
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jianwei Wu
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qian Wang
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yifeng Wang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Xiao Yi
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, 511462, China.
| |
Collapse
|
2
|
Chakraborty S, Dutta A, Roy A, Joshi A, Basak T. The theatrics of collagens in the myocardium: the supreme architect of the fibrotic heart. Am J Physiol Cell Physiol 2025; 328:C1893-C1920. [PMID: 40257077 DOI: 10.1152/ajpcell.01043.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/24/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Heart failure (HF) mediated by cardiac fibrosis (CF) is characterized by an excessive accumulation of collagen-based extracellular matrix (ECM) in the myocardium. CF is a common pathophysiological condition in many heart diseases and can be distinctly categorized into two types: replacement and interstitial. In ischemic heart diseases, sudden loss of cardiomyocytes leads to the replacement of CF to prevent ventricular rupture. In contrast, excessive collagen deposition in the interstitial space between cardiomyocytes (often in response to pressure overload, chronic cardiac stress, hypertension, etc.) is termed interstitial CF. The progression of HF due to cardiac fibrosis is mainly driven by compromised diastolic function, resulting from increased stiffness of the heart wall muscle due to collagen-based scar formation. Increased myocardial stiffness is primarily catalyzed by the differential cross linking of deposited collagens forming the scar in the fibrotic heart. Although collagen deposition remained a hallmark of fibrosis, the pathophysiological progression due to biochemical alterations and mechanistic discrepancy of collagens across cardiac fibrosis subtypes remains elusive. With the advent of next-generation RNA sequencing and high-resolution mass spectrometry, mechanistic insights into collagen-mediated scar maturation have gained impetus. A deeper understanding of the spatiocellular transcriptional heterogeneity and site-specific collagen posttranslational modifications (PTMs) in maneuvering ECM remodeling is gaining attention. The unexplored mechanisms of posttranslational modifications and subsequent collagen cross linking in various cardiac fibrosis may provide the prime target for therapeutic interventions. This review comprehensively summarizes the detailed pattern, role, signaling, and mechanical contributions of different collagens and their PTMs, including cross-linking patterns as newer therapeutic regimens during cardiac fibrosis.
Collapse
Affiliation(s)
- Sanchari Chakraborty
- School of Biosciences and Bioengineering, Indian Institute of Technology - Mandi, Mandi, Himachal Pradesh, India
| | - Abhi Dutta
- School of Biosciences and Bioengineering, Indian Institute of Technology - Mandi, Mandi, Himachal Pradesh, India
| | - Antara Roy
- School of Biosciences and Bioengineering, Indian Institute of Technology - Mandi, Mandi, Himachal Pradesh, India
| | - Ashutosh Joshi
- School of Biosciences and Bioengineering, Indian Institute of Technology - Mandi, Mandi, Himachal Pradesh, India
| | - Trayambak Basak
- School of Biosciences and Bioengineering, Indian Institute of Technology - Mandi, Mandi, Himachal Pradesh, India
| |
Collapse
|
3
|
Wang C, Fan M, Heo SC, Adams SM, Li T, Liu Y, Li Q, Loebel C, Burdick JA, Lu XL, Birk DE, Alisafaei F, Mauck RL, Han L. Structure, Mechanics, and Mechanobiology of Fibrocartilage Pericellular Matrix Mediated by Type V Collagen. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e14750. [PMID: 40407177 DOI: 10.1002/advs.202414750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/01/2025] [Indexed: 05/28/2025]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding cells in various tissues, regulating matrix turnover, cell-matrix interactions, and disease. This study elucidates the structure-mechanical properties and mechanobiology of the PCM in fibrocartilage, using the murine meniscus as the model. The fibrocartilage PCM is comprised of thin, randomly oriented collagen fibrils that entrap proteoglycans, contrasting with the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). Compared to the ECM, the PCM exhibits lower modulus and greater isotropy, but has similar relative viscoelastic properties. In Col5a1+/- menisci, the reduction of collagen V results in thicker, more heterogeneous collagen fibrils, reduced modulus, loss of isotropy and faster viscoelastic relaxation in the PCM. Such altered PCM leads to impaired matrix-to-cell strain transmission, and in turn, disrupts mechanotransduction of meniscal cells, as illustrated by reduced calcium signaling activities and alters expression of matrix genes. In vitro, Col5a1+/- cells produce a weakened PCM with inferior properties and reduced protection of cells against tensile stretch. These findings highlight the PCM as a distinctive microstructure in fibrocartilage mechanobiology, underscoring a pivotal role of collagen V in PCM function. Targeting the PCM or its constituents offers potential for improving meniscus regeneration, osteoarthritis intervention and broader fibrocartilage-related therapies.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Su Chin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, 80309, USA
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA, 19104, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
4
|
Jiang H, Lu X, Bu T, Yang X, Li X, Ren X, Xu X, Fan C, He J, Zhang X, Song W, Tian W, Xu B. Mechanics Mediated Semi-Convertible Hydrogel Enabled Sustained Drug Release. Adv Healthc Mater 2025:e2500845. [PMID: 40400499 DOI: 10.1002/adhm.202500845] [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: 02/15/2025] [Revised: 05/09/2025] [Indexed: 05/23/2025]
Abstract
The dynamic mechanic environment surrounding the wound may retard wound healing, and even lead to an exacerbation of inflammation and scar. How to actively promote wound healing under a dynamic mechanical environment during human motion is still a long-standing challenge. Therefore, a mechanics mediated semi-convertible hydrogel (MechSCH) loaded with drug is proposed in this study employing the synergistic interaction between mechanics mediated supramolecular non-covalent networks and polyvinyl alcohol/Gelatin polymer networks for enhancing dynamic wound healing. The formed MechSCH exhibits a partial gel-sol transition even under a shear stress of ≈9.04 Pa that is satisfied with most tissues or organs' stress. The sustained release of encapsulated drugs would be efficiently compared with the mechanics of non-sensitive polyvinyl alcohol/Gelatin hydrogel. The loaded platelet-derived growth factor (PDGF) of the MechSCH exhibited a rapid onset of therapeutic effect in a mice dorsal full-thickness dermal wound model, which demonstrated sustaining drug release through mechanics of skin tension at the wound site, along with alleviating the inflammation and promoting rapid vascular regeneration. This mechanics mediated semi-convertible hydrogel presents potential clinical applications for the dynamic management of chronic wounds.
Collapse
Affiliation(s)
- Hongyue Jiang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Xing Lu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, P. R. China
- Nanhu Laboratory, Jiaxing, 314051, P. R. China
| | - Tianshi Bu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Xuhao Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Xiang Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Xue Ren
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Xinyi Xu
- Nanhu Laboratory, Jiaxing, 314051, P. R. China
| | | | - Jingxuan He
- Nanhu Laboratory, Jiaxing, 314051, P. R. China
| | - Xiaopeng Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, P. R. China
- Nanhu Laboratory, Jiaxing, 314051, P. R. China
| | - Wenlong Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Wenjing Tian
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Bin Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| |
Collapse
|
5
|
Timmer LT, den Hertog E, Versteeg D, Post H, Verdonschot JAJ, Monshouwer-Kloots J, Kyriakopoulou E, Perini I, Koopmans T, van der Kraak P, Zentilin L, Heymans SRB, Vink A, Giacca M, Heck AJR, van Rooij E. Cardiomyocyte SORBS2 expression increases in heart failure and regulates integrin interactions and extracellular matrix composition. Cardiovasc Res 2025; 121:585-600. [PMID: 39957251 PMCID: PMC12054630 DOI: 10.1093/cvr/cvaf021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/26/2024] [Accepted: 12/05/2024] [Indexed: 02/18/2025] Open
Abstract
AIMS In this study, we aimed to uncover genes associated with stressed cardiomyocytes by combining single-cell transcriptomic data sets from failing cardiac tissue from both humans and mice. METHODS AND RESULTS Our bioinformatic analysis identified SORBS2 as conserved NPPA-correlated gene. Using mouse models and cardiac tissue from human heart failure patients, we demonstrated that SORBS2 expression is consistently increased during pathological remodelling, correlates to disease severity, and is regulated by GATA4. By affinity purification mass spectrometry, we showed SORBS2 to interact with the integrin-cytoskeleton connections. Cardiomyocyte-specific genetic loss of Sorbs2 in adult mice changed integrin interactions, indicated by the increased expression of several integrins and altered extracellular matrix components connecting to these integrins, leading to an exacerbated fibrotic response during pathological remodelling. CONCLUSION Sorbs2 is a cardiomyocyte-enriched gene that is increased during progression to heart failure in a GATA4-dependent manner and correlates to phenotypical hallmarks of cardiac failure. Our data indicate SORBS2 to function as a crucial regulator of integrin interactions and cardiac fibrosis.
Collapse
Affiliation(s)
- Louk T Timmer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Elvira den Hertog
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Job A J Verdonschot
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart)
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Eirini Kyriakopoulou
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Ilaria Perini
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Petra van der Kraak
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Stephane R B Heymans
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, King’s College London, London, UK
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
6
|
Su L, Sun Q, Li Y, Alvarez JF, Tao B, Zhang G, Gu Y, Hanudel MR, Espinoza A, Zhang L, Pan C, Hilser JR, Hartiala JA, Li S, Pellegrini M, Allayee H, Lusis AJ, Deb A. Collagen V regulates renal function after kidney injury and can be pharmacologically targeted to enhance kidney repair in mice. Sci Transl Med 2025; 17:eads7714. [PMID: 40203084 DOI: 10.1126/scitranslmed.ads7714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/03/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
Kidney fibrosis determines clinical outcomes in individuals with chronic kidney disease (CKD). The stoichiometric ratio of collagens in renal scar differs from that of healthy kidney extracellular matrix (ECM), but the functional importance of altered collagen types in injured kidneys remains unclear. Using human population studies, we show that circulating protein and renal mRNA amounts of collagen V A1 (COL5A1) exhibited associations with kidney disease and incident CKD risk. We show that Col5a1 regulates the degree of postinjury fibrosis and renal function. Mice with conditionally knocked out Col5a1 (Col5a1 CKO) exhibited decreased renal function and greater renal fibrosis after dietary adenine- or ureteric obstruction-mediated kidney injury. Renal fibroblasts in Col5a1 CKO animals up-regulated the profibrotic αvβ3 integrin. Inhibition of αvβ3 signaling with a small molecule, cilengitide, rescued postinjury renal function in Col5a1 CKO animals. Using the hybrid mouse diversity panel that comprises 100 diverse inbred strains of mice, we observed that gene expression of Col5a1 after injury exhibited genetic variation across 100 strains. Strains with low Col5a1 expression after injury exhibited worse renal function compared with animals that had higher degrees of expression. We next measured Col5a1 expression in peripheral blood mononuclear cells in mice to identify nonresponder strains that did not have increased Col5a1 expression after kidney injury. We observed that administration of cilengitide in nonresponder strains significantly rescued postinjury renal fibrosis and function. These studies point to the feasibility of precision medicine approaches to target Col5a1 for enhancing renal repair.
Collapse
Affiliation(s)
- Lianjiu Su
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qihao Sun
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yusheng Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Juan Felipe Alvarez
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Tao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yiqian Gu
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark R Hanudel
- Department of Pediatric Nephrology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Espinoza
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linlin Zhang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - James R Hilser
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jaana A Hartiala
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hooman Allayee
- Departments of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Departments of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Koopmans T, van Rooij E. Molecular gatekeepers of endogenous adult mammalian cardiomyocyte proliferation. Nat Rev Cardiol 2025:10.1038/s41569-025-01145-y. [PMID: 40195566 DOI: 10.1038/s41569-025-01145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
Irreversible cardiac fibrosis, cardiomyocyte death and chronic cardiac dysfunction after myocardial infarction pose a substantial global health-care challenge, with no curative treatments available. To regenerate the injured heart, cardiomyocytes must proliferate to replace lost myocardial tissue - a capability that adult mammals have largely forfeited to adapt to the demanding conditions of life. Using various preclinical models, our understanding of cardiomyocyte proliferation has progressed remarkably, leading to the successful reactivation of cell cycle induction in adult animals, with functional recovery after cardiac injury. Central to this success is the targeting of key pathways and structures that drive cardiomyocyte maturation after birth - nucleation and ploidy, sarcomere structure, developmental signalling, chromatin and epigenetic regulation, the microenvironment and metabolic maturation - forming a complex regulatory framework that allows efficient cellular contraction but restricts cardiomyocyte proliferation. In this Review, we explore the molecular pathways underlying these core mechanisms and how their manipulation can reactivate the cell cycle in cardiomyocytes, potentially contributing to cardiac repair.
Collapse
Affiliation(s)
- Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands.
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
8
|
Lu J, Wang Z, Zhang L. Single-cell transcriptome analysis revealing mechanotransduction via the Hippo/YAP pathway in promoting fibroblast-to-myofibroblast transition and idiopathic pulmonary fibrosis development. Gene 2025; 943:149271. [PMID: 39855369 DOI: 10.1016/j.gene.2025.149271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is an irreversible and fatal interstitial lung disease, characterized by excessive extracellular matrix (ECM) secretion that disrupts normal alveolar structure. This study aims to explore the potential molecular mechanisms underlying the promotion of IPF development. METHODS Firstly, we compared the transcriptome and single-cell sequencing data from lung tissue samples of patients with IPF and healthy individuals. Subsequently, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on the differentially expressed genes (DEGs). Furthermore, we employed sodium alginate hydrogels with varying degrees of crosslinking to provide differential mechanical stress, mimicking the mechanical microenvironment in vivo during lung fibrosis. On this basis, we examined cytoskeletal remodeling in fibroblasts MRC-5, mRNA expression of multiple related genes, immunofluorescence localization, and cellular proliferation capacity. RESULTS Bioinformatics analysis revealed a series of DEGs associated with IPF. Further functional and pathway enrichment analyses indicated that these DEGs were primarily enriched in ECM-related biological processes. Single-cell sequencing data revealed that fibroblasts and myofibroblasts are the main contributors to excessive ECM secretion and suggested activation of mechanotransduction and the Hippo/YAP signaling pathway in myofibroblasts. Cellular experiments demonstrated that sodium alginate hydrogels with different stiffness can simulate different mechanical stress environments, thereby affecting cytoskeletal rearrangement and Hippo/YAP pathway activity in MRC-5 lung fibroblasts. Notably, high levels of mechanical stress promoted YAP nuclear translocation, increased expression of type I collagen and α-SMA, and enhanced proliferative capacity. Additionally, we also found that fibroblasts primarily participate in mechanotransduction through the Rho/ROCK and Integrin/FAK pathways under high mechanical stress conditions, ultimately upregulating the gene expression of CCNE1/2, CTGF, and FGF1. CONCLUSION Our study uncovers the crucial role of cytoskeletal mechanotransduction in myofibroblast transformation and IPF development through activation of the Hippo/YAP pathway, providing new insights into understanding the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China.
| | - Zhenhua Wang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| | - Liguo Zhang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| |
Collapse
|
9
|
Chirivì M, Maiullari F, Milan M, Ceraolo MG, Fratini N, Fasciani A, Bousselmi S, Stirm M, Scalera F, Gervaso F, Villa M, Viganò R, Brambilla F, Mauri P, De Falco E, Silvestre DD, Costantini M, Wolf E, Bearzi C, Rizzi R. Mimicking the Dystrophic Cardiac Extracellular Environment through DystroGel. Adv Healthc Mater 2025; 14:e2404251. [PMID: 39962811 PMCID: PMC11973943 DOI: 10.1002/adhm.202404251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/03/2025] [Indexed: 04/08/2025]
Abstract
Advances in understanding the mechanisms behind genetic diseases like Duchenne muscular dystrophy (DMD) underscore the critical role of the extracellular matrix (ECM) composition in disease progression. Effective in vitro models must replicate the intercellular relationships and physicochemical properties of native ECM to fully capture disease-specific characteristics. Although recent biomaterials support the in vitro biofabrication of pathophysiological environments, they often lack disease-specific ECM features. In this study, DystroGel, a hydrogel derived from the cardiac ECM of a porcine DMD model, replicates the distinct molecular composition of dystrophic cardiac tissue for the first time. The findings indicate that the dystrophic ECM matrix exhibits a unique protein profile, impacting cellular processes critical to DMD pathology. This work demonstrates the importance of using a 3D substrate that recreates intercellular dynamics within a defined pathological environment, enhancing the ability to model genetic disorders and providing a valuable tool for advancing personalized therapeutic strategies.
Collapse
Affiliation(s)
- Maila Chirivì
- Department of Molecular MedicineSapienza UniversityViale Regina Elena, 324Rome00161Italy
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
| | - Fabio Maiullari
- Ph.D. Program in Cellular and Molecular BiologyDepartment of BiologyUniversity of Rome “Tor Vergata”Via della Ricerca Scientifica, 1Rome00133Italy
| | - Marika Milan
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
| | - Maria Grazia Ceraolo
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
| | - Nicole Fratini
- Department of Molecular MedicineSapienza UniversityViale Regina Elena, 324Rome00161Italy
| | - Alessandra Fasciani
- Fondazione Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi”Via Francesco Sforza, 35Milan20122Italy
| | - Salma Bousselmi
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoVia Francesco Sforza, 35Milan20122Italy
- Ph.D. Program in Cellular and Molecular BiologyDepartment of BiologyUniversity of Rome “Tor Vergata”Via della Ricerca Scientifica, 1Rome00133Italy
| | - Michael Stirm
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU Munich81377MunichGermany
- Center for Innovative Medical Models (CiMM)Department of Veterinary SciencesLMU Munich85764OberschleißheimGermany
| | - Francesca Scalera
- Institute of NanotechnologyNational Research Councilc/o Campus Ecoteknevia MonteroniLecce73100Italy
| | - Francesca Gervaso
- Institute of NanotechnologyNational Research Councilc/o Campus Ecoteknevia MonteroniLecce73100Italy
| | - Michela Villa
- Fondazione Istituto Nazionale di Genetica Molecolare “Romeo ed Enrica Invernizzi”Via Francesco Sforza, 35Milan20122Italy
- Department of BiosciencesUniversity of MilanVia Celoria, 26Milan20133Italy
| | - Raffaello Viganò
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Francesca Brambilla
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Pierluigi Mauri
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Elena De Falco
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Dario Di Silvestre
- Department of BiosciencesUniversity of MilanVia Celoria, 26Milan20133Italy
| | - Marco Costantini
- Institute of Physical Chemistry – Polish Academy of SciencesMarcina Kasprzaka 44/52Warsaw01–224Poland
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and BiotechnologyGene Center and Department of Veterinary SciencesLMU Munich81377MunichGermany
| | - Claudia Bearzi
- Institute for Biomedical TechnologiesNational Research CouncilVia Fratelli Cervi, 93, SegrateMilan20054Italy
| | - Roberto Rizzi
- Department of Medical‐Surgical Sciences and BiotechnologiesSapienza University of RomeC.so della Repubblica 79Latina04100Italy
| |
Collapse
|
10
|
Matias Z, Lopes CS, Santos NC, Carvalho FA. Nanotechnology meets medicine: applications of atomic force microscopy in disease. Biophys Rev 2025; 17:359-384. [PMID: 40376402 PMCID: PMC12075069 DOI: 10.1007/s12551-025-01306-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 05/18/2025] Open
Abstract
Atomic force microscopy (AFM) is a scanning imaging technique able to work at the nanoscale. It uses a cantilever with a tip to move across samples' surface and a laser to measure the cantilever bending, enabling the assessment of interaction forces between tip and sample and creating a three-dimensional visual representation of its surface. AFM has been gaining notoriety in the biomedical field due to its high-resolution images, as well as due to its ability to measure the inter- and intramolecular interaction forces involved in the pathophysiology of many diseases. Here, we highlight some of the current applications of AFM in the biomedical field. First, a brief overview of the AFM technique is presented. This theoretical framework is then used to link AFM to its novel translational applications, handling broad clinical questions in different areas, such as infectious diseases, cardiovascular diseases, cancer, and neurodegenerative diseases. Morphological and nanomechanical characteristics such as cell height, volume, stiffness, and adhesion forces may serve as novel parameters used to tailor patient care through nanodiagnostics, individualized risk stratification, and therapeutic monitoring. Despite an increasing development of AFM biomedical research with patient cells, showing its unique capabilities in terms of resolution, speed, and accuracy, there is a notable need for applied AFM research in clinical settings. More translational research with AFM may provide new grounds for the valuable collaboration between biomedical researchers and healthcare professionals.
Collapse
Affiliation(s)
- Zita Matias
- Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- ULSLO – Unidade Local de Saúde Lisboa Ocidental, Lisbon, Portugal
| | - Catarina S. Lopes
- GIMM – Gulbenkian Institute for Molecular Medicine, Av. Prof. Egas Moniz, 1649-035 Lisbon, Portugal
| | - Nuno C. Santos
- Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- GIMM – Gulbenkian Institute for Molecular Medicine, Av. Prof. Egas Moniz, 1649-035 Lisbon, Portugal
| | - Filomena A. Carvalho
- Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
- GIMM – Gulbenkian Institute for Molecular Medicine, Av. Prof. Egas Moniz, 1649-035 Lisbon, Portugal
| |
Collapse
|
11
|
Ye W, Meng X, Xu S. [Research progress on collagen secretion mechanisms in scarring]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2025; 54:266-278. [PMID: 40194913 PMCID: PMC12062945 DOI: 10.3724/zdxbyxb-2024-0535] [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: 09/24/2024] [Revised: 12/12/2024] [Accepted: 02/22/2025] [Indexed: 04/09/2025]
Abstract
Scar formation is characterized by dynamic alterations in collagen secretion, which critically determine scar morphology and pathological progression. In fibroblasts, collagen secretion is initiated through the activation of cytokine- and integrin-mediated signaling pathways, which promote collagen gene transcription. The procollagen polypeptide α chains undergo extensive post-translational modifications, including hydroxylation and glycosylation, within the endoplasmic reticulum (ER), followed by folding and assembly into triple-helical procollagen. Subsequent intracellular trafficking involves the sequential transport of procollagen through the ER, Golgi apparatus, and plasma membrane, accompanied by further structural refinements prior to extracellular secretion. Once secreted, procollagen is enzymatically processed to form mature collagen fibrils, which drive scar tissue remodeling. Recent advances in elucidating regulation of collagen secretion have identified pivotal molecular targets, such as transforming growth factor-beta 1 (TGF-β1), prolyl 4-hydroxylase (P4H), heat shock protein 47 (HSP47), and transport and Golgi organization protein 1 (TANGO1), providing novel therapeutic strategies to mitigate pathological scar hyperplasia and improve regenerative outcomes. This review provides a comprehensive analysis of the molecular mechanisms governing collagen secretion during scar formation, with emphasis on signaling cascades, procollagen biosynthesis, intracellular transport dynamics, and post-translational modifications, thereby offering a framework for developing targeted anti-scar therapies.
Collapse
Affiliation(s)
- Wenkai Ye
- Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Xinan Meng
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, International Institutes of Medicine, Zhejiang University, Center for Membrane Receptors and Brain Medicine, International School of Medicine, Zhejiang University, Yiwu 322000, Zhejiang Province, China
| | - Suhong Xu
- Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
12
|
Wang Y, Meng D, Shi X, Hou Y, Zang S, Chen L, Spanos M, Li G, Cretoiu D, Zhou Q, Xiao J. Injectable hydrogel with miR-222-engineered extracellular vesicles ameliorates myocardial ischemic reperfusion injury via mechanotransduction. Cell Rep Med 2025; 6:101987. [PMID: 40037358 PMCID: PMC11970392 DOI: 10.1016/j.xcrm.2025.101987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/05/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025]
Abstract
Cardiac ischemic reperfusion injury (IRI) significantly exacerbates cardiac dysfunction and heart failure, causing high mortality. Despite the severity of IRI, effective therapeutic strategies remain elusive. Acellular cardiac patches have shown considerable efficacy in delivering therapeutics directly to cardiac tissues. Herein, we develop injectable GelMA (GEL) hydrogels with controlled mechanical properties. Targeting miR-222-engineered extracellular vesicles (TeEVs), tailored with cardiac-ischemia-targeting peptides (CTPs), are developed as ischemic TeEV therapeutics. These TeEVs are encapsulated within mechanical hydrogels to create injectable TeEV-loaded cardiac patches, enabling minimal invasiveness to attenuate IRI. The injectable patches facilitate the precise targeting of TeEVs for the efficient rescue of damaged cells. Persistent delivery of TeEVs into the infarcted region alleviates acute IRI and mitigated remodeling post IRI. This is linked to focal adhesion activation, cytoskeleton force enhancement, and nuclear force-sensing preservation. These findings may pave the way for force-sensing approaches to cardiac therapy using bioengineered therapeutic patches.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Danni Meng
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xiaohui Shi
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Yan Hou
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
| | - Shihui Zang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China
| | - Lei Chen
- Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dragos Cretoiu
- Department of Medical Genetics, Carol Davila University of Medicine and Pharmacy, 020031 Bucharest, Romania; Materno-Fetal Assistance Excellence Unit, Alessandrescu-Rusescu National Institute for Mother and Child Health, 011062 Bucharest, Romania
| | - Qiulian Zhou
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong) and School of Life Science, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
13
|
Sun W, Shahrajabian MH, Ma K, Wang S. Advances in Molecular Function and Recombinant Expression of Human Collagen. Pharmaceuticals (Basel) 2025; 18:430. [PMID: 40143206 PMCID: PMC11945623 DOI: 10.3390/ph18030430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/02/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Collagen is the main protein found in skin, bone, cartilage, ligaments, tendons and connective tissue, and it can exhibit properties ranging from compliant to rigid or form gradients between these states. The collagen family comprises 28 members, each containing at least one triple-helical domain. These proteins play critical roles in maintaining mechanical characteristics, tissue organization, and structural integrity. Collagens regulate cellular processes such as proliferation, migration, and differentiation through interactions with cell surface receptors. Fibrillar collagens, the most abundant extracellular matrix (ECM) proteins, provide organs and tissues with structural stability and connectivity. In the mammalian myocardial interstitium, types I and III collagens are predominant: collagen I is found in organs, tendons, and bones; collagen II is found in cartilage; collagen III is found in reticular fibers; collagen IV is found in basement membranes; and collagen V is found in nails and hair. Recombinant human collagens, particularly in sponge-like porous formats combined with bone morphogenetic proteins, serve as effective scaffolds for bone repair. Due to their biocompatibility and low immunogenicity, collagens are pivotal in tissue engineering applications for skin, bone, and wound regeneration. Recombinant technology enables the production of triple-helical collagens with amino acid sequences identical to human tissue-derived collagens. This review summarizes recent advances in the molecular functions and recombinant expression of human collagens, with a focus on their biomedical applications.
Collapse
Affiliation(s)
- Wenli Sun
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100086, China;
| | - Mohamad Hesam Shahrajabian
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100086, China;
| | - Kun Ma
- Hantide Biomedical Group Co., Ltd., Zibo 256300, China;
| | - Shubin Wang
- Hantide Biomedical Group Co., Ltd., Zibo 256300, China;
| |
Collapse
|
14
|
Karsdal M, Cox TR, Parker AL, Willumsen N, Sand JMB, Jenkins G, Hansen HH, Oldenburger A, Geillinger-Kaestle KE, Larsen AT, Black D, Genovese F, Eckersley A, Heinz A, Nyström A, Holm Nielsen S, Bennink L, Johannsson L, Bay-Jensen AC, Orange DE, Friedman S, Røpke M, Fiore V, Schuppan D, Rieder F, Simona B, Borthwick L, Skarsfeldt M, Wennbo H, Thakker P, Stoffel R, Clarke GW, Kalluri R, Ruane D, Zannad F, Mortensen JH, Sinkeviciute D, Sundberg F, Coseno M, Thudium C, Croft AP, Khanna D, Cooreman M, Broermann A, Leeming DJ, Mobasheri A, Ricard-Blum S. Advances in Extracellular Matrix-Associated Diagnostics and Therapeutics. J Clin Med 2025; 14:1856. [PMID: 40142664 PMCID: PMC11943371 DOI: 10.3390/jcm14061856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 03/28/2025] Open
Abstract
The extracellular matrix (ECM) is the common denominator of more than 50 chronic diseases. Some of these chronic pathologies lead to enhanced tissue formation and deposition, whereas others are associated with increased tissue degradation, and some exhibit a combination of both, leading to severe tissue alterations. To develop effective therapies for diseases affecting the lung, liver, kidney, skin, intestine, musculoskeletal system, heart, and solid tumors, we need to modulate the ECM's composition to restore its organization and function. Across diverse organ diseases, there are common denominators and distinguishing factors in this fibroinflammatory axis, which may be used to foster new insights into drug development across disease indications. The 2nd Extracellular Matrix Pharmacology Congress took place in Copenhagen, Denmark, from 17 to 19 June 2024 and was hosted by the International Society of Extracellular Matrix Pharmacology. The event was attended by 450 participants from 35 countries, among whom were prominent scientists who brought together state-of-the-art research on organ diseases and asked important questions to facilitate drug development. We highlight key aspects of the ECM in the liver, kidney, skin, intestine, musculoskeletal system, lungs, and solid tumors to advance our understanding of the ECM and its central targets in drug development. We also highlight key advances in the tools and technology that enable this drug development, thereby supporting the ECM.
Collapse
Affiliation(s)
- Morten Karsdal
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Thomas R. Cox
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Amelia L. Parker
- Garvan Institute of Medical Research, Sydney 2010, Australia; (T.R.C.); (A.L.P.)
- School of Clinical Medicine, St Vincent’s Clinical Campus, UNSW Medicine & Health, UNSW, Sydney 2010, Australia
| | - Nicholas Willumsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Jannie Marie Bülow Sand
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London SW7 2AZ, UK;
| | | | | | - Kerstin E. Geillinger-Kaestle
- Department of Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Anna Thorsø Larsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | - Federica Genovese
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Alexander Eckersley
- Wellcome Centre for Cell Matrix Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK;
| | - Andrea Heinz
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center—University of Freiburg, 79106 Breisgau, Germany;
| | - Signe Holm Nielsen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | | | | | - Anne-Christine Bay-Jensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dana E. Orange
- Hospital for Special Surgery, The Rockefeller University, New York, NY 10065, USA;
| | - Scott Friedman
- Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA;
| | | | - Vincent Fiore
- Boehringer Ingelheim, 55218 Ingelheim am Rhein, Germany;
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | | | - Lee Borthwick
- FibroFind Ltd., FibroFind Laboratories, Medical School, Newcastle upon Tyne NE2 4HH, UK;
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Mark Skarsfeldt
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Haakan Wennbo
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Paresh Thakker
- Takeda, Translational Medicine Biomarkers Gastrointestinal & Global, Boston, MA 02110, USA; (H.W.); (P.T.)
| | - Ruedi Stoffel
- Roche Diagnostics International Ltd., 6343 Rotkreuz, Switzerland;
| | - Graham W. Clarke
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden;
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College, London E1 9RT, UK
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Darren Ruane
- Janssen Immunology, Translational Sciences and Medicine, La Jolla, CA 92037, USA;
| | - Faiez Zannad
- Division of Heart Failure and Hypertension, and of the Inserm CIC, University of Lorraine, 54000 Metz, France;
| | - Joachim Høg Mortensen
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Dovile Sinkeviciute
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Fred Sundberg
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Molly Coseno
- Sengenics Corporation LLC, Wilmington, DE 19801, USA; (F.S.); (M.C.)
| | - Christian Thudium
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Adam P. Croft
- National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, University of Birmingham, Birmingham B15 2TT, UK;
- Institute of Inflammation and Ageing, Queen Elizabeth Hospital, University of Birmingham, Birmingham B15 2TT, UK
| | - Dinesh Khanna
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | | | - Andre Broermann
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riss, Germany;
| | - Diana Julie Leeming
- Nordic Bioscience, 2730 Herlev, Denmark; (N.W.); (J.M.B.S.); (A.T.L.); (F.G.); (S.H.N.); (A.-C.B.-J.); (J.H.M.); (D.S.); (D.J.L.)
| | - Ali Mobasheri
- Faculty of Medicine, University of Oulu, 90570 Oulu, Finland;
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania
- Faculté de Médecine, Université de Liège, 4000 Liège, Belgium
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Sylvie Ricard-Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), UMR 5246 CNRS, ICBMS, University Lyon 1, 69622 Villeurbanne Cedex, France;
| |
Collapse
|
15
|
Tan Y, Li H, Cao G, Xin J, Yan D, Liu Y, Li P, Zhang Y, Shi L, Zhang B, Yi W, Sun Y. N-terminal domain of CTRP9 promotes cardiac fibroblast activation in myocardial infarction via Rap1/Mek/Erk pathway. J Transl Med 2025; 23:300. [PMID: 40065407 PMCID: PMC11892279 DOI: 10.1186/s12967-025-06274-z] [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: 12/07/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND In developed nations, myocardial infarction (MI) is one of the main causes of morbidity and mortality, resulting in a significant economic burden and becoming a global public health problem. C1q/tumor necrosis factor-related protein 9 (CTRP9) is a secreted protein comprising a variable domain, a collagenous region, and a C-terminal trimerizing globular C1q (gC1q) domain. In vivo, the full-length CTRP9 (fCTRP9) can be cleaved into the globular domain of CTRP9 (gCTRP9). Here, we tested the cardio-protective impacts of fCTRP9, gCTRP9, and N-terminal domain, including the variable and collagenous domain, of CTRP9 (nCTRP9) in the context of MI. METHODS Studies comparing the protective properties of fCTRP9 and gCTRP9 against MI in mice hearts were performed both in vitro and in vivo. The role of matrix metalloproteinase-9 (MMP9) in CTRP9 cleavage was examined, and the effects of different CTRP9 domains on cardiac fibrosis and cardiac fibroblast (CF) activation were investigated. RESULTS gCTRP9 exerted better protective effects than fCTRP9 against MI, demonstrating superior anti-apoptotic and anti-fibrotic properties. fCTRP9 was cleaved by MMP9, resulting in gCTRP9 and nCTRP9. MMP9 overexpression enhanced the cardioprotective effects of fCTRP9, while nCTRP9 supplementation aggravated cardiac fibrosis in MI mice. Mechanistically, nCTRP9 activated CFs via an increase in Rap1 expression and MEK 1/2 and ERK1/2 phosphorylation. CONCLUSIONS Different domains of CTRP9 have distinct cardioprotective effects. gCTRP9 shows beneficial effects, while nCTRP9 promotes cardiac fibrosis. These findings highlight the importance of CTRP9 in cardiac function regulation and suggest prospective therapeutic options for MI treatment.
Collapse
Affiliation(s)
- Yanzhen Tan
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Hong Li
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Department of General Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Guojie Cao
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jialin Xin
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dongxu Yan
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yingying Liu
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Panpan Li
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuxi Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lei Shi
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bing Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, No.127, Changlexi Road, Xi'an, Shaanxi, 710032, China.
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, No.127, Changlexi Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
16
|
Wang X, Yu S, Xie L, Xiang M, Ma H. The role of the extracellular matrix in cardiac regeneration. Heliyon 2025; 11:e41157. [PMID: 39834404 PMCID: PMC11745795 DOI: 10.1016/j.heliyon.2024.e41157] [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: 07/08/2024] [Revised: 11/16/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
The extracellular matrix (ECM) is a complex and dynamic three-dimensional network that functions as an architectural scaffold to maintain cardiac homeostasis. Important biochemical and mechanical signals associated with cell‒cell communication are provided via the reciprocal interaction between cells and the ECM. By converting mechanical cues into biochemical signals, the ECM regulates many cell processes, including migration, adhesion, growth, differentiation, proliferation, and apoptosis. Moreover, the ECM facilitates the replacement of dead cells and preserves the structural integrity of the heart, making it essential in conditions such as myocardial infarction and other pathological states. When excessive ECM deposition or abnormal production of ECM components occurs, the heart undergoes fibrosis, leading to cardiac dysfunction and heart failure. However, emerging evidence suggests that the ECM may contribute to heart regeneration following cardiac injury. The present review offers a complete overview of the existing information and novel discoveries regarding the involvement of the ECM in heart regeneration from both mechanical and biochemical perspectives. Understanding the ECM and its involvement in mechanotransduction holds significant potential for advancing therapeutic approaches in heart repair and regeneration.
Collapse
Affiliation(s)
- Xiying Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lan Xie
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China
| |
Collapse
|
17
|
Bellon B, Pippenger B, Stähli A, Degen M, Parisi L. Cementum and enamel surface mimicry influences soft tissue cell behavior. J Periodontal Res 2025; 60:64-76. [PMID: 38828886 PMCID: PMC11840463 DOI: 10.1111/jre.13295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024]
Abstract
AIMS To test whether titanium surface roughness disparity might be used to specifically guide the behavior of gingiva fibroblasts and keratinocytes, thereby improving the quality of soft tissue (ST) integration around abutments. METHODS Titanium discs resembling the roughness of enamel (M) or cementum (MA) were created with normal or increased hydrophilicity and used as substrates for human fibroblasts and keratinocytes. Adhesion and proliferation assays were performed to assess cell-type specific responses upon encountering the different surfaces. Additionally, immunofluorescence and qPCR analyses were performed to study more in depth the behavior of fibroblasts and keratinocytes on MA and M surfaces, respectively. RESULTS While enamel-like M surfaces supported adhesion, growth and a normal differentiation potential of keratinocytes, cementum-emulating MA surfaces specifically impaired the growth of keratinocytes. Vice versa, MA surfaces sustained regular adhesion and proliferation of fibroblasts. Yet, a more intimate adhesion between fibroblasts and titanium was achieved by an increased hydrophilicity of MA surfaces, which was associated with an increased expression of elastin. CONCLUSION The optimal titanium implant abutment might be achieved by a bimodal roughness design, mimicking the roughness of enamel (M) and cementum with increased hydrophilicity (hMA), respectively. These surfaces can selectively elicit cell responses favoring proper ST barrier by impairing epithelial downgrowth and promoting firm adhesion of fibroblasts.
Collapse
Affiliation(s)
- Benjamin Bellon
- Faculty of Medicine and Health TechnologyUniversity of TampereTampereFinland
- Preclinical and Translational ResearchInstitut Straumann AGBaselSwitzerland
- Clinic of Conservative and Preventive DentistryUniversity of ZurichZürichSwitzerland
| | - Benjamin Pippenger
- Preclinical and Translational ResearchInstitut Straumann AGBaselSwitzerland
- Department of PeriodontologyUniversity of BernBernSwitzerland
| | | | - Martin Degen
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial OrthopedicsUniversity of BernBernSwitzerland
| | - Ludovica Parisi
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial OrthopedicsUniversity of BernBernSwitzerland
| |
Collapse
|
18
|
Mao Q, Zhang X, Yang J, Kong Q, Cheng H, Yu W, Cao X, Li Y, Li C, Liu L, Ding Z. HSPA12A acts as a scaffolding protein to inhibit cardiac fibroblast activation and cardiac fibrosis. J Adv Res 2025; 67:217-229. [PMID: 38219869 PMCID: PMC11725103 DOI: 10.1016/j.jare.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/12/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024] Open
Abstract
INTRODUCTION Cardiac fibrosis is the main driver for adverse remodeling and progressive functional decline in nearly all types of heart disease including myocardial infarction (MI). The activation of cardiac fibroblasts (CF) into myofibroblasts is responsible for cardiac fibrosis. Unfortunately, no ideal approach for controlling CF activation currently exists. OBJECTIVES This study investigated the role of Heat shock protein A12A (HSPA12A), an atypical member of the HSP70 family, in CF activation and MI-induced cardiac fibrosis. METHODS Primary CF and Hspa12a knockout mice were used in the experiments. CF activation was indicated by the upregulation of myofibroblast characters including alpha-Smooth muscle actin (αSMA), Collagen, and Fibronectin. Cardiac fibrosis was illustrated by Masson's trichrome and picrosirius staining. Cardiac function was examined using echocardiography. Glycolytic activity was indicated by levels of extracellular lactate and the related protein expression. Protein stability was examined following cycloheximide and MG132 treatment. Protein-protein interaction was examined by immunoprecipitation-immunoblotting analysis. RESULTS HSPA12A displayed a high expression level in quiescent CF but showed a decreased expression in activated CF, while ablation of HSPA12A in mice promoted CF activation and cardiac fibrosis following MI. HSPA12A overexpression inhibited the activation of primary CF through inhibiting glycolysis, while HSPA12A knockdown showed the opposite effects. Moreover, HSPA12A upregulated the protein expression of transcription factor p53, by which mediated the HSPA12A-induced inhibition of glycolysis and CF activation. Mechanistically, this action of HSPA12A was achieved by acting as a scaffolding protein to bind p53 and ubiquitin specific protease 10 (USP10), thereby promoting the USP10-mediated p53 protein stability and the p53-medicated glycolysis inhibition. CONCLUSION The present study provided clear evidence that HSPA12A is a novel endogenous inhibitor of CF activation and cardiac fibrosis. Targeting HSPA12A in CF could represent a promising strategy for the management of cardiac fibrosis in patients.
Collapse
Affiliation(s)
- Qian Mao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinna Yang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Cheng
- Department of Anesthesiology, The First Affiliated Hospital with Wannan Medical College, Wuhu, China
| | - Wansu Yu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaofei Cao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yuehua Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Chuanfu Li
- Departments of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
19
|
Ma W, Chen H, Tian Y, Huang W, Ren Z, Li J, Ouyang Q, Hu Y, Wang X, Ji H, Liu X, Liu Y, Wang X, Liu Y, Tian Y, Li F, Yang B, Wang N, Cai B. The highly conserved PIWI-interacting RNA CRAPIR antagonizes PA2G4-mediated NF110-NF45 disassembly to promote heart regeneration in mice. NATURE CARDIOVASCULAR RESEARCH 2025; 4:102-118. [PMID: 39814981 DOI: 10.1038/s44161-024-00592-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/28/2024] [Indexed: 01/18/2025]
Abstract
Targeting the cardiomyocyte cell cycle is a promising strategy for heart repair following injury. Here, we identify a cardiac-regeneration-associated PIWI-interacting RNA (CRAPIR) as a regulator of cardiomyocyte proliferation. Genetic ablation or antagomir-mediated knockdown of CRAPIR in mice impairs cardiomyocyte proliferation and reduces heart regenerative potential. Conversely, overexpression of CRAPIR promotes cardiomyocyte proliferation, reduces infarct size and improves heart function after myocardial infarction. Mechanistically, CRAPIR promotes cardiomyocyte proliferation by competing with NF110 for binding to the RNA-binding protein PA2G4, thereby preventing the interaction of PA2G4 with the NF110-NF45 heterodimer and reducing NF110 degradation. The ability of CRAPIR to promote proliferation was confirmed in human embryonic stem cell-derived cardiomyocytes. Notably, CRAPIR serum levels are lower in individuals with ischemic heart disease and negatively correlate with levels of N-terminal pro-brain natriuretic peptide. These findings position CRAPIR both as a potential diagnostic marker for cardiac injury and as a therapeutic target for heart regeneration through the PA2G4-NF110-NF45 signaling axis.
Collapse
Affiliation(s)
- Wenya Ma
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Hongyang Chen
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China.
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China.
| | - Yanan Tian
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Wei Huang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Zhongyu Ren
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Jianglong Li
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Qimeng Ouyang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Yu Hu
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Haoyu Ji
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Xu Liu
- Department of Laboratory Medicine at the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yu Liu
- Department of Laboratory Medicine at the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - XiuXiu Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Yining Liu
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
| | - Ye Tian
- Department of Pathophysiology and the Key Laboratory of Cardiovascular Pathophysiology, Harbin Medical University, Harbin, China
| | - Faqian Li
- Department of Pathology and Laboratory Medicine at Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Baofeng Yang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Harbin, China
| | - Ning Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China.
| | - Benzhi Cai
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education; National Key Laboratory of Frigid Zone Cardiovascular Diseases), Harbin Medical University, Harbin, China.
- NHC Key Laboratory of Cell Transplantation, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Li R, Xu S, Guo Y, Cao C, Xu J, Hao L, Luo S, Chen X, Du Y, Li Y, Xie Y, Gao W, Li J, Xu B. Application of collagen in bone regeneration. J Orthop Translat 2025; 50:129-143. [PMID: 40171103 PMCID: PMC11960539 DOI: 10.1016/j.jot.2024.10.002] [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: 07/09/2024] [Revised: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 04/03/2025] Open
Abstract
At present, there is a significant population of individuals experiencing bone deficiencies caused by injuries, ailments affecting the bones, congenital abnormalities, and cancer. The management of substantial bone defects a significant global orthopedic challenge due to the intricacies involved in promoting and restoring the growth of fresh osseous tissue. Autografts are widely regarded as the "gold standard" for repairing bone defects because of their superior tissue acceptance and ability to control osteogenesis. However, patients undergoing autografts may encounter various challenges, including but not limited to hernia, bleeding, nerve impairment, tissue death. Therefore, researchers in regenerative medicine are striving to find alternatives. Collagen is the most abundant protein in the human body, and its triple helix structure gives it unique characteristics that contribute to its strength and functionality in various tissues. Collagen is commonly processed into various forms such as scaffolds, sponges, membranes, hydrogels, and composite materials, due to its unique compatibility with the human body, affinity for water, minimal potential for immune reactions, adaptability, and ability to transport nutrients or drugs. As an alternative material in the field of bone regeneration, collagen is becoming increasingly important. The objective of this review is to provide a comprehensive analysis of the primary types and sources of collagen, their processes of synthesis and degradation, as well as the advancements made in bone regeneration research and its potential applications. A comprehensive investigation into the role of collagen in bone regeneration is undertaken, providing valuable points of reference for a more profound comprehension of collagen applications in this field. The concluding section provides a comprehensive overview of the prospective avenues for collagen research, underscoring their promising future and highlighting their significant potential in the field of bone regeneration. The Translational Potential of this Article. The comprehensive exploration into the diverse functions and translational potential of collagen in bone regeneration, as demonstrated in this review, these findings underscore their promising potential as a treatment option with significant clinical implications, thus paving the way for innovative and efficacious therapeutic strategies in this domain.
Collapse
Affiliation(s)
- Rou Li
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
- China Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| | - Shiqing Xu
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Yanning Guo
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Cong Cao
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Jingchen Xu
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Lijun Hao
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Sai Luo
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Xinyao Chen
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Yuyang Du
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Heilongjiang Province, PR China
| | - Ye Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guang Zhou 510515, PR China
| | - Yong Xie
- Department of Cardiac Surgery, The First Affiliated Hospital of Tsinghua University, Beijing 100036, PR China
| | - Weitong Gao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, PR China
| | - Jing Li
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
| | - Baohua Xu
- China–Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing 100029, PR China
- China Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, PR China
| |
Collapse
|
21
|
Li S, Tao B, Wan J, Montecino-Rodriguez E, Wang P, Ma F, Sun B, Gu Y, Ramadoss S, Su L, Sun Q, Hoeve JT, Stiles L, Collins J, van Dam RM, Tamboline M, Taschereau R, Shirihai O, Kitchen DB, Pellegrini M, Graeber T, Dorshkind K, Xu S, Deb A. A humanized monoclonal antibody targeting an ectonucleotidase rescues cardiac metabolism and heart function after myocardial infarction. Cell Rep Med 2024; 5:101795. [PMID: 39454569 PMCID: PMC11604407 DOI: 10.1016/j.xcrm.2024.101795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024]
Abstract
Myocardial infarction (MI) results in aberrant cardiac metabolism, but no therapeutics have been designed to target cardiac metabolism to enhance heart repair. We engineer a humanized monoclonal antibody against the ectonucleotidase ENPP1 (hENPP1mAb) that targets metabolic crosstalk in the infarcted heart. In mice expressing human ENPP1, systemic administration of hENPP1mAb metabolically reprograms myocytes and non-myocytes and leads to a significant rescue of post-MI heart dysfunction. Using metabolomics, single-nuclear transcriptomics, and cellular respiration studies, we show that the administration of the hENPP1mAb induces organ-wide metabolic and transcriptional reprogramming of the heart that enhances myocyte cellular respiration and decreases cell death and fibrosis in the infarcted heart. Biodistribution and safety studies showed specific organ-wide distribution with the antibody being well tolerated. In humanized animals, with drug clearance kinetics similar to humans, we demonstrate that a single "shot" of the hENPP1mAb after MI is sufficient to rescue cardiac dysfunction.
Collapse
Affiliation(s)
- Shen Li
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bo Tao
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jijun Wan
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Enca Montecino-Rodriguez
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ping Wang
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Feiyang Ma
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Baiming Sun
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yiqian Gu
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sivakumar Ramadoss
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lianjiu Su
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Qihao Sun
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Johanna Ten Hoeve
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute of Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Linsey Stiles
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey Collins
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute of Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - R Michael van Dam
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute of Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mikayla Tamboline
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute of Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard Taschereau
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute of Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Orian Shirihai
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | - Matteo Pellegrini
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Thomas Graeber
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute of Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kenneth Dorshkind
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Shili Xu
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute of Molecular Imaging, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA; Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Shin K, Rodriguez-Parks A, Kim C, Silaban IM, Xia Y, Sun J, Dong C, Keles S, Wang J, Cao J, Kang J. Harnessing the regenerative potential of interleukin11 to enhance heart repair. Nat Commun 2024; 15:9666. [PMID: 39516197 PMCID: PMC11549343 DOI: 10.1038/s41467-024-54060-0] [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: 11/02/2023] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Balancing between regenerative processes and fibrosis is crucial for heart repair, yet strategies regulating this balance remain a barrier to developing therapies. The role of Interleukin 11 (IL11) in heart regeneration remains controversial, as both regenerative and fibrotic functions have been reported. We uncovered that il11a, an Il11 homolog in zebrafish, can trigger robust regenerative programs in zebrafish hearts, including cardiomyocytes proliferation and coronary expansion, even in the absence of injury. Notably, il11a induction in uninjured hearts also activates the quiescent epicardium to produce epicardial progenitor cells, which later differentiate into cardiac fibroblasts. Consequently, prolonged il11a induction indirectly leads to persistent fibroblast emergence, resulting in cardiac fibrosis. While deciphering the regenerative and fibrotic effects of il11a, we found that il11-dependent fibrosis, but not regeneration, is mediated through ERK activity, suggesting to potentially uncouple il11a dual effects on regeneration and fibrosis. To harness the il11a's regenerative ability, we devised a combinatorial treatment through il11a induction with ERK inhibition. This approach enhances cardiomyocyte proliferation with mitigated fibrosis, achieving a balance between regenerative processes and fibrosis. Thus, we unveil the mechanistic insights into regenerative il11 roles, offering therapeutic avenues to foster cardiac repair without exacerbating fibrosis.
Collapse
Affiliation(s)
- Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Anjelica Rodriguez-Parks
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Chanul Kim
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Isabella M Silaban
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Yu Xia
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Jisheng Sun
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, USA
| | - Chenyang Dong
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, USA
| | - Sunduz Keles
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, USA
| | - Jinhu Wang
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA.
- UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
23
|
Chandrasekaran P, Alanazi A, Kwok B, Li Q, Viraraghavan G, Balasubramanian S, Frank DB, Lu XL, Birk DE, Mauck RL, Dyment NA, Koyama E, Han L. Type V collagen exhibits distinct regulatory activities in TMJ articular disc versus condylar cartilage during postnatal growth and remodeling. Acta Biomater 2024; 189:192-207. [PMID: 39362448 PMCID: PMC11640222 DOI: 10.1016/j.actbio.2024.09.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 09/02/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Understanding matrix molecular activities that regulate the postnatal growth and remodeling of the temporomandibular joint (TMJ) articular disc and condylar cartilage will enable the development of effective regenerative strategies targeting TMJ disorders. This study elucidated the distinct roles of type V collagen (collagen V) in regulating these two units. Studying the TMJ of young adult Col5a1+/- mice, we found that loss of collagen V resulted in substantial changes in the proliferation, clustering and density of progenitors in condylar cartilage, but did not have a major impact on disc cells that are more fibroblast-like. Although loss of collagen V led to thickened collagen fibrils with increased heterogeneity in the disc, there were no significant changes in local micromodulus, except for a reduction at the posterior end of the inferior side. Following the induction of aberrant occlusal loading by the unilateral anterior crossbite (UAC) procedure, both wild-type (WT) and Col5a1+/- condylar cartilage exhibited salient remodeling, and Col5a1+/- condyle developed more pronounced degeneration and tissue hypertrophy at the posterior end than the WT. In contrast, neither UAC nor collagen V deficiency induced marked changes in the morphology or biomechanical properties of the disc. Together, our findings highlight the distinct roles of collagen V in regulating these two units during postnatal growth and remodeling, emphasizing its more crucial role in condylar cartilage due to its impact on the highly mechanosensitive progenitors. These results provide the foundation for using collagen V to improve the regeneration of TMJ and the care of patients with TMJ disorders. STATEMENT OF SIGNIFICANCE: Successful regeneration of the temporomandibular joint (TMJ) articular disc and condylar cartilage remains a significant challenge due to the limited understanding of matrix molecular activities that regulate the formation and remodeling of these tissues. This study demonstrates that collagen V plays distinct and critical roles in these processes. In condylar cartilage, collagen V is essential for regulating progenitor cell fate and maintaining matrix integrity. In the disc, collagen V also regulates fibril structure and local micromechanics, but has a limited impact on cell phenotype or its remodeling response. Our findings establish collagen V as a key component in maintaining the integrity of these two units, with a more crucial role in condylar cartilage due to its impact on progenitor cell activities.
Collapse
Affiliation(s)
- Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Abdulaziz Alanazi
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Girish Viraraghavan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Sriram Balasubramanian
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Division of Pediatric Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Eiki Koyama
- Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
24
|
Ramadoss S, Qin J, Tao B, Thomas NE, Cao E, Wu R, Sandoval DR, Piermatteo A, Grunddal KV, Ma F, Li S, Sun B, Zhou Y, Wan J, Pellegrini M, Holst B, Lusis AJ, Gordts PLSM, Deb A. Bone-marrow macrophage-derived GPNMB protein binds to orphan receptor GPR39 and plays a critical role in cardiac repair. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1356-1373. [PMID: 39455836 DOI: 10.1038/s44161-024-00555-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane protein initially identified in nonmetastatic melanomas and has been associated with human heart failure; however, its role in cardiac injury and function remains unclear. Here we show that GPNMB expression is elevated in failing human and mouse hearts after myocardial infarction (MI). Lineage tracing and bone-marrow transplantation reveal that bone-marrow-derived macrophages are the main source of GPNMB in injured hearts. Using genetic loss-of-function models, we demonstrate that GPNMB deficiency leads to increased mortality, cardiac rupture and rapid post-MI left ventricular dysfunction. Conversely, increasing circulating GPNMB levels through viral delivery improves heart function after MI. Single-cell transcriptomics show that GPNMB enhances myocyte contraction and reduces fibroblast activation. Additionally, we identified GPR39 as a receptor for circulating GPNMB, with its absence negating the beneficial effects. These findings highlight a pivotal role of macrophage-derived GPNMBs in post-MI cardiac repair through GPR39 signaling.
Collapse
MESH Headings
- Animals
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Humans
- Macrophages/metabolism
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/genetics
- Mice, Knockout
- Disease Models, Animal
- Myocytes, Cardiac/metabolism
- Male
- Mice, Inbred C57BL
- Signal Transduction
- Ventricular Function, Left
- Heart Failure/metabolism
- Heart Failure/genetics
- Female
- Mice
- Cells, Cultured
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/genetics
- Bone Marrow Transplantation
- Protein Binding
- Regeneration
- Eye Proteins
Collapse
Affiliation(s)
- Sivakumar Ramadoss
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Juan Qin
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Bo Tao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Nathan E Thomas
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
- Glycobiology Research and Training Center, University of California-San Diego, La Jolla, CA, USA
| | - Edward Cao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Rimao Wu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Daniel R Sandoval
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
| | - Ann Piermatteo
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
| | - Kaare V Grunddal
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
| | - Feiyang Ma
- Department of Cell and Development Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Baiming Sun
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Yonggang Zhou
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Jijun Wan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA
| | - Birgitte Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Philip L S M Gordts
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, CA, USA
- Glycobiology Research and Training Center, University of California-San Diego, La Jolla, CA, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California-Los Angeles, Los Angeles, CA, USA.
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California-Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA, USA.
- California Nanosystems Institute, University of California-Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Xie Z, Xie H, Xie C, Yang S, Feng Y, Su Z, Tang T, Zhang B, Yang J, Wang Y, Huang L, Zhu H, Cao J, Jiang R, Li T, Lu W. A combined analysis of bulk RNA-seq and scRNA-seq was performed to investigate the molecular mechanisms associated with the occurrence of myocardial infarction. BMC Genomics 2024; 25:921. [PMID: 39363266 PMCID: PMC11448016 DOI: 10.1186/s12864-024-10813-1] [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: 04/10/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Myocardial infarction (MI) induces complex transcriptional changes across diverse cardiac cell types. Single-cell RNA sequencing (scRNA-seq) provides an unparalleled ability to discern cellular diversity during infarction, yet the veracity of these discoveries necessitates confirmation. This investigation sought to elucidate MI mechanisms by integrating scRNA-seq and bulk RNA-seq data. METHODS Publicly available scRNA-seq (GSE136088) and bulk RNA-seq (GSE153485) data from mice MI models were analyzed. Cell types were annotated, and differential expression analysis conducted. Bulk RNA-seq underwent quality control, principal component analysis, and differential expression analysis. RESULTS In scRNA-seq data, the comparison between MI and sham groups unveiled a reduction in endothelial cell populations, but macrophages and monocytes increased. Within fibroblast subgroups, three distinct categories were discerned, with two exhibiting upregulation in MI. Notably, endothelial cells exhibited an elevated expression of genes associated with apoptosis and ferroptosis. In bulk RNA-seq analysis, distinct patterns emerged when comparing MI and sham groups. Specifically, six genes linked to endothelial ferroptosis exhibited heightened expression in MI group, thereby corroborating the scRNA-seq findings. Moreover, the examination of isolated cardiac macrophages from mice MI model revealed increased expression of Spp1, Col1a2, Col3a1, Ctsd, and Lgals3 compared to sham group, thus substantiating the dysregulation of macrophage apoptosis-related proteins following MI. CONCLUSION MI altered the transcriptomic landscapes of cardiac cells with increased expression of apoptotic genes. Moreover, the upregulation of macrophage apoptosis marker was confirmed within MI models. The presence of endothelial cell depletion and ferroptosis in MI has been demonstrated.
Collapse
Affiliation(s)
- Zheng Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Huicong Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Chen Xie
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Saichao Yang
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Yun Feng
- Department of General Practice, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Zhaohai Su
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Tao Tang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Bilong Zhang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Jiangyong Yang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Yueting Wang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Ling Huang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Hengqing Zhu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Jun Cao
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Rengui Jiang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Weiling Lu
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital (Gannan Medical University Affiliated Municipal Hospital), 49 Dagong Road, Ganzhou, 341000, China.
| |
Collapse
|
26
|
Zhang D, Wen Q, Zhang R, Kou K, Lin M, Zhang S, Yang J, Shi H, Yang Y, Tan X, Yin S, Ou X. From Cell to Gene: Deciphering the Mechanism of Heart Failure With Single-Cell Sequencing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308900. [PMID: 39159065 PMCID: PMC11497092 DOI: 10.1002/advs.202308900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Heart failure (HF) is a prevalent cardiovascular disease with significant morbidity and mortality rates worldwide. Due to the intricate structure of the heart, diverse cell types, and the complex pathogenesis of HF, further in-depth investigation into the underlying mechanisms is required. The elucidation of the heterogeneity of cardiomyocytes and the intercellular communication network is particularly important. Traditional high-throughput sequencing methods provide an average measure of gene expression, failing to capture the "heterogeneity" between cells and impacting the accuracy of gene function knowledge. In contrast, single-cell sequencing techniques allow for the amplification of the entire genome or transcriptome at the individual cell level, facilitating the examination of gene structure and expression with unparalleled precision. This approach offers valuable insights into disease mechanisms, enabling the identification of changes in cellular components and gene expressions during hypertrophy associated with HF. Moreover, it reveals distinct cell populations and their unique roles in the HF microenvironment, providing a comprehensive understanding of the cellular landscape that underpins HF pathogenesis. This review focuses on the insights provided by single-cell sequencing techniques into the mechanisms underlying HF and discusses the challenges encountered in current cardiovascular research.
Collapse
Affiliation(s)
- Dan Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
- Department of Rehabilitation MedicineSouthwest Medical UniversityLuzhouSichuan646000China
| | - Qiang Wen
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang RdWuhanHubei430022China
| | - Rui Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Kun Kou
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Miao Lin
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Shiyu Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Jun Yang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Hangchuan Shi
- Department of Clinical & Translational ResearchUniversity of Rochester Medical Center265 Crittenden BlvdRochesterNY14642USA
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical Center601 Elmwood AveRochesterNY14642USA
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
- Department of PhysiologySchool of Basic Medical SciencesSouthwest Medical UniversityLuzhouSichuan646000China
| | - Shigang Yin
- Luzhou Key Laboratory of Nervous system disease and Brain FunctionSouthwest Medical UniversityLuzhouSichuan646000China
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal ResourcesGuangxi Normal UniversityGuilinGuangxi541004China
| |
Collapse
|
27
|
Kelly-Scumpia KM, Archang MM, Purbey PK, Yokota T, Wu R, McCourt J, Li S, Crosbie RH, Scumpia PO, Deb A. Modulating the extracellular matrix to treat wound healing defects in Ehlers-Danlos syndrome. iScience 2024; 27:110676. [PMID: 39262784 PMCID: PMC11389543 DOI: 10.1016/j.isci.2024.110676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/13/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024] Open
Abstract
Classic Ehlers-Danlos syndrome (cEDS) is a genetic disorder of the connective tissue that is characterized by mutations in genes coding type V collagen. Wound healing defects are characteristic of cEDS and no therapeutic strategies exist. Herein we describe a murine model of cEDS that phenocopies wound healing defects seen in humans. Our model features mice with conditional loss of Col5a1 in Col1a2 + fibroblasts (Col5a1CKO). This model shows that an abnormal extracellular matrix (ECM) characterized by fibrillar disarray, altered mechanical properties, and decreased collagen deposition contribute to the wound healing defect. The cEDS animals exhibit decreased expression of epidermal genes and increased inflammation. Finally, we demonstrate that inhibiting mechanosensitive integrin signaling or by injecting wild-type (WT) fibroblasts into cEDS animals enhances epidermal gene expression, decreases inflammation, and augments wound closure. These findings suggest that cell delivery and/or blocking integrin signaling are potentially therapeutic strategies to rescue wound healing defects in cEDS.
Collapse
Affiliation(s)
- Kindra M Kelly-Scumpia
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Maani M Archang
- Bioengineering Department, University of California, Los Angeles, Los Angeles, CA, USA
- Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System-West Los Angeles, Los Angeles, CA 90073, USA
| | - Prabhat K Purbey
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tomohiro Yokota
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rimao Wu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jackie McCourt
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Philip O Scumpia
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System-West Los Angeles, Los Angeles, CA 90073, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
28
|
Ayhan S, Dursun A. ELFN1 is a new extracellular matrix (ECM)-associated protein. Life Sci 2024; 352:122900. [PMID: 38986898 DOI: 10.1016/j.lfs.2024.122900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
AIMS The ELFN1, discovered in 2007, is a single-pass transmembrane protein. Studies conducted thus far to elucidate the function of the Elfn1 have been limited only to animal studies. These studies have reported that ELFN1 is a universal binding partner of metabotropic glutamate receptors (mGluRs) in the central nervous system and its functional deficiency has been associated with the pathogenesis of neurological and neuropsychiatric diseases. In 2021, we described the first disease-associated human ELFN1 pathogenic gene mutation. Severe joint laxity, which was the most striking finding of this new disease and was clearly seen in the patients since early infancy, showed that the ELFN1 may have a possible function in the connective tissue besides the nervous system. Here, we present the first experimental evidence of the extracellular matrix (ECM)-related function of the ELFN1. MATERIALS AND METHODS Primary skin fibroblasts were isolated from the skin biopsies of ELFN1 mutated patients and healthy foreskin donors. For the clinical trial in a dish, in vitro ECM and DEM (decellularized ECM) models were created from skin fibroblasts. All the in vitro models were comparatively characterized and analyzed. KEY FINDINGS The mutation in the ELFN1 signal peptide region of patients resulted in a severe lack of ELFN1 expression and dramatically altered the characteristic morphology and behavior (growth, proliferation, and motility) of fibroblasts. SIGNIFICANCE We propose that ELFN1 is involved in the cell-ECM attachment, and its deficiency is critical enough to cause a loss of cell motility and soft ECM stiffness.
Collapse
Affiliation(s)
- Selda Ayhan
- Department of Pediatrics Metabolism, Institute of Child Health, Hacettepe University, Sıhhıye, Ankara 06100, Turkey.
| | - Ali Dursun
- Department of Pediatrics Metabolism, Faculty of Medicine, Hacettepe University, Sıhhıye, Ankara 06100, Turkey.
| |
Collapse
|
29
|
Correia JC, Jannig PR, Gosztyla ML, Cervenka I, Ducommun S, Præstholm SM, Dias JM, Dumont KD, Liu Z, Liang Q, Edsgärd D, Emanuelsson O, Gregorevic P, Westerblad H, Venckunas T, Brazaitis M, Kamandulis S, Lanner JT, Teixeira AI, Yeo GW, Ruas JL. Zfp697 is an RNA-binding protein that regulates skeletal muscle inflammation and remodeling. Proc Natl Acad Sci U S A 2024; 121:e2319724121. [PMID: 39141348 PMCID: PMC11348326 DOI: 10.1073/pnas.2319724121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 06/27/2024] [Indexed: 08/15/2024] Open
Abstract
Skeletal muscle atrophy is a morbidity and mortality risk factor that happens with disuse, chronic disease, and aging. The tissue remodeling that happens during recovery from atrophy or injury involves changes in different cell types such as muscle fibers, and satellite and immune cells. Here, we show that the previously uncharacterized gene and protein Zfp697 is a damage-induced regulator of muscle remodeling. Zfp697/ZNF697 expression is transiently elevated during recovery from muscle atrophy or injury in mice and humans. Sustained Zfp697 expression in mouse muscle leads to a gene expression signature of chemokine secretion, immune cell recruitment, and extracellular matrix remodeling. Notably, although Zfp697 is expressed in several cell types in skeletal muscle, myofiber-specific Zfp697 genetic ablation in mice is sufficient to hinder the inflammatory and regenerative response to muscle injury, compromising functional recovery. We show that Zfp697 is an essential mediator of the interferon gamma response in muscle cells and that it functions primarily as an RNA-interacting protein, with a very high number of miRNA targets. This work identifies Zfp697 as an integrator of cell-cell communication necessary for tissue remodeling and regeneration.
Collapse
Affiliation(s)
- Jorge C. Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Paulo R. Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Maya L. Gosztyla
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA92093
- Sanford Stem Cell Institute Innovation Center and Stem Cell Program, University of California San Diego, La Jolla, CA92093
- Center for RNA Technologies and Therapeutics, University of California San Diego, La Jolla, CA92093
| | - Igor Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Serge Ducommun
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Stine M. Præstholm
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - José M. Dias
- Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska, StockholmSE-171 77, Sweden
- Department of Cell and Molecular Biology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Kyle D. Dumont
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Zhengye Liu
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Qishan Liang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA92093
- Center for RNA Technologies and Therapeutics, University of California San Diego, La Jolla, CA92093
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA92093
| | - Daniel Edsgärd
- Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Biotechnology, Chemistry and Health, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Olof Emanuelsson
- Science for Life Laboratory, Department of Gene Technology, School of Engineering Sciences in Biotechnology, Chemistry and Health, KTH Royal Institute of Technology, Stockholm SE-100 44, Sweden
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Håkan Westerblad
- Muscle Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Tomas Venckunas
- Institute of Sports Science and Innovations, Lithuanian Sports University, Kaunas44221, Lithuania
| | - Marius Brazaitis
- Institute of Sports Science and Innovations, Lithuanian Sports University, Kaunas44221, Lithuania
| | - Sigitas Kamandulis
- Institute of Sports Science and Innovations, Lithuanian Sports University, Kaunas44221, Lithuania
| | - Johanna T. Lanner
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
| | - Ana I. Teixeira
- Nanomedicine and Spatial Biology, Department of Physiology and Pharmacology, Biomedicum, Karolinska, StockholmSE-171 77, Sweden
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA92093
- Sanford Stem Cell Institute Innovation Center and Stem Cell Program, University of California San Diego, La Jolla, CA92093
- Center for RNA Technologies and Therapeutics, University of California San Diego, La Jolla, CA92093
| | - Jorge L. Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, StockholmSE-171 77, Sweden
- Department of Pharmacology and Stanley & Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI48109
| |
Collapse
|
30
|
Gao C, Xiong Z, Liu Y, Wang M, Wang M, Liu T, Liu J, Ren S, Cao N, Yan H, Drucker DJ, Rau CD, Yokota T, Huang J, Wang Y. Glucagon Receptor Antagonist for Heart Failure With Preserved Ejection Fraction. Circ Res 2024; 135:614-628. [PMID: 39011638 PMCID: PMC11325917 DOI: 10.1161/circresaha.124.324706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is an emerging major unmet need and one of the most significant clinic challenges in cardiology. The pathogenesis of HFpEF is associated with multiple risk factors. Hypertension and metabolic disorders associated with obesity are the 2 most prominent comorbidities observed in patients with HFpEF. Although hypertension-induced mechanical overload has long been recognized as a potent contributor to heart failure with reduced ejection fraction, the synergistic interaction between mechanical overload and metabolic disorders in the pathogenesis of HFpEF remains poorly characterized. METHOD We investigated the functional outcome and the underlying mechanisms from concurrent mechanic and metabolic stresses in the heart by applying transverse aortic constriction in lean C57Bl/6J or obese/diabetic B6.Cg-Lepob/J (ob/ob) mice, followed by single-nuclei RNA-seq and targeted manipulation of a top-ranked signaling pathway differentially affected in the 2 experimental cohorts. RESULTS In contrast to the post-transverse aortic constriction C57Bl/6J lean mice, which developed pathological features of heart failure with reduced ejection fraction over time, the post-transverse aortic constriction ob/ob mice showed no significant changes in ejection fraction but developed characteristic pathological features of HFpEF, including diastolic dysfunction, worsened cardiac hypertrophy, and pathological remodeling, along with further deterioration of exercise intolerance. Single-nuclei RNA-seq analysis revealed significant transcriptome reprogramming in the cardiomyocytes stressed by both pressure overload and obesity/diabetes, markedly distinct from the cardiomyocytes singularly stressed by pressure overload or obesity/diabetes. Furthermore, glucagon signaling was identified as the top-ranked signaling pathway affected in the cardiomyocytes associated with HFpEF. Treatment with a glucagon receptor antagonist significantly ameliorated the progression of HFpEF-related pathological features in 2 independent preclinical models. Importantly, cardiomyocyte-specific genetic deletion of the glucagon receptor also significantly improved cardiac function in response to pressure overload and metabolic stress. CONCLUSIONS These findings identify glucagon receptor signaling in cardiomyocytes as a critical determinant of HFpEF progression and provide proof-of-concept support for glucagon receptor antagonism as a potential therapy for the disease.
Collapse
MESH Headings
- Animals
- Heart Failure/physiopathology
- Heart Failure/metabolism
- Heart Failure/drug therapy
- Heart Failure/etiology
- Stroke Volume/drug effects
- Mice, Inbred C57BL
- Mice
- Male
- Receptors, Glucagon/antagonists & inhibitors
- Receptors, Glucagon/metabolism
- Receptors, Glucagon/genetics
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Mice, Obese
- Ventricular Function, Left/drug effects
- Obesity/metabolism
- Obesity/physiopathology
- Obesity/complications
- Disease Models, Animal
- Signal Transduction
Collapse
Affiliation(s)
- Chen Gao
- Department of Pharmacology and Systems Physiology, University of Cincinnati, OH (C.G., T.L.)
| | - Zhaojun Xiong
- Department of Cardiovascular Medicine, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China (Z.X.)
| | - Yunxia Liu
- Signature Research Program in Cardiovascular and Metabolic Diseases, DukeNUS School of Medicine and National Heart Center of Singapore, Singapore (Y.L., Meng Wang, S.R., Y.W.)
| | - Meng Wang
- Signature Research Program in Cardiovascular and Metabolic Diseases, DukeNUS School of Medicine and National Heart Center of Singapore, Singapore (Y.L., Meng Wang, S.R., Y.W.)
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, China (Menglong Wang, J.L.)
| | - Tian Liu
- Department of Pharmacology and Systems Physiology, University of Cincinnati, OH (C.G., T.L.)
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, China (Menglong Wang, J.L.)
| | - Shuxun Ren
- Signature Research Program in Cardiovascular and Metabolic Diseases, DukeNUS School of Medicine and National Heart Center of Singapore, Singapore (Y.L., Meng Wang, S.R., Y.W.)
| | - Nancy Cao
- School of Medicine and Public Health, University of Wisconsin, Madison (N.C.)
| | - Hai Yan
- REMD Biotherapeutics, Camarillo, CA (Y.H.)
| | - Daniel J. Drucker
- Department of Medicine, Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada (D.J.D.)
| | - Christoph Daniel Rau
- Computational Medicine Program and Department of Human Genetics, University of North Carolina at Chapel Hill (C.D.R.)
| | - Tomohiro Yokota
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, and the VA Greater Los Angeles Healthcare System (T.Y.)
| | - Jijun Huang
- Division of Endocrinology, Department of medicine, David Geffen School of Medicine, University of California, Los Angeles (J.H.)
| | - Yibin Wang
- Signature Research Program in Cardiovascular and Metabolic Diseases, DukeNUS School of Medicine and National Heart Center of Singapore, Singapore (Y.L., Meng Wang, S.R., Y.W.)
- Department of Medicine, Duke University School of Medicine, Durham, NC (Y.W.)
| |
Collapse
|
31
|
Marvin Tan XH, Wang Y, Zhu X, Mendes FN, Chung PS, Chow YT, Man T, Lan H, Lin YJ, Zhang X, Zhang X, Nguyen T, Ardehali R, Teitell MA, Deb A, Chiou PY. Massive field-of-view sub-cellular traction force videography enabled by Single-Pixel Optical Tracers (SPOT). Biosens Bioelectron 2024; 258:116318. [PMID: 38701538 DOI: 10.1016/j.bios.2024.116318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024]
Abstract
We report a massive field-of-view and high-speed videography platform for measuring the sub-cellular traction forces of more than 10,000 biological cells over 13 mm2 at 83 frames per second. Our Single-Pixel Optical Tracers (SPOT) tool uses 2-dimensional diffraction gratings embedded into a soft substrate to convert cells' mechanical traction force into optical colors detectable by a video camera. The platform measures the sub-cellular traction forces of diverse cell types, including tightly connected tissue sheets and near isolated cells. We used this platform to explore the mechanical wave propagation in a tightly connected sheet of Neonatal Rat Ventricular Myocytes (NRVMs) and discovered that the activation time of some tissue regions are heterogeneous from the overall spiral wave behavior of the cardiac wave.
Collapse
Affiliation(s)
- Xing Haw Marvin Tan
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States; Department of Bioengineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States; Department of Electronics and Photonics, Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), 1 Fusionopolis Way, 138632, Singapore
| | - Yijie Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E Young Dr S, Los Angeles, CA, 90095, United States
| | - Xiongfeng Zhu
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Felipe Nanni Mendes
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Pei-Shan Chung
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States; Department of Bioengineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Yu Ting Chow
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Tianxing Man
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Hsin Lan
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Yen-Ju Lin
- Department of Electrical and Computer Engineering, University of California at Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Xiang Zhang
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Xiaohe Zhang
- Department of Mathematics, University of California Los Angeles, 520 Portola Plaza, Los Angeles, CA, 90095, United States
| | - Thang Nguyen
- Department of Bioengineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E Young Dr S, Los Angeles, CA, 90095, United States
| | - Michael A Teitell
- Department of Bioengineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E Young Dr S, Los Angeles, CA, 90095, United States
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E Young Dr S, Los Angeles, CA, 90095, United States
| | - Pei-Yu Chiou
- Department of Mechanical and Aerospace Engineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States; Department of Bioengineering, University of California Los Angeles, Westwood Plaza, Los Angeles, CA, 90095, United States.
| |
Collapse
|
32
|
Zhu G, Wang Y, Wang Y, Huang H, Li B, Chen P, Chen C, Zhang H, Li Y, Liu H, Chen J. Myofibroblasts derived type V collagen promoting tissue mechanical stress and facilitating metastasis and therapy resistance of lung adenocarcinoma cells. Cell Death Dis 2024; 15:493. [PMID: 38987529 PMCID: PMC11237033 DOI: 10.1038/s41419-024-06873-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/22/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
Lung cancer is a leading cause of cancer-related mortality globally, with a dismal 5-year survival rate, particularly for Lung Adenocarcinoma (LUAD). Mechanical changes within the tumor microenvironment, such as extracellular matrix (ECM) remodeling and fibroblast activity, play pivotal roles in cancer progression and metastasis. However, the specific impact of the basement membrane (BM) on the mechanical characteristics of LUAD remains unclear. This study aims to identify BM genes influencing internal mechanical stress in tumors, elucidating their effects on LUAD metastasis and therapy resistance, and exploring strategies to counteract these effects. Using Matrigel overlay and Transwell assays, we found that mechanical stress, mimicked by matrix application, augmented LUAD cell migration and invasion, correlating with ECM alterations and activation of the epithelial-mesenchymal transition (EMT) pathway. Employing machine learning, we developed the SVM_Score model based on relevant BM genes, which accurately predicted LUAD patient prognosis and EMT propensity across multiple datasets. Lower SVM_Scores were associated with worse survival outcomes, elevated cancer-related pathways, increased Tumor Mutation Burden, and higher internal mechanical stress in LUAD tissues. Notably, the SVM_Score was closely linked to COL5A1 expression in myofibroblasts, a key marker of mechanical stress. High COL5A1 expression from myofibroblasts promoted tumor invasiveness and EMT pathway activation in LUAD cells. Additionally, treatment with Sorafenib, which targets COL5A1 secretion, attenuated the tumor-promoting effects of myofibroblast-derived COL5A1, inhibiting LUAD cell proliferation, migration, and enhancing chemosensitivity. In conclusion, this study elucidates the complex interplay between mechanical stress, ECM alterations, and LUAD progression. The SVM_Score emerges as a robust prognostic tool reflecting tumor mechanical characteristics, while Sorafenib intervention targeting COL5A1 secretion presents a promising therapeutic strategy to mitigate LUAD aggressiveness. These findings deepen our understanding of the biomechanical aspects of LUAD and offer insights for future research and clinical applications.
Collapse
Affiliation(s)
- Guangsheng Zhu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yanan Wang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yingjie Wang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Hua Huang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Boshi Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Peijie Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Chen Chen
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.
| |
Collapse
|
33
|
Sun Q, Chen W, Wu R, Tao B, Wang P, Sun B, Alvarez JF, Ma F, Galindo DC, Maroney SP, Saviola AJ, Hansen KC, Li S, Deb A. Serine protease inhibitor, SerpinA3n, regulates cardiac remodelling after myocardial infarction. Cardiovasc Res 2024; 120:943-953. [PMID: 38666458 PMCID: PMC12098007 DOI: 10.1093/cvr/cvae075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/07/2024] [Accepted: 02/02/2024] [Indexed: 07/03/2024] Open
Abstract
AIMS Following myocardial infarction (MI), the heart repairs itself via a fibrotic repair response. The degree of fibrosis is determined by the balance between deposition of extracellular matrix (ECM) by activated fibroblasts and breakdown of nascent scar tissue by proteases that are secreted predominantly by inflammatory cells. Excessive proteolytic activity and matrix turnover has been observed in human heart failure, and protease inhibitors in the injured heart regulate matrix breakdown. Serine protease inhibitors (Serpins) represent the largest and the most functionally diverse family of evolutionary conserved protease inhibitors, and levels of the specific Serpin, SerpinA3, have been strongly associated with clinical outcomes in human MI as well as non-ischaemic cardiomyopathies. Yet, the role of Serpins in regulating cardiac remodelling is poorly understood. The aim of this study was to understand the role of Serpins in regulating scar formation after MI. METHODS AND RESULTS Using a SerpinA3n conditional knockout mice model, we observed the robust expression of Serpins in the infarcted murine heart and demonstrate that genetic deletion of SerpinA3n (mouse homologue of SerpinA3) leads to increased activity of substrate proteases, poorly compacted matrix, and significantly worse post-infarct cardiac function. Single-cell transcriptomics complemented with histology in SerpinA3n-deficient animals demonstrated increased inflammation, adverse myocyte hypertrophy, and expression of pro-hypertrophic genes. Proteomic analysis of scar tissue demonstrated decreased cross-linking of ECM peptides consistent with increased proteolysis in SerpinA3n-deficient animals. CONCLUSION Our study demonstrates a hitherto unappreciated causal role of Serpins in regulating matrix function and post-infarct cardiac remodelling.
Collapse
Affiliation(s)
- Qihao Sun
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Wei Chen
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Rimao Wu
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Bo Tao
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Ping Wang
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Baiming Sun
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Juan F Alvarez
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Feiyang Ma
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David Ceja Galindo
- Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sean P Maroney
- Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Anthony J Saviola
- Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kirk C Hansen
- Biochemistry and Molecular Genetics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Cardiovascular Theme, David Geffen School of Medicine, University of California, 675 Charles E Young Drive South, Los Angeles, California, 90095 CA, USA
- Department of Molecular, Cell & Developmental Biology, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, 615 Charles E Young Drive S, Los Angeles, California, 90095 CA, USA
- Molecular Biology Institute, University of California, 610 Charles E Young Dr S, Los Angeles, California, 90095 CA, USA
- California NanoSystems Institute, University of California, 570 Westwood Plaza, Los Angeles, California, 90095 CA, USA
| |
Collapse
|
34
|
Reese-Petersen AL, Holm Nielsen S, Bülow Sand JM, Schattenberg JM, Bugianesi E, Karsdal MA. The sclerotic component of metabolic syndrome: Fibroblast activities may be the central common denominator driving organ function loss and death. Diabetes Obes Metab 2024; 26:2554-2566. [PMID: 38699780 DOI: 10.1111/dom.15615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis is a common feature of more than 50 different diseases and the cause of more than 35% of deaths worldwide, of which liver, kidney, skin, heart and, recently, lungs are receiving the most attention. Tissue changes, resulting in loss of organ function, are both a cause and consequence of disease and outcome. Fibrosis is caused by an excess deposition of extracellular matrix proteins, which over time results in impaired organ function and organ failure, and the pathways leading to increased fibroblast activation are many. This narrative review investigated the common denominator of fibrosis, fibroblasts, and the activation of fibroblasts, in response to excess energy consumption in liver, kidney, heart, skin and lung fibrosis. Fibroblasts are the main drivers of organ function loss in lung, liver, skin, heart and kidney disease. Fibroblast activation in response to excess energy consumption results in the overproduction of a range of collagens, of which types I, III and VI seem to be the essential drivers of disease progression. Fibroblast activation may be quantified in serum, enabling profiling and selection of patients. Activation of fibroblasts results in the overproduction of collagens, which deteriorates organ function. Patient profiling of fibroblast activities in serum, quantified as collagen production, may identify an organ death trajectory, better enabling identification of the right treatment for use in different metabolic interventions. As metabolically activated patients have highly elevated risk of kidney, liver and heart failure, it is essential to identify which organ to treat first and monitor organ status to correct treatment regimes. In direct alignment with this, it is essential to identify the right patients with the right organ deterioration trajectory for enrolment in clinical studies.
Collapse
Affiliation(s)
| | | | | | - Jörn M Schattenberg
- Saarland University Medical Center, Homburg, Germany
- University of the Saarland, Saarbrücken, Germany
| | | | | |
Collapse
|
35
|
Wang C, Fan M, Heo SJ, Adams SM, Li T, Liu Y, Li Q, Loebel C, Alisafaei F, Burdick JA, Lu XL, Birk DE, Mauck RL, Han L. Structure-Mechanics Principles and Mechanobiology of Fibrocartilage Pericellular Matrix: A Pivotal Role of Type V Collagen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600498. [PMID: 38979323 PMCID: PMC11230444 DOI: 10.1101/2024.06.26.600498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The pericellular matrix (PCM) is the immediate microniche surrounding resident cells in various tissue types, regulating matrix turnover, cell-matrix cross-talk and disease initiation. This study elucidated the structure-mechanical properties and mechanobiological functions of the PCM in fibrocartilage, a family of connective tissues that sustain complex tensile and compressive loads in vivo. Studying the murine meniscus as the model tissue, we showed that fibrocartilage PCM contains thinner, random collagen fibrillar networks that entrap proteoglycans, a structure distinct from the densely packed, highly aligned collagen fibers in the bulk extracellular matrix (ECM). In comparison to the ECM, the PCM has a lower modulus and greater isotropy, but similar relative viscoelastic properties. In Col5a1 +/- menisci, the reduction of collagen V, a minor collagen localized in the PCM, resulted in aberrant fibril thickening with increased heterogeneity. Consequently, the PCM exhibited a reduced modulus, loss of isotropy and faster viscoelastic relaxation. This disrupted PCM contributes to perturbed mechanotransduction of resident meniscal cells, as illustrated by reduced intracellular calcium signaling, as well as upregulated biosynthesis of lysyl oxidase and tenascin C. When cultured in vitro, Col5a1 +/- meniscal cells synthesized a weakened nascent PCM, which had inferior properties towards protecting resident cells against applied tensile stretch. These findings underscore the PCM as a distinctive microstructure that governs fibrocartilage mechanobiology, and highlight the pivotal role of collagen V in PCM function. Targeting the PCM or its molecular constituents holds promise for enhancing not only meniscus regeneration and osteoarthritis intervention, but also addressing diseases across various fibrocartilaginous tissues.
Collapse
Affiliation(s)
- Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Mingyue Fan
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sheila M. Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Thomas Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Yuchen Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Claudia Loebel
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Farid Alisafaei
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ 07102, United States
| | - Jason A. Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David E. Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| |
Collapse
|
36
|
Ortega M, Fábrega-García MM, Molina-García T, Gavara J, de Dios E, Pérez-Solé N, Marcos-Garcés V, Padilla-Esquivel JJ, Diaz A, Martinez-Dolz L, Jimenez-Navarro M, Rios-Navarro C, Bodí V, Ruiz-Saurí A. Novel Fibrillar and Non-Fibrillar Collagens Involved in Fibrotic Scar Formation after Myocardial Infarction. Int J Mol Sci 2024; 25:6625. [PMID: 38928330 PMCID: PMC11204374 DOI: 10.3390/ijms25126625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Following myocardial infarction (MI), adverse remodeling depends on the proper formation of fibrotic scars, composed of type I and III collagen. Our objective was to pinpoint the participation of previously unreported collagens in post-infarction cardiac fibrosis. Gene (qRT-PCR) and protein (immunohistochemistry followed by morphometric analysis) expression of fibrillar (types II and XI) and non-fibrillar (types VIII and XII) collagens were determined in RNA-sequencing data from 92 mice undergoing myocardial ischemia; mice submitted to permanent (non-reperfused MI, n = 8) or transient (reperfused MI, n = 8) coronary occlusion; and eight autopsies from chronic MI patients. In the RNA-sequencing analysis of mice undergoing myocardial ischemia, increased transcriptomic expression of collagen types II, VIII, XI, and XII was reported within the first week, a tendency that persisted 21 days afterwards. In reperfused and non-reperfused experimental MI models, their gene expression was heightened 21 days post-MI induction and positively correlated with infarct size. In chronic MI patients, immunohistochemistry analysis demonstrated their presence in fibrotic scars. Functional analysis indicated that these subunits probably confer tensile strength and ensure the cohesion of interstitial components. Our data reveal that novel collagens are present in the infarcted myocardium. These data could lay the groundwork for unraveling post-MI fibrotic scar composition, which could ultimately influence patient survivorship.
Collapse
Affiliation(s)
- María Ortega
- INCLIVA Biomedical Research Institute, 46100 Valencia, Spain; (M.O.); (T.M.-G.); (N.P.-S.); (V.M.-G.); (A.R.-S.)
| | | | - Tamara Molina-García
- INCLIVA Biomedical Research Institute, 46100 Valencia, Spain; (M.O.); (T.M.-G.); (N.P.-S.); (V.M.-G.); (A.R.-S.)
| | - Jose Gavara
- Centro de Biomateriales e Ingeniería Tisular, Universidad Politécnica de Valencia, 46022 Valencia, Spain;
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
| | - Elena de Dios
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
| | - Nerea Pérez-Solé
- INCLIVA Biomedical Research Institute, 46100 Valencia, Spain; (M.O.); (T.M.-G.); (N.P.-S.); (V.M.-G.); (A.R.-S.)
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
| | - Víctor Marcos-Garcés
- INCLIVA Biomedical Research Institute, 46100 Valencia, Spain; (M.O.); (T.M.-G.); (N.P.-S.); (V.M.-G.); (A.R.-S.)
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain
| | | | - Ana Diaz
- Unidad Central de Investigación Médica, University of Valencia, 46010 Valencia, Spain;
| | - Luis Martinez-Dolz
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
- Cardiology Departament, Hospital Universitario Politécnico La Fe, 46026 Valencia, Spain
- Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Manuel Jimenez-Navarro
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
- Servicio de Cardiología y Cirugía Cardiovascular-Área del Corazón, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), 29590 Málaga, Spain
- Departamento de Medicina y Dermatología, Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Cesar Rios-Navarro
- INCLIVA Biomedical Research Institute, 46100 Valencia, Spain; (M.O.); (T.M.-G.); (N.P.-S.); (V.M.-G.); (A.R.-S.)
- Department of Pathology, University of Valencia, 46010 Valencia, Spain;
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
| | - Vicente Bodí
- INCLIVA Biomedical Research Institute, 46100 Valencia, Spain; (M.O.); (T.M.-G.); (N.P.-S.); (V.M.-G.); (A.R.-S.)
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
- Cardiology Department, Hospital Clínico Universitario, 46010 Valencia, Spain
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Amparo Ruiz-Saurí
- INCLIVA Biomedical Research Institute, 46100 Valencia, Spain; (M.O.); (T.M.-G.); (N.P.-S.); (V.M.-G.); (A.R.-S.)
- Department of Pathology, University of Valencia, 46010 Valencia, Spain;
- Centro de Investigación Biomédica en Red (CIBER)-CV, 28029 Madrid, Spain; (E.d.D.); (L.M.-D.); (M.J.-N.)
| |
Collapse
|
37
|
Hilgendorf I, Frantz S, Frangogiannis NG. Repair of the Infarcted Heart: Cellular Effectors, Molecular Mechanisms and Therapeutic Opportunities. Circ Res 2024; 134:1718-1751. [PMID: 38843294 PMCID: PMC11164543 DOI: 10.1161/circresaha.124.323658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024]
Abstract
The adult mammalian heart has limited endogenous regenerative capacity and heals through the activation of inflammatory and fibrogenic cascades that ultimately result in the formation of a scar. After infarction, massive cardiomyocyte death releases a broad range of damage-associated molecular patterns that initiate both myocardial and systemic inflammatory responses. TLRs (toll-like receptors) and NLRs (NOD-like receptors) recognize damage-associated molecular patterns (DAMPs) and transduce downstream proinflammatory signals, leading to upregulation of cytokines (such as interleukin-1, TNF-α [tumor necrosis factor-α], and interleukin-6) and chemokines (such as CCL2 [CC chemokine ligand 2]) and recruitment of neutrophils, monocytes, and lymphocytes. Expansion and diversification of cardiac macrophages in the infarcted heart play a major role in the clearance of the infarct from dead cells and the subsequent stimulation of reparative pathways. Efferocytosis triggers the induction and release of anti-inflammatory mediators that restrain the inflammatory reaction and set the stage for the activation of reparative fibroblasts and vascular cells. Growth factor-mediated pathways, neurohumoral cascades, and matricellular proteins deposited in the provisional matrix stimulate fibroblast activation and proliferation and myofibroblast conversion. Deposition of a well-organized collagen-based extracellular matrix network protects the heart from catastrophic rupture and attenuates ventricular dilation. Scar maturation requires stimulation of endogenous signals that inhibit fibroblast activity and prevent excessive fibrosis. Moreover, in the mature scar, infarct neovessels acquire a mural cell coat that contributes to the stabilization of the microvascular network. Excessive, prolonged, or dysregulated inflammatory or fibrogenic cascades accentuate adverse remodeling and dysfunction. Moreover, inflammatory leukocytes and fibroblasts can contribute to arrhythmogenesis. Inflammatory and fibrogenic pathways may be promising therapeutic targets to attenuate heart failure progression and inhibit arrhythmia generation in patients surviving myocardial infarction.
Collapse
Affiliation(s)
- Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen and Faculty of Medicine at the University of Freiburg, Freiburg, Germany
| | - Stefan Frantz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY
| |
Collapse
|
38
|
Jeong JH, John KA, Hong J, Lee JH. Heterogeneous nuclear ribonucleoprotein A2/B1, a key regulator of myocardial fibrosis. CLINICAL AND TRANSLATIONAL DISCOVERY 2024; 4:e319. [PMID: 40017489 PMCID: PMC11864630 DOI: 10.1002/ctd2.319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 03/01/2025]
Affiliation(s)
- Ji-Hoon Jeong
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, USA
| | - Kayode Abidemi John
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, USA
| | - Juyeong Hong
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, USA
| | - Ji Hoon Lee
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, Texas, USA
| |
Collapse
|
39
|
Li H, Song S, Shi A, Hu S. Identification of Potential lncRNA-miRNA-mRNA Regulatory Network Contributing to Arrhythmogenic Right Ventricular Cardiomyopathy. J Cardiovasc Dev Dis 2024; 11:168. [PMID: 38921668 PMCID: PMC11204167 DOI: 10.3390/jcdd11060168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) can lead to sudden cardiac death and life-threatening heart failure. Due to its high fatality rate and limited therapies, the pathogenesis and diagnosis biomarker of ARVC needs to be explored urgently. This study aimed to explore the lncRNA-miRNA-mRNA competitive endogenous RNA (ceRNA) network in ARVC. The mRNA and lncRNA expression datasets obtained from the Gene Expression Omnibus (GEO) database were used to analyze differentially expressed mRNA (DEM) and lncRNA (DElnc) between ARVC and non-failing controls. Differentially expressed miRNAs (DEmiRs) were obtained from the previous profiling work. Using starBase to predict targets of DEmiRs and intersecting with DEM and DElnc, a ceRNA network of lncRNA-miRNA-mRNA was constructed. The DEM and DElnc were validated by real-time quantitative PCR in human heart tissue. Protein-protein interaction network and weighted gene co-expression network analyses were used to identify hub genes. A logistic regression model for ARVC diagnostic prediction was established with the hub genes and their ceRNA pairs in the network. A total of 448 DEMs (282 upregulated and 166 downregulated) were identified, mainly enriched in extracellular matrix and fibrosis-related GO terms and KEGG pathways, such as extracellular matrix organization and collagen fibril organization. Four mRNAs and two lncRNAs, including COL1A1, COL5A1, FBN1, BGN, XIST, and LINC00173 identified through the ceRNA network, were validated by real-time quantitative PCR in human heart tissue and used to construct a logistic regression model. Good ARVC diagnostic prediction performance for the model was shown in both the training set and the validation set. The potential lncRNA-miRNA-mRNA regulatory network and logistic regression model established in our study may provide promising diagnostic methods for ARVC.
Collapse
Affiliation(s)
| | | | | | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China; (H.L.); (S.S.); (A.S.)
| |
Collapse
|
40
|
Ge LY, Wu TH, Liu YQ, Jiang C, Yin X. Management of experimental trabeculectomy filtering blebs via crosslinking of the scleral flap inhibited vascularization. Graefes Arch Clin Exp Ophthalmol 2024; 262:1507-1517. [PMID: 37943331 DOI: 10.1007/s00417-023-06306-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/10/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023] Open
Abstract
PURPOSE The aim of this study was to evaluate whether UVA-light-activated riboflavin-induced collagen crosslinking (UVA-CXL) can maintain the function of filtering blebs after trabeculectomy (TRAB) in rabbits. METHODS Thirty-six healthy rabbits were randomized to one of the following groups with 12 rabbits in each group: Trabeculectomy group (TRAB group), trabeculectomy combined with CXL group (CXL group), and trabeculectomy combined with MMC group (MMC group). Six rabbits of each group were performed with intraocular pressure (IOP), optical coherence tomography (OCT), and OCT angiography (OCTA). Bleb structure was observed via hematoxylin & eosin (H&E) and Masson staining. Immunohistochemistry, proteomic study, western blot, and tensile test were performed between CXL group and the control. In vitro, cell viability was evaluated by CCK-8 and Calcein/PI staining. TRPV4 and VEGF-a expression levels were measured by Q-PCR. Ca2+ concentration was observed with Fluo-4 AM. RESULTS The IOP and bleb median survival day were significantly modified in CXL (5.92 ± 0.32 mmHg and 15.5 days) than TRAB group (7.50 ± 0.43 mmHg and 9 days). The bleb area and height increased. CXL inhibited vascularization, and vascularization peaked at postoperative day (POD) 14 and then decreased gradually. In proteomic analyses, Z disc, actin filament binding, and sarcomere organization were significantly enriched. CXL inhibited scleral stress‒strain in tensile tests. Compared with TRAB group, TRPV4 expression was significantly increased, but VEGF-a and TGF-β1 levels were reduced in the CXL group in western blot. Meanwhile, TRPV4 expression colocalized with CD31. In vitro, CXL inhibited HUVECs cell viability. After CXL, expression level of TRPV4 was increased and calcium influx was activated, but VEGF-a was decreased in HUVECs. CONCLUSIONS This study demonstrates that intraoperative UV-RF CXL can significantly improve the success rate of TRAB via inhibiting filtering bleb vascularization. CXL increased sclera stiffness, in turn, induced TRPV4 activation, thus contributing to vascular endothelial cells suppression.
Collapse
Affiliation(s)
- Ling Ying Ge
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 21006, Jiangsu Province, China
| | - Tian Hong Wu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 21006, Jiangsu Province, China
| | - Yue Qi Liu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 21006, Jiangsu Province, China
| | - Chun Jiang
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 21006, Jiangsu Province, China
| | - Xue Yin
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 21006, Jiangsu Province, China.
| |
Collapse
|
41
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
42
|
Mei T, Cao H, Zhang L, Cao Y, Ma T, Sun Z, Liu Z, Hu Y, Le W. 3D Printed Conductive Hydrogel Patch Incorporated with MSC@GO for Efficient Myocardial Infarction Repair. ACS Biomater Sci Eng 2024; 10:2451-2462. [PMID: 38429076 DOI: 10.1021/acsbiomaterials.3c01837] [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] [Indexed: 03/03/2024]
Abstract
Myocardial infarction (MI) results in an impaired heart function. Conductive hydrogel patch-based therapy has been considered as a promising strategy for cardiac repair after MI. In our study, we fabricated a three-dimensional (3D) printed conductive hydrogel patch made of fibrinogen scaffolds and mesenchymal stem cells (MSCs) combined with graphene oxide (GO) flakes (MSC@GO), capitalizing on GO's excellent mechanical property and electrical conductivity. The MSC@GO hydrogel patch can be attached to the epicardium via adhesion to provide strong electrical integration with infarcted hearts, as well as mechanical and regeneration support for the infarcted area, thereby up-regulating the expression of connexin 43 (Cx43) and resulting in effective MI repair in vivo. In addition, MI also triggers apoptosis and damage of cardiomyocytes (CMs), hindering the normal repair of the infarcted heart. GO flakes exhibit a protective effect against the apoptosis of implanted MSCs. In the mouse model of MI, MSC@GO hydrogel patch implantation supported cardiac repair by reducing cell apoptosis, promoting gap connexin protein Cx43 expression, and then boosting cardiac function. Together, this study demonstrated that the conductive hydrogel patch has versatile conductivity and mechanical support function and could therefore be a promising candidate for heart repair.
Collapse
Affiliation(s)
- Tianxiao Mei
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200092, China
| | - Hao Cao
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Laihai Zhang
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Yunfei Cao
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Teng Ma
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Zeyi Sun
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200092, China
| | - Yihui Hu
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200092, China
| | - Wenjun Le
- Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200092, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200092, China
| |
Collapse
|
43
|
Sun M, Acosta AC, Emerick V, Adams S, Avila MY, Margo CE, Espana EM. Dysfunctional latent transforming growth factor β activation after corneal injury in a classical Ehlers-Danlos model. Matrix Biol 2024; 128:21-30. [PMID: 38340967 PMCID: PMC10996040 DOI: 10.1016/j.matbio.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/17/2023] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Patients with classical Ehlers Danlos syndrome (cEDS) suffer impaired wound healing and from scars formed after injuries that are atrophic and difficult to close surgically. Haploinsufficiency in COL5A1 creates systemic morphological and functional alterations in the entire body. We investigated mechanisms that impair wound healing from corneal lacerations (full thickness injuries) in a mouse model of cEDS (Col5a1+/-). We found that collagen V reexpression in this model is upregulated during corneal tissue repair and that wound healing is delayed, impaired, and results in large atrophic corneal scars. We noted that in a matrix with a 50 % content of collagen V, activation of latent Transforming Growth Factor (TGF) β is dysregulated. Corneal myofibroblasts with a haploinsufficiency of collagen V failed to mechanically activate latent TGF β. Second harmonic imaging microscopy showed a disorganized, undulated, and denser collagen matrix in our Col5a1+/- model that suggested alterations in the extracellular matrix structure and function. We hypothesize that a regenerated collagen matrix with only 50 % content of collagen V is not resistant enough mechanically to allow adequate activation of latent TGF β by fibroblasts and myofibroblasts.
Collapse
Affiliation(s)
- Mei Sun
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Ana Carolina Acosta
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Victoria Emerick
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Sheila Adams
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA
| | - Marcel Y Avila
- Departament of Ophthalmology, Universidad Nacional de Colombia, Bogota, Colombia
| | - Curtis E Margo
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA; Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Edgar M Espana
- Department of Ophthalmology, Cornea and External Disease, Morsani College of Medicine, University of South Florida, 13330 USF Laurel Dr., 4th floor, MDC11, Tampa, FL 33612, USA; Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
44
|
Sun AR, Hengst RM, Young JL. All the small things: Nanoscale matrix alterations in aging tissues. Curr Opin Cell Biol 2024; 87:102322. [PMID: 38277866 DOI: 10.1016/j.ceb.2024.102322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/28/2024]
Abstract
Cellular aging stems from multifaceted intra- and extracellular molecular changes that lead to the gradual deterioration of biological function. Altered extracellular matrix (ECM) properties that include biochemical, structural, and mechanical perturbations direct cellular- and tissue-level dysfunction. With recent advancements in high-resolution imaging modalities and nanomaterial strategies, the importance of nanoscale ECM features has come into focus. Here, we provide an updated window into micro- to nano-scale ECM properties that are altered with age and in age-related disease, and the impact these altered small-scale ECM properties have on cellular function. We anticipate future impactful research will incorporate nanoscale ECM features in the design of new biomaterials and call on the tissue biology field to work collaboratively with the nanomaterials community.
Collapse
Affiliation(s)
- Avery Rui Sun
- Mechanobiology Institute (MBI), National University of Singapore, 5A Engineering Drive 1, 117411, Singapore; Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, 117583, Singapore
| | - Ranmadusha M Hengst
- Mechanobiology Institute (MBI), National University of Singapore, 5A Engineering Drive 1, 117411, Singapore
| | - Jennifer L Young
- Mechanobiology Institute (MBI), National University of Singapore, 5A Engineering Drive 1, 117411, Singapore; Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, 117583, Singapore.
| |
Collapse
|
45
|
Feng J, Li Y, Li Y, Yin Q, Li H, Li J, Zhou B, Meng J, Lian H, Wu M, Li Y, Dou K, Song W, Lu B, Liu L, Hu S, Nie Y. Versican Promotes Cardiomyocyte Proliferation and Cardiac Repair. Circulation 2024; 149:1004-1015. [PMID: 37886839 DOI: 10.1161/circulationaha.123.066298] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND The adult mammalian heart is incapable of regeneration, whereas a transient regenerative capacity is maintained in the neonatal heart, primarily through the proliferation of preexisting cardiomyocytes. Neonatal heart regeneration after myocardial injury is accompanied by an expansion of cardiac fibroblasts and compositional changes in the extracellular matrix. Whether and how these changes influence cardiomyocyte proliferation and heart regeneration remains to be investigated. METHODS We used apical resection and myocardial infarction surgical models in neonatal and adult mice to investigate extracellular matrix components involved in heart regeneration after injury. Single-cell RNA sequencing and liquid chromatography-mass spectrometry analyses were used for versican identification. Cardiac fibroblast-specific Vcan deletion was achieved using the mouse strains Col1a2-2A-CreER and Vcanfl/fl. Molecular signaling pathways related to the effects of versican were assessed through Western blot, immunostaining, and quantitative reverse transcription polymerase chain reaction. Cardiac fibrosis and heart function were evaluated by Masson trichrome staining and echocardiography, respectively. RESULTS Versican, a cardiac fibroblast-derived extracellular matrix component, was upregulated after neonatal myocardial injury and promoted cardiomyocyte proliferation. Conditional knockout of Vcan in cardiac fibroblasts decreased cardiomyocyte proliferation and impaired neonatal heart regeneration. In adult mice, intramyocardial injection of versican after myocardial infarction enhanced cardiomyocyte proliferation, reduced fibrosis, and improved cardiac function. Furthermore, versican augmented the proliferation of human induced pluripotent stem cell-derived cardiomyocytes. Mechanistically, versican activated integrin β1 and downstream signaling molecules, including ERK1/2 and Akt, thereby promoting cardiomyocyte proliferation and cardiac repair. CONCLUSIONS Our study identifies versican as a cardiac fibroblast-derived pro-proliferative proteoglycan and clarifies the role of versican in promoting adult cardiac repair. These findings highlight its potential as a therapeutic factor for ischemic heart diseases.
Collapse
Affiliation(s)
- Jie Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Yandong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Yan Li
- State Key Laboratory of Tribology, Tsinghua University, Beijing, China (Y.L.)
| | - Qianqian Yin
- Institute of Medical Innovation and Research, Peking University Third Hospital, Peking University, Beijing, China (Q.Q.Y.)
| | - Haotong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Jun Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai (B.Z.)
| | - Jian Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Hong Lian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Mengge Wu
- Experimental Animal Center, Fuwai Central-China Hospital, Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou (M.G.W.)
| | - Yahuan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Kefei Dou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Weihua Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Bin Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Lihui Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (J.F., Y.D.L., H.T.L., J.L., J.M., H.L., Y.H.L., K.F.D., W.H.S., B.L., L.H.L., S.S.H., Y.N.)
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences (Y.N.)
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou (Y.N.)
| |
Collapse
|
46
|
Chang M, Wang H, Lei Y, Yang H, Xu J, Tang S. Proteomic study of left ventricle and cortex in rats after myocardial infarction. Sci Rep 2024; 14:6866. [PMID: 38514755 PMCID: PMC10958002 DOI: 10.1038/s41598-024-56816-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
Myocardial infarction (MI) induces neuroinflammation indirectly, chronic neuroinflammation may cause neurodegenerative diseases. Changes in the proteomics of heart and brain tissue after MI may shed new light on the mechanisms involved in neuroinflammation. This study explored brain and heart protein changes after MI with a data-independent acquisition (DIA) mode proteomics approach. Permanent ligation of the left anterior descending coronary artery (LAD) was performed in the heart of rats, and the immunofluorescence of microglia in the brain cortex was performed at 1d, 3d, 5d, and 7d after MI to detect the neuroinflammation. Then proteomics was accomplished to obtain the vital proteins in the heart and brain post-MI. The results show that the number of microglia was significantly increased in the Model-1d group, the Model-3d group, the Model-5d group, and the Model-7d group compared to the Sham group. Various proteins were obtained through DIA proteomics. Linking to key targets of brain disease, 14 proteins were obtained in the brain cortex. Among them, elongation of very long chain fatty acids protein 5 (ELOVL5) and ATP-binding cassette subfamily G member 4 (ABCG4) were verified through western blotting (WB). The results of WB were consistent with the proteomics results. Therefore, these proteins may be related to the pathogenesis of neuroinflammation after MI.
Collapse
Affiliation(s)
- Mengli Chang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huanhuan Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuxin Lei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Hongjun Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jing Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Shihuan Tang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
47
|
Delgado-Arija M, Genovés P, Pérez-Carrillo L, González-Torrent I, Giménez-Escamilla I, Martínez-Dolz L, Portolés M, Tarazón E, Roselló-Lletí E. Plasma fibroblast activation protein is decreased in acute heart failure despite cardiac tissue upregulation. J Transl Med 2024; 22:124. [PMID: 38297310 PMCID: PMC10832198 DOI: 10.1186/s12967-024-04900-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Cardiac fibroblast activation protein (FAP) has an emerging role in heart failure (HF). A paradoxical reduction in its levels in pathological conditions associated with acute processes has been observed. We aimed to identify FAP cardiac tissue expression and its relationship with the main cardiac fibrosis-related signaling pathways, and to compare plasma FAP levels in acute and chronic HF patients. METHODS Transcriptomic changes were assessed via mRNA/ncRNA-seq in left ventricle tissue from HF patients (n = 57) and controls (n = 10). Western blotting and immunohistochemistry were used to explore FAP protein levels and localization in cardiac tissue. ELISA was performed to examine plasma FAP levels in acute HF (n = 48), chronic HF (n = 15) and control samples (n = 7). RESULTS FAP overexpression in cardiac tissue is related to the expression of molecules directly involved in cardiac fibrosis, such as POSTN, THBS4, MFAP5, COL1A2 and COL3A1 (P < 0.001), and is directly and inversely related to pro- and antifibrotic microRNAs, respectively. The observed FAP overexpression is not reflected in plasma. Circulating FAP levels were lower in acute HF patients than in controls (P < 0.05), while chronic HF patients did not show significant changes. The clinical variables analyzed, such as functional class or etiology, do not affect plasma FAP concentrations. CONCLUSIONS We determined that in HF cardiac tissue, FAP is related to the main cardiac fibrosis signaling pathways as well as to pro- and antifibrotic microRNAs. Additionally, an acute phase of HF decreases plasma FAP levels despite the upregulation observed in cardiac tissue and regardless of other clinical conditions.
Collapse
Affiliation(s)
- Marta Delgado-Arija
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Patricia Genovés
- Department of Physiology, Faculty of Medicine, Universitat de València, Avd. de Blasco Ibañez, 15, 46010, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Lorena Pérez-Carrillo
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Irene González-Torrent
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Isaac Giménez-Escamilla
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Luis Martínez-Dolz
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
- Heart Failure and Transplantation Unit, Cardiology Department, University and Polytechnic La Fe Hospital, Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Manuel Portolés
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Estefanía Tarazón
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain.
| | - Esther Roselló-Lletí
- Clinical and Translational Research in Cardiology Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avd. Fernando Abril Martorell 106, 46026, Valencia, Spain.
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Avd. Monforte de Lemos 3-5, 28029, Madrid, Spain.
| |
Collapse
|
48
|
Kostin S, Richter M, Ganceva N, Sasko B, Giannakopoulos T, Ritter O, Szalay Z, Pagonas N. Atrial fibrillation in human patients is associated with increased collagen type V and TGFbeta1. INTERNATIONAL JOURNAL OF CARDIOLOGY. HEART & VASCULATURE 2024; 50:101327. [PMID: 38419608 PMCID: PMC10899732 DOI: 10.1016/j.ijcha.2023.101327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 03/02/2024]
Abstract
Background and aim Atrial fibrosis is an important factor in initiating and maintaining atrial fibrillation (AF). Collagen V belongs to fibrillar collagens. There are, however no data on collagen V in AF. The aim of this work was to study the quantity of collagen V and its relationship with the number of fibroblasts and TGF- b 1 expression in patients in sinus rhythm (SR) and in patients with atrial fibrillation (AF). Methods We used quantitative immuhistochemistry to study collagen V in right and left atrial biopsies obtained from 35 patients in SR, 35 patients with paroxysmal AF (pAF) and 27 patients with chronic, long-standing persistent AF (cAF). In addition, we have quantified the number of vimentin-positive fibroblasts and expression levels of TGF-β1. Results Compared to patients in SR, collagen V was increased 1.8- and 3.1-fold in patients with pAF and cAF, respectively. In comparison with SR patients, the number of vimentin-positive cells increased significantly 1.46- and 1.8-fold in pAF and cAF patients, respectively.Compared to SR patients, expression levels of TGF-ß1, expressed as fluorescence units per tissue area, was significantly increased by 77 % and 300 % in patients with pAF and cAF, respectively. Similar to intensity measurements, the number of TGFß1-positive cells per 1 mm2 atrial tissue increased significantly from 35.5 ± 5.5 cells in SR patients to 61.9 ± 12.4 cells in pAF and 131.5 ± 23.5 cells in cAF. In both types of measurements, there was a statistically significant difference between pAF and cAF groups. Conclusions This is the first study to show that AF is associated with increased expression levels of collagen V and TGF-ß1indicating its role in the pathogenesis of atrial fibrosis. In addition, increases in collagen V correlate with increased number of fibroblasts and TGF-β1 and are more pronounced in cAF patients than those in pAF patients.
Collapse
Affiliation(s)
- Sawa Kostin
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Natalia Ganceva
- Department of Anesthesiology and Intensive Care, Kerckoff-Clinic, Bad Nauheim, Germany
| | - Benjamin Sasko
- Medical Department II, Marien Hospital Herne, Ruhr-University of Bochum, Germany
| | | | - Oliver Ritter
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Cardiology, University Hospital Brandenburg, Brandenburg an der Havel, Germany
| | - Zoltan Szalay
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Nikolaos Pagonas
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Internal Medicine, University Hospital Ruppin-Brandenburg, Neuruppin, Germany
| |
Collapse
|
49
|
Van Wagoner DR. Collagen type V, interstitial fibrosis and the substrate for atrial fibrillation. IJC HEART & VASCULATURE 2024; 50:101356. [PMID: 38419609 PMCID: PMC10899731 DOI: 10.1016/j.ijcha.2024.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/02/2024]
Affiliation(s)
- David R. Van Wagoner
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, M/S ND50, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
50
|
Abstract
Permanent fibrosis and chronic deterioration of heart function in patients after myocardial infarction present a major health-care burden worldwide. In contrast to the restricted potential for cellular and functional regeneration of the adult mammalian heart, a robust capacity for cardiac regeneration is seen during the neonatal period in mammals as well as in the adults of many fish and amphibian species. However, we lack a complete understanding as to why cardiac regeneration takes place more efficiently in some species than in others. The capacity of the heart to regenerate after injury is controlled by a complex network of cellular and molecular mechanisms that form a regulatory landscape, either permitting or restricting regeneration. In this Review, we provide an overview of the diverse array of vertebrates that have been studied for their cardiac regenerative potential and discuss differential heart regeneration outcomes in closely related species. Additionally, we summarize current knowledge about the core mechanisms that regulate cardiac regeneration across vertebrate species.
Collapse
Affiliation(s)
- Michael Weinberger
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|