1
|
Ye D, Feng S, Yang X, Su Y, Zhang J, Feng H, Zhou M, Zhou B, Duan L, Peng T, Wang C. Hedgehog-interacting protein orchestrates alveologenesis and protects against bronchopulmonary dysplasia and emphysema. SCIENCE ADVANCES 2025; 11:eadu2958. [PMID: 40333979 PMCID: PMC12057671 DOI: 10.1126/sciadv.adu2958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025]
Abstract
Most of the lung's gas-exchange surface forms during alveologenesis and its disruption causes bronchopulmonary dysplasia (BPD) in infants, characterized by alveolar simplification and myofibroblast accumulation. BPD also increases the risk of adult emphysema, marked by alveolar loss. Despite this connection, mechanisms linking these conditions and effective treatments are still lacking. We identify hedgehog-interacting protein (HHIP), associated with both BPD and emphysema, as a critical regulator of alveologenesis. During this process, Hhip-expressing cells expanded, accompanied by hedgehog (Hh) signaling inhibition and myofibroblast transition. Stromal-specific Hhip deletion led to hyperactivation of Hh-IGF1 signaling axis, causing persistent SMA+ myofibroblasts and epithelial stem/progenitor cell senescence. Hyperactivation of this pathway was also observed in human BPD and hyperoxia-induced BPD models. Early Hhip deficiency resulted in adult emphysema with myofibroblast accumulation. We developed a therapeutic Fc-fused HHIP protein that mitigated BPD in neonatal mice and prevented adult emphysema. These findings establish HHIP as a critical regulator of alveologenesis and a therapeutic target for BPD and emphysema.
Collapse
Affiliation(s)
- Datian Ye
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Shiyun Feng
- Perfect Life Science Research Institute, Perfect (GuangDong) Co. Ltd., Zhongshan 528402, China
| | - Xinguo Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yanjing Su
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Jing Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Haixin Feng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Minqi Zhou
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bin Zhou
- University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lihui Duan
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tien Peng
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chaoqun Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
2
|
Wang Q, Ismahil MA, Zhu Y, Rokosh G, Hamid T, Zhou G, Pogwizd SM, Prabhu SD. CD206 +IL-4Rα + Macrophages Are Drivers of Adverse Cardiac Remodeling in Ischemic Cardiomyopathy. Circulation 2025. [PMID: 40308203 DOI: 10.1161/circulationaha.124.072411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND The role of cardiac CD (cluster of differentiation) 206+ macrophages in chronic heart failure (HF) is unknown. We examined whether CD206+ macrophages expressing IL (interleukin)-4Rα are key drivers of adverse left ventricular (LV) remodeling in HF. METHODS Adult C57BL/6 mice underwent nonreperfused myocardial infarction to induce HF. Macrophages in murine and human hearts were profiled using flow cytometry and immunostaining. In vivo myeloid-specific IL-4Rα deletion and intramyocardial macrophage adoptive transfer defined the functional effects of M[IL-4] macrophages. Antisense oligonucleotides were used for in vivo IL-4Rα gene silencing in mice. RESULTS CD206+ macrophages steadily expanded in hearts after myocardial infarction, such that at 8 weeks after myocardial infarction, they comprised ≈85% of all macrophages. These macrophages were proliferative, predominantly CCR2- (C-C motif chemokine receptor) and MHC (major histocompatibility complex) IIhi, and correlated with LV dysfunction and fibrosis. Nearly half of CD206+ macrophages expressed IL-4Rα, and the majority of CD206+IL-4Rα+ macrophages coexpressed profibrotic FIZZ (found in inflammatory zone) 1. IL-4-polarized bone marrow-derived CD206+ macrophages also exhibited marked upregulation of FIZZ1 and induced FIZZ1-dependent myofibroblast differentiation of both cardiac mesenchymal stem cells and cardiac fibroblasts, in part related to DLL-4/Jagged1-Notch1 signaling in cardiac mesenchymal stem cells. Intramyocardial adoptive transfer of M[IL-4], but not M[IL-10], CD206+ macrophages to naïve mice induced progressive LV remodeling over 4 weeks, increasing fibrosis, cardiomyocyte hypertrophy, and apoptosis. Myeloid-specific IL-4Rα gene deletion in HF (initiated 4 weeks after myocardial infarction) in IL-4Rαf/fLysM-CreERT2 mice significantly reduced CD206+ macrophage proliferation and effectively depleted CD206+IL-4Rα+ cardiac macrophages. This was associated with abrogation of LV remodeling progression, reduction of cardiac fibrosis, and improved neovascularization. In vivo IL-4Rα gene silencing in mice with established HF effectively depleted cardiac CD206+IL-4Rα+ macrophages and reversed LV remodeling, improving fibrosis, neovascularization, and dysfunction, and suppressed both local and systemic inflammation. Last, alternatively activated CD206+ and CD163+ macrophages were significantly expanded in human failing hearts and correlated with fibrosis. The majority of CD163+ macrophages expressed IL-4Rα and FIZZ3, the human homolog of FIZZ1. CONCLUSIONS Cardiac CD206+IL-4Rα+ macrophages proliferate and expand in HF and are key mediators of pathological remodeling and fibrosis, in part through the secretion of FIZZ1. Inhibition of CD206+ macrophage IL-4Rα signaling alleviates LV remodeling in ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Qiongxin Wang
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO (Q.W., M.A.I., Y.Z., G.R., T.H., S.D.P.)
- Department of Cardiology, Zhongnan Hospital of Wuhan University, China (Q.W.)
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| | - Mohamed Ameen Ismahil
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO (Q.W., M.A.I., Y.Z., G.R., T.H., S.D.P.)
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| | - Yujie Zhu
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO (Q.W., M.A.I., Y.Z., G.R., T.H., S.D.P.)
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| | - Gregg Rokosh
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO (Q.W., M.A.I., Y.Z., G.R., T.H., S.D.P.)
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| | - Tariq Hamid
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO (Q.W., M.A.I., Y.Z., G.R., T.H., S.D.P.)
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| | - Guihua Zhou
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| | - Steven M Pogwizd
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| | - Sumanth D Prabhu
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO (Q.W., M.A.I., Y.Z., G.R., T.H., S.D.P.)
- Department of Medicine (Cardiovascular Disease), University of Alabama at Birmingham (Q.W., M.A.I., Y.Z., G.R., T.H., G.Z., S.M.P., S.D.P.)
| |
Collapse
|
3
|
Kamm DR, Shaji A, Bohnert KL, Keener JD, Pathak A, Meyer GA. Adipose stromal cells in the human rotator cuff are resistant to fibrotic microenvironmental cues. J Physiol 2025. [PMID: 40198859 DOI: 10.1113/jp286563] [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: 09/12/2024] [Accepted: 02/14/2025] [Indexed: 04/10/2025] Open
Abstract
Rotator cuff tears are the most common upper extremity orthopaedic injury, causing degenerative changes within the bone, tendon, joint capsule, bursa and muscle. These degenerative changes are linked to poor rehabilitative and surgical outcomes, which has launched investigations into co-therapeutic biologics. Specifically, mesenchymal stem cells (MSCs) have shown promise in mitigating degenerative changes in animal models of rotator cuff tears, but reports of their impact on clinical outcomes remain mixed. Here we describe an alternative source of MSCs in the human shoulder, adipose stromal cells (ASCs) from the subacromial fat (SAF) pad. Compared to the gold-standard subcutaneous (SQ) fat, we show that SAF ASCs are less sensitive to chemical and mechanical fibrotic cues, (1) retaining smaller cell area with reduced actin stress fibre alignment across a range of physiological and pathological stiffnesses, (2) having reduced traction forces and extracellular matrix production, and (3) having reduced myofibroblastic conversion in response to cytokine challenge. Furthermore, we show that SAF ASCs enhance fusion of primary human myoblasts via paracrine signalling. Despite a fibrotic signature in SAF from rotator cuffs with tendon tears, SAF ASCs sourced from torn rotator cuffs were equally effective at resisting fibroblastic conversion and promoting myogenesis as those from intact rotator cuffs, further supporting autologous clinical use of these cells. In conclusion, this study describes human SAF ASCs as an alternative, and potentially superior, cell source for rotator cuff therapies. KEY POINTS: Adipose tissue within the rotator cuff is a novel and understudied source of therapeutic adipose stromal cells. Here, we detail the impact rotator cuff tears have on adipose tissue within the shoulder, its resident adipose stromal cells, and make a comparison of shoulder adipose stromal cells to subcutaneous adipose stromal cells. Rotator cuff tears cause fibrosis of rotator cuff adipose tissue; this fibrosis does not impact downstream adipose stromal cell morphology or pro-myogenic signaling. Rotator cuff adipose stromal cells resist fibrotic microenvironmental cues and have enhanced pro-myogenic paracrine signaling compared with traditional subcutaneous adipose stromal cells. Rotator cuff adipose stromal cells represent a new cell type that can be impactful in advancing rotator cuff therapies.
Collapse
Affiliation(s)
- Dakota R Kamm
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Akash Shaji
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - Jay D Keener
- Washington University in St. Louis, St. Louis, Missouri, USA
| | - Amit Pathak
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | |
Collapse
|
4
|
Kureel SK, Maroto R, Davis K, Sheetz M. Cellular mechanical memory: a potential tool for mesenchymal stem cell-based therapy. Stem Cell Res Ther 2025; 16:159. [PMID: 40165288 PMCID: PMC11960036 DOI: 10.1186/s13287-025-04249-x] [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: 08/21/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Recent studies have shown that mechanical properties such as extracellular matrix stiffness, fluid flow, weight loading, compression, and stretching can affect cellular functions. Some examples of cell responses to mechanical properties could be the migration of cancer cells from rigid to soft surfaces or the differentiation of fibroblasts into myofibroblasts. Cellular responses to mechanical changes can modify the insertion of proteins in the extracellular matrix (ECM), causing an increase in tissue stiffness with functional consequences. In general, mechanical and physical factors can affect any kind of cell phenotype in culture conditions and in vivo tissues. Cells sense mechanical stimuli by applying force and restructuring their shape and functions in response to the resistance of the stimuli. Furthermore, mechanical triggers can develop a "memory" for altering cellular plasticity and adaptation. This phenomenon is called cellular mechanical memory (CMM), a singular feature of mesenchymal stem cells (MSCs). Controlled targeting of CMM may resolve the scarcity of viable cells needed for cell based therapy (CBT) and implement studies concerning cancer research, fibrosis, and senescence. This review focusses on cells from the mesodermal lineage, such as MSCs, fibroblasts and chondrocytes, and the role of CMM as a potential target for CBT.
Collapse
Affiliation(s)
- Sanjay Kumar Kureel
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Rosario Maroto
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kristen Davis
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Michael Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
5
|
Walker JT, Cooper TT, Dunmore-Buyze J, Serack FE, Brooks C, Grant A, Drangova M, Lajoie G, Dekaban GA, Flynn LE. Syngeneic adipose-derived stromal cells modulate the immune response but have limited persistence within decellularized adipose tissue implants in C57BL/6 mice. Acta Biomater 2025; 195:169-182. [PMID: 39922513 DOI: 10.1016/j.actbio.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/14/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
The delivery of adipose-derived stromal cells (ASCs) on cell-instructive decellularized adipose tissue (DAT) scaffolds is a promising strategy for stimulating host-derived soft tissue regeneration. However, a better understanding of the mechanisms through which ASCs modulate regeneration in vivo is needed to harness these cells more effectively. In this study, DAT scaffolds, both with and without seeded syngeneic DsRED+ mouse ASCs, were implanted into immunocompetent C57BL/6 mice. Downstream analyses focused on assessing donor ASC persistence and phenotype, as well as the effects of ASC seeding on host macrophage polarization and the perfused host vascular network. Notably, most donor ASCs were cleared from the scaffolds by 2 weeks. Mass spectrometry-based proteomics indicated that the transplanted ASCs maintained their pre-implantation phenotype up to 1 week in vivo, suggesting that the cells were not undergoing programmed cell death. A higher fraction of the infiltrating host macrophages expressed CD68 and Arginase-1 in the ASC-seeded implants up to 1-week post-implantation. Interestingly, a small population of phagocytic macrophages, identified by uptake of DsRED protein, was present in the DAT implants in the first 2 weeks and showed enhanced expression of CD68, Arginase-1, and CD163, along with reduced expression of iNOS. MicroCT angiography revealed a similar perfused vessel network in the seeded and unseeded groups at 4- and 8-weeks post-implantation. Overall, seeding with syngeneic ASCs modulated the host macrophage response to the DAT bioscaffolds at early timepoints, but did not impact long-term regenerative outcomes, potentially due to the rapid clearance of the donor cell population in this model. STATEMENT OF SIGNIFICANCE: Decellularized adipose tissue (DAT) is a promising biomaterial for treating soft tissue defects. Seeding with adipose-derived stromal cells (ASCs) can augment fat regeneration within DAT in pre-clinical models, but our understanding of how ASCs contribute to tissue regeneration in vivo remains limited. Furthermore, ASC clearance from implanted biomaterials is well described, but poorly understood. Here, ASC-seeded DAT was implanted subcutaneously in immunocompetent mice to assess how ASCs altered the host macrophage response, functional vascular regeneration, and long-term integration with the host tissues. Additionally, ASC phenotype and persistence were assessed to determine how these cells might be cleared from the implants. Such understanding is critical to design biomaterials that can better harness the therapeutic benefits of ASCs.
Collapse
Affiliation(s)
- John T Walker
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Tyler T Cooper
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Joy Dunmore-Buyze
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Fiona E Serack
- School of Biomedical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Aaron Grant
- Division of Plastic and Reconstructive Surgery, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, Ontario N6A 5C1, Canada
| | - Maria Drangova
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada; Department of Medical Biophysics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Gilles Lajoie
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Gregory A Dekaban
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A 5C1, Canada; Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Lauren E Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada; School of Biomedical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario N6A 3K7, Canada; Department of Chemical and Biochemical Engineering, Faculty of Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada.
| |
Collapse
|
6
|
Chen X, Ji X, Lao Z, Pan B, Qian Y, Yang W. Role of YAP/TAZ in bone diseases: A transductor from mechanics to biology. J Orthop Translat 2025; 51:13-23. [PMID: 39902099 PMCID: PMC11787699 DOI: 10.1016/j.jot.2024.12.003] [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: 08/01/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Wolff's Law and the Mechanostat Theory elucidate how bone tissues detect and convert mechanical stimuli into biological signals, crucial for maintaining bone equilibrium. Abnormal mechanics can lead to diseases such as osteoporosis, osteoarthritis, and nonunion fractures. However, the detailed molecular mechanisms by which mechanical cues are transformed into biological responses in bone remain underexplored. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of bone homeostasis, are instrumental in this process. Emerging research highlights bone cells' ability to sense various mechanical stimuli and relay these signals intracellularly. YAP/TAZ are central in receiving these mechanical cues and converting them into signals that influence bone cell behavior. Abnormal YAP/TAZ activity is linked to several bone pathologies, positioning these proteins as promising targets for new treatments. Thus, this review aims to provide an in-depth examination of YAP/TAZ's critical role in the interpretation of mechanical stimuli to biological signals, with a special emphasis on their involvement in bone cell mechanosensing, mechanotransduction, and mechanoresponse. The translational potential of this article: Clinically, appropriate stress stimulation promotes fracture healing, while bed rest can lead to disuse osteoporosis and excessive stress can cause osteoarthritis or bone spurs. Recent advancements in the understanding of YAP/TAZ-mediated mechanobiological signal transduction in bone diseases have been significant, yet many aspects remain unknown. This systematic review summarizes current research progress, identifies unaddressed areas, and highlights potential future research directions. Advancements in this field facilitate a deeper understanding of the molecular mechanisms underlying bone mechanics regulation and underscore the potential of YAP/TAZ as therapeutic targets for bone diseases such as fractures, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Xing Ji
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
7
|
Alabrahim OAA, Fytory M, Abou-Shanab AM, Lababidi J, Fritzsche W, El-Badri N, Azzazy HMES. A biocompatible β-cyclodextrin inclusion complex containing natural extracts: a promising antibiofilm agent. NANOSCALE ADVANCES 2025; 7:1405-1420. [PMID: 39845135 PMCID: PMC11748956 DOI: 10.1039/d4na00916a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Biofilms formed by several bacterial strains still pose a significant challenge to healthcare due to their resistance to conventional treatment approaches, including antibiotics. This study explores the potential of loading natural extracts with antimicrobial activities into β-cyclodextrin (βCD) nanoparticles, which are FDA-approved and have superior biocompatibility owing to their cyclic sugar structures, for biofilm eradication. An inclusion complex of βCD carrying Boswellia sacra essential oils (BOS) was prepared and characterized with regard to its physicochemical properties, antimicrobial efficacy, and antibiofilm activities. Encapsulation of BOS into βCD significantly enhanced the antimicrobial activity of BOS by 4-fold against Gram-positive (Staphylococcus aureus and Bacillus subtilis) and by 8-fold against Gram-negative (Escherichia coli and Pseudomonas putida) bacteria, with minimum inhibitory concentrations ranging from 2.5 to 5 mg mL-1. Furthermore, the BOS-βCD complex demonstrated a dual-action against bacterial biofilms where it prevented biofilm formation and disrupted established biofilms. This resulted in a significant reduction in biofilm biomass, with prevention and disruption rates reaching up to 93.78% and 82.17%, respectively. Additionally, the formula revealed an excellent biocompatibility profile with no induction of oxidative stress in human skin fibroblast cells. Our findings suggest that βCD nanoparticles loaded with BOS essential oils hold promise as an effective formula for preventing the formation of bacterial biofilms and combating preformed ones for use in relevant medical applications.
Collapse
Affiliation(s)
- Obaydah Abd Alkader Alabrahim
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt +202 2615 2559
| | - Mostafa Fytory
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt +202 2615 2559
- Material Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University 62511 Beni-Suef Egypt
| | - Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology Giza 12578 Egypt
| | - Jude Lababidi
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt +202 2615 2559
| | - Wolfgang Fritzsche
- Department of Nanobiophotonics, Leibniz Institute of Photonic Technology Jena 07745 Germany
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology Giza 12578 Egypt
| | - Hassan Mohamed El-Said Azzazy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt +202 2615 2559
- Department of Nanobiophotonics, Leibniz Institute of Photonic Technology Jena 07745 Germany
| |
Collapse
|
8
|
Koczkowska M, Kostecka A, Zawrzykraj M, Myszczyński K, Skoniecka A, Deptuła M, Tymińska A, Czerwiec K, Jąkalski M, Zieliński J, Crossman DK, Crowley MR, Cichorek M, Skowron PM, Pikuła M, Piotrowski A. Identifying differentiation markers between dermal fibroblasts and adipose-derived mesenchymal stromal cells (AD-MSCs) in human visceral and subcutaneous tissues using single-cell transcriptomics. Stem Cell Res Ther 2025; 16:64. [PMID: 39934849 PMCID: PMC11818286 DOI: 10.1186/s13287-025-04185-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: 05/16/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stromal cells (AD-MSCs) and fibroblasts are both widely used in regenerative medicine, demonstrating significant potential for personalized cell therapy. A major challenge in their use lies in their high biological similarity, encompassing morphology, differentiation capabilities, and flow cytometric markers, making their distinction difficult. METHODS In our study, we aimed to compare AD-MSCs obtained from two types of adipose tissue, subcutaneous and visceral, alongside skin fibroblasts. Notably, all tissue samples were sourced from the same donors. We analyzed the cells for surface antigens via flow cytometry and conducted single-cell RNA sequencing, followed by verification with quantitative PCR (qPCR). RESULTS Our results revealed phenotypic similarities between the isolated AD-MSCs and dermal fibroblasts, particularly in the expression of markers characteristic of AD-MSCs. However, through in-depth analyses, we identified distinct differences between these cell types. Specifically, we pinpointed 30 genes exhibiting the most significant variations in expression between AD-MSCs and fibroblasts. These genes are associated with biological processes such as tissue remodeling, cell movement, and activation in response to external stimuli. Among them, MMP1, MMP3, S100A4, CXCL1, PI16, IGFBP5, COMP were further validated using qPCR, clearly demonstrating their potential to differentiate between AD-MSCs and fibroblasts. CONCLUSIONS Our scRNA-seq analysis elucidates the transcriptional landscape of AD-MSCs and fibroblasts with unprecedented resolution, highlighting both the population-specific markers and the intrapopulation heterogeneity. Our findings underscore the importance of employing high-resolution techniques for cell identification.
Collapse
Affiliation(s)
| | - Anna Kostecka
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Małgorzata Zawrzykraj
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Kamil Myszczyński
- Centre of Biostatistics and Bioinformatics Analysis, Medical University of Gdansk, Gdansk, Poland
| | - Aneta Skoniecka
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Milena Deptuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Agata Tymińska
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Katarzyna Czerwiec
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Jąkalski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Jacek Zieliński
- Department of Surgical Oncology, Transplant Surgery and General Surgery, Medical University of Gdansk, Gdansk, Poland
| | - David K Crossman
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael R Crowley
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mirosława Cichorek
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Michał Pikuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland.
| | - Arkadiusz Piotrowski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland.
| |
Collapse
|
9
|
Rai D, Sardar A, Raj A, Maji B, Verma S, Tripathi AK, Gupta S, Sharma A, Dhar YV, Trivedi R. miR4352b a cross-species modulator of SOSTDC1, targets dual pathway to regulate bone health and fracture healing. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167514. [PMID: 39326466 DOI: 10.1016/j.bbadis.2024.167514] [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: 03/29/2024] [Revised: 08/28/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
Mutations in SOST can lead to various monogenic bone diseases. Its paralog, SOSTDC1, shares 55 % protein sequence homology and belongs to the BMP antagonist class. Sostdc1-/- mice exhibit distinct effects on cortical and trabecular bone. Genetic polymorphisms in SOSTDC1 impacting peak bone mass makes SOSTDC1 gene, a candidate for influencing BMD variation in humans. SOSTDC1 is upregulated in bone loss conditions, altering BMP-responsive genes and signaling modulators, suggesting its dual BMP/Wnt antagonist role may enhance both pathways. Overexpression of SOSTDC1 confirmed its role as an osteogenic antagonist. Glycine max (Soy)-derived miR4352b, identified for cross-kingdom applications, precisely targets SOSTDC1, a key regulator of bone. SOSTDC1 competitively binds to BMP2 receptor, BMPR1A. Gma-miR4352b suppresses SOSTDC1 expression, enhancing osteogenesis and countering SOSTDC1's inhibition of osteogenic potential. Modeling estrogen deficiency to mimic elevated SOSTDC1 levels, we observed an inverse correlation with SOSTDC1 expression, while serum BMP2 and PINP levels increased following gma-miR4352b supplementation. In fracture healing, SOSTDC1's crucial role becomes evident in conditions of delayed fracture healing. As healing progresses, SOSTDC1 expression decreases. Gma-miR4352b, compared to scrambled miRNA, remarkably promotes callus formation, achieving 68 % healing by day 10, surpassing the scrambled group at 44 %. By the day 13, the treatment group exhibits advanced healing, challenging to find the callus, while the scrambled group maintains a healing rate similar to day10. The accelerated healing in the treatment group underscores the importance of SOSTDC1 in influencing early fracture healing, potentially through the activation of both BMP2 and Wnt signaling pathways.
Collapse
Affiliation(s)
- Divya Rai
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anirban Sardar
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anuj Raj
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Bhaskar Maji
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shikha Verma
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | | | - Sanchita Gupta
- Computational Biology lab, CSIR-National Botanical Research Institute, 226001, India
| | - Ashish Sharma
- CSIR- Central Institute of Medicinal and Aromatic Plants (CSIR-CIMAP) P.O. CIMAP, Near Kukrail Picnic Spot, Lucknow 226 015, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Yogeshwar Vikram Dhar
- Computational Biology lab, CSIR-National Botanical Research Institute, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ritu Trivedi
- Endocrinology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
10
|
Barasa P, Simoliunas E, Grybas A, Zilinskaite-Tamasauske R, Dasevicius D, Alksne M, Rinkunaite I, Buivydas A, Baltrukonyte E, Tamulyte R, Megur A, Verkauskas G, Baltriukiene D, Bukelskiene V. Development of multilayered artificial urethra graft for urethroplasty. J Biomed Mater Res A 2025; 113:e37796. [PMID: 39268589 DOI: 10.1002/jbm.a.37796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/11/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
To enhance the treatment of patients' urethral defects, such as strictures and hypospadias, we investigated the potential of using artificial urethral tissue. Our study aimed to generate this tissue and assess its effectiveness in a rabbit model. Two types of bioprinted grafts, based on methacrylated gelatin-silk fibroin (GelMA-SF) hydrogels, were produced: acellular, as well as loaded with autologous rabbit stem cells. Rabbit adipose stem cells (RASC) were differentiated toward smooth muscle in the GelMA-SF hydrogel, while rabbit buccal mucosa stem cells (RBMC), differentiated toward the epithelium, were seeded on its surface, forming two layers of the cell-laden tissue. The constructs were then reinforced with polycaprolactone-polylactic acid meshes to create implantable multilayered artificial urethral grafts. In vivo experiments showed that the cell-laden tissue integrated into the urethra with less fibrosis and inflammation compared to its acellular counterpart. Staining to trace the implanted cells confirmed integration into the host organism 3 months postsurgery.
Collapse
Affiliation(s)
- Povilas Barasa
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Egidijus Simoliunas
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Aivaras Grybas
- Urology Center, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Ramune Zilinskaite-Tamasauske
- Children's Surgery, Orthopaedic and Traumatology Centre, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Darius Dasevicius
- Centre of Pathology, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Milda Alksne
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ieva Rinkunaite
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Andrius Buivydas
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Emilija Baltrukonyte
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rimgaile Tamulyte
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | | | - Gilvydas Verkauskas
- Children's Surgery, Orthopaedic and Traumatology Centre, Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Daiva Baltriukiene
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Virginija Bukelskiene
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
11
|
Ma Z, Chawla S, Lan X, Zhou E, Mulet-Sierra A, Kunze M, Sommerfeldt M, Adesida AB. Functional heterogeneity of meniscal fibrochondrocytes and microtissue models is dependent on modality of fibrochondrocyte isolation. Cell Prolif 2025; 58:e13735. [PMID: 39377189 DOI: 10.1111/cpr.13735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 10/09/2024] Open
Abstract
Collagenase digestion (d) and cellular outgrowth (og) are the current modalities of meniscus fibrochondrocytes (MFC) isolation for bioengineering and mechanobiology-related studies. However, the impact of these modalities on study outcomes is unknown. Here, we show that og- and d-isolated MFC have distinct proliferative capacities, transcriptomic profiles via RNA sequencing (RNAseq), extracellular matrix (ECM)-forming, and migratory capacities. Our data indicate that microtissue pellet models developed from og-isolated MFC display a contractile phenotype with higher expressions of alpha-smooth muscle actin (ACTA2) and transgelin (TAGLN) and are mechanically stiffer than their counterparts from d-MFC. Moreover, we introduce a novel method of MFC isolation designated digestion-after-outgrowth (dog). The transcriptomic profile of dog-MFC is distinct from d- and og-MFC, including a higher expression of mechanosensing caveolae-associated caveolin-1 (CAV1). Additionally, dog-MFC were superior chondrogenically and generated larger-size microtissue pellet models containing a higher frequency of smaller collagen fibre diameters. Thus, we demonstrate that the modalities of MFC isolation influence the downstream outcomes of bioengineering and mechanobiology-related studies.
Collapse
Affiliation(s)
- Zhiyao Ma
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shikha Chawla
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaoyi Lan
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Eva Zhou
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Melanie Kunze
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mark Sommerfeldt
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Adetola B Adesida
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
12
|
Papp FR, Katko M, Csiki R, Galgoczi E, Molnar Z, Erdei A, Bodor M, Steiber Z, Ujhelyi B, Nagy EV. Characteristics of Hyaluronan Metabolism During Myofibroblast Differentiation in Orbital Fibroblasts. Invest Ophthalmol Vis Sci 2024; 65:13. [PMID: 39504052 PMCID: PMC11549924 DOI: 10.1167/iovs.65.13.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/12/2024] [Indexed: 11/11/2024] Open
Abstract
Purpose To study the impact of myofibroblast differentiation (MD) on hyaluronan (HA) turnover in orbital fibroblasts (OFs) focusing on the expression of its key enzymes and their potential implications in the pathogenesis of thyroid eye disease (TED). Methods Primary cultures of OFs were established from tissue samples (TED OFs, n = 4; non-TED OFs, n = 5). MD was induced by TGF-β1 (5 ng/mL). Measurements were performed after 24- and 72-hour treatments. The proliferation rate was determined by 5-bromo-2'-deoxyuridine (BrdU) incorporation. HA level and size were measured using an aggrecan-based ELISA-like method and agarose gel electrophoresis, respectively. mRNA expressions of myofibroblast markers and enzymes with a role in HA metabolism were determined using real-time PCR. Results Upregulation of type I collagen alpha1 chain, alpha-smooth muscle actin, and fibronectin indicated that OFs underwent MD after stimulation by TGF-β. After 72 hours, proliferation of untreated cultures declined, but it remained higher in myofibroblasts. Pericellular HA content, but not HA in the supernatant of myofibroblasts, increased compared to untreated cells. TGF-β was a potent stimulator of hyaluronan synthase 1 (HAS1) expression. The expression of hyaluronidase-1 and cell migration-inducing protein (CEMIP) diminished following MD, whereas the expression of transmembrane protein 2, the regulator of HA catabolism through CEMIP, was elevated. The size distribution of HA shifted toward a high-molecular-weight form following treatment with TGF-β. Conclusions OFs undergoing MD are characterized by decreased HA turnover as a consequence of the inhibition of hyaluronidases and HAS1 induction. Our results suggest that hyaluronidases could be potential targets in the treatment of TED.
Collapse
Affiliation(s)
- Fruzsina R. Papp
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Monika Katko
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Robert Csiki
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Erika Galgoczi
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsanett Molnar
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Annamaria Erdei
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Miklos Bodor
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zita Steiber
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Bernadett Ujhelyi
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Endre V. Nagy
- Division of Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
13
|
Liu D, Wang X, Gao C, Zhang Z, Wang Q, Pei Y, Wang H, Tang Y, Li K, Yu Y, Cai Q, Zhang X. Biodegradable Piezoelectric-Conductive Integrated Hydrogel Scaffold for Repair of Osteochondral Defects. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409400. [PMID: 39267457 DOI: 10.1002/adma.202409400] [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: 07/01/2024] [Revised: 08/20/2024] [Indexed: 09/17/2024]
Abstract
Osteochondral injury is a prevalent condition for which no specific treatment is currently available. This study presents a piezoelectric-conductive scaffold composed of a piezoelectric cartilage-decellularized extracellular matrix (dECM) and piezoelectric-conductive modified gelatin (Gel-PC). The piezoelectricity of the scaffold is achieved through the modification of diphenylalanine (FF) assembly on the pore surface, while the conductive properties of scaffold are achieved by the incorporating poly(3,4-ethylenedioxythiophene). In vitro experiments demonstrate that bone marrow mesenchymal stem cells (BMSCs) undergo biphasic division during differentiation. In vivo studies using a Parma pig model of osteochondral defects demonstrate that the piezoelectric-conductive scaffold exhibits superior reparative efficacy. Notably, the generation of electrical stimulation is linked to joint movement. During joint activity, mechanical forces compress the scaffold, leading to deformation and the subsequent generation of an electric potential difference. The positive charges accumulated on the upper layer of the scaffold attract BMSCs, promoting their migration to the upper layer and chondrogenic differentiation. Meanwhile, the negative charges in the lower layer induce the osteogenic differentiation of BMSCs. Overall, this piezoelectric-conducive scaffold provides a promising platform for the effective repair of osteochondral defects.
Collapse
Affiliation(s)
- Dingge Liu
- Institute of Sports MedicineBeijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Xinyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chenyuan Gao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhihua Zhang
- Institute of Sports MedicineBeijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Qi Wang
- Institute of Sports MedicineBeijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Yin Pei
- Institute of Sports MedicineBeijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Haijun Wang
- Institute of Sports MedicineBeijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| | - Yujing Tang
- SINOPEC Beijing Research Institute of Chemical Industry Co. Ltd, Beijing, 100728, China
| | - Ke Li
- SINOPEC Beijing Research Institute of Chemical Industry Co. Ltd, Beijing, 100728, China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xin Zhang
- Institute of Sports MedicineBeijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China
| |
Collapse
|
14
|
Park G, Hwang DY, Kim DY, Han JY, Lee E, Hwang H, Park JS, Kim DW, Hong S, Yim SV, Hong HS, Son Y. Identification of CD141 +vasculogenic precursor cells from human bone marrow and their endothelial engagement in the arteriogenesis by co-transplantation with mesenchymal stem cells. Stem Cell Res Ther 2024; 15:388. [PMID: 39482744 PMCID: PMC11526567 DOI: 10.1186/s13287-024-03994-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is a condition characterized by insufficient blood flow to the lower limbs, resulting in severe ischemia and potentially leading to amputation. This study aims to identify novel vasculogenic precursor cells (VPCs) in human bone marrow and evaluate their efficacy in combination with bone marrow-derived mesenchymal stem cells (BM-MSCs) for the treatment of CLI. METHODS Ex vivo cultured VPCs and BM-MSCs from bone marrow were characterized and their effects on neovascularization and long-term tissue regeneration were tested in a mouse CLI model. RESULTS VPCs, expressing high levels of hepatocyte growth factor and c-MET, were identified from human bone marrow aspirates. These cells exhibited strong vasculogenic capacity in vitro but possessed a cellular phenotype distinct from those of previously reported endothelial precursor cells in circulation or cord blood. They also expressed most surface markers of BM-MSCs and demonstrated multipotent differentiation ability. Screening of 376 surface markers revealed that VPCs uniquely display CD141 (thrombomodulin). CD141+VPCs are present in BM aspirates as a rare population and can be expanded ex vivo with a population doubling time of approximately 20 h, generating an elaborate vascular network even under angiogenic factor-deficient conditions and recruiting BM-MSCs to the network as pericyte-like cells. Intramuscular transplantation of a combination of human CD141+VPCs and BM-MSCs at a ratio of 2:1 resulted in limb salvage, blood flow recovery, and regeneration of large vessels in the femoral artery-removed CLI model, with an efficacy superior to that of singular transplantation. Importantly, large arteries and arterioles in dual cell transplantation expressed human CD31 in the intima and human α-smooth muscle actin in media layer at 4 and 12 weeks, likely indicating their lineage commitment to endothelial cells and vascular smooth muscle, respectively, in vivo. CONCLUSION Dual-cell therapy using BM-derived CD141+ VPCs and BM-MSCs holds potential for further development in clinical trials to treat peripheral artery disease and diabetic ulcers.
Collapse
Affiliation(s)
- Gabee Park
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Dae Yeon Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Do Young Kim
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Ji Young Han
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Euiseon Lee
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Hwakyung Hwang
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Jeong Seop Park
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea
| | - Dae Wook Kim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Seonmin Hong
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
| | - Sung Vin Yim
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduated School, Kyung Hee University, Seoul, Korea.
- East-West Medical Research Institute, Kyung Hee University, Seoul, Korea.
| | - Youngsook Son
- R&D Center, Elphis Cell Therapeutics Inc, Yong In, 17095, Korea.
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea.
| |
Collapse
|
15
|
Arciero I, Buonvino S, Melino S. Slow H 2S-Releasing Donors and 3D Printable Arrays Cellular Models in Osteo-Differentiation of Mesenchymal Stem Cells for Personalized Therapies. Biomolecules 2024; 14:1380. [PMID: 39595557 PMCID: PMC11592188 DOI: 10.3390/biom14111380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The effects of the hydrogen sulfide (H2S) slow-releasing donor, named GSGa, a glutathione-conjugate water-soluble garlic extract, on human mesenchymal stem cells (hMSCs) in both bidimensional (2D) and three-dimensional (3D) cultures were investigated, demonstrating increased expression of the antioxidant enzyme HO-1 and decreased expression of the pro-inflammatory cytokine interleukin-6 (IL-6). The administration of the H2S donor can therefore increase the expression of antioxidant enzymes, which may have potential therapeutic applications in osteoarthritis (OA). Moreover, GSGa was able to promote the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), but not of cardiac mesenchymal stem cells (cMSCs) in a 2D culture system. This result highlights the varying sensitivity of hMSCs to the H2S donor GSGa, suggesting that the induction of osteogenic differentiation in stem cells by chemical factors is dependent on the tissue of origin. Additionally, a 3D-printable mesenchymal stem cells-bone matrix array (MSCBM), designed to closely mimic the stiffness of bone tissue, was developed to serve as a versatile tool for evaluating the effects of drugs and stem cells on bone repair in chronic diseases, such as OA. We demonstrated that the osteogenic differentiation process in cMSCs can be induced just by simulating bone stiffness in a 3D system. The expression of osteocalcin, RUNX2, and antioxidant enzymes was also assessed after treating MSCs with GSGa and/or increasing the stiffness of the culture environment. The printability of the array may enable better customization of the cavities, enabling an accurate replication of real bone defects. This could optimize the BM array to mimic bone defects not only in terms of stiffness, but also in terms of shape. This culture system may enable a rapid screening of antioxidant and anti-inflammatory compounds, facilitating a more personalized approach to regenerative therapy.
Collapse
Affiliation(s)
- Ilaria Arciero
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy;
| | - Silvia Buonvino
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| | - Sonia Melino
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
16
|
Zhu YF, Wan MC, Gao P, Shen MJ, Zhu YN, Hao JX, Lu WC, Wang CY, Tay F, Ehrlich H, Niu LN, Jiao K. Fibrocyte: A missing piece in the pathogenesis of fibrous epulis. Oral Dis 2024; 30:4376-4389. [PMID: 38148479 DOI: 10.1111/odi.14847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/18/2023] [Accepted: 12/10/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVES To explore the role of fibrocytes in the recurrence and calcification of fibrous epulides. METHODS Different subtypes of fibrous epulides and normal gingival tissue specimens were first collected for histological and immunofluorescence analyses to see if fibrocytes were present and whether they differentiated into myofibroblasts and osteoblasts upon stimulated by transforming growth factor-β1 (TGF-β1). Electron microscopy and elemental analysis were used to characterize the extracellular microenvironment in different subtypes of fibrous epulides. Human peripheral blood mononuclear cells (PBMCs) were subsequently isolated from in vitro models to mimic the microenvironment in fibrous epulides to identify whether TGF-β1 as well as the calcium and phosphorus ion concentration in the extracellular matrix (ECM) of a fibrous epulis trigger fibrocyte differentiation. RESULTS Fibrous epulides contain fibrocytes that accumulate in the local inflammatory environment and have the ability to differentiate into myofibroblasts or osteoblasts. TGF-β1 promotes fibrocytes differentiation into myofibroblasts in a concentration-dependent manner, while TGF-β1 stimulates the fibrocytes to differentiate into osteoblasts when combined with a high calcium and phosphorus environment. CONCLUSIONS Our study revealed fibrocytes play an important role in the fibrogenesis and osteogenesis in fibrous epulis, and might serve as a therapeutic target for the inhibition of recurrence of fibrous epulides.
Collapse
Affiliation(s)
- Yi-Fei Zhu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Mei-Chen Wan
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Peng Gao
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Min-Juan Shen
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yi-Na Zhu
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jia-Xin Hao
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Wei-Cheng Lu
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Chen-Yu Wang
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Franklin Tay
- The Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hermann Ehrlich
- Institute of Electronic and Sensor Materials, Freiberg, Germany
| | - Li-Na Niu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Prosthodontics, State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
17
|
Ahmed AI, Dowidar MF, Negm AF, Abdellatif H, Alanazi A, Alassiri M, Samy W, Mekawy DM, Abdelghany EMA, El-Naseery NI, Ibrahem MA, Albadawi EA, Salah W, Eldesoqui M, Tîrziu E, Bucur IM, Arisha AH, Khamis T. Bone marrow mesenchymal stem cells expressing Neat-1, Hotair-1, miR-21, miR-644, and miR-144 subsided cyclophosphamide-induced ovarian insufficiency by remodeling the IGF-1-kisspeptin system, ovarian apoptosis, and angiogenesis. J Ovarian Res 2024; 17:184. [PMID: 39267091 PMCID: PMC11396253 DOI: 10.1186/s13048-024-01498-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 08/14/2024] [Indexed: 09/14/2024] Open
Abstract
Ovarian insufficiency is one of the common reproductive disorders affecting women with limited therapeutic aids. Mesenchymal stem cells have been investigated in such disorders before yet, the exact mechanism of MSCs in ovarian regeneration regarding their epigenetic regulation remains elusive. The current study is to investigate the role of the bone marrow-derived mesenchymal stem cells (BM-MSCs) lncRNA (Neat-1 and Hotair1) and miRNA (mir-21-5p, mir-144-5p, and mir-664-5p) in mitigating ovarian granulosa cell apoptosis as well as searching BM-MSCs in altering the expression of ovarian and hypothalamic IGF-1 - kisspeptin system in connection to HPG axis in a cyclophosphamide-induced ovarian failure rat model. Sixty mature female Sprague Dawley rats were divided into 3 equal groups; control group, premature ovarian insufficiency (POI) group, and POI + BM-MSCs. POI female rat model was established with cyclophosphamide. The result revealed that BM-MSCs and their conditioned media displayed a significant expression level of Neat-1, Hotair-1, mir-21-5p, mir-144-5p, and mir-664-5p. Moreover, BM-MSCs transplantation in POI rats improves; the ovarian and hypothalamic IGF-1 - kisspeptin, HPG axis, ovarian granulosa cell apoptosis, steroidogenesis, angiogenesis, energy balance, and oxidative stress. BM-MSCs expressed higher levels of antiapoptotic lncRNAs and microRNAs that mitigate ovarian insufficiency.
Collapse
Affiliation(s)
- Amany I Ahmed
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed F Dowidar
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Asmaa F Negm
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Hussein Abdellatif
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman
- Human Anatomy and Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Asma Alanazi
- Collage of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mohammed Alassiri
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- Department of Basic Medical Sciences, College of Science and Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences, Riyadh, Kingdom of Saudi Arabia
| | - Walaa Samy
- Medical biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Dina Mohamed Mekawy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University in Cairo, Badr City, 11829, Egypt
| | - Eman M A Abdelghany
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Nesma I El-Naseery
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed A Ibrahem
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Emad Ali Albadawi
- Department of Anatomy, College of Medicine, Taibah University, Medina, Saudi Arabia
| | - Wed Salah
- Department of Anatomy, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O.Box 71666, Riyadh, 11597, Saudi Arabia
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Emil Tîrziu
- Department of Animal Production and Veterinary Public Health, Faculty of Veterinary Medicine, University of Life Sciences, "King Mihai I" from Timisoara [ULST], Aradului St. 119, Timisoara, 300645, Romania
| | - Iulia Maria Bucur
- Department of Animal Production and Veterinary Public Health, Faculty of Veterinary Medicine, University of Life Sciences, "King Mihai I" from Timisoara [ULST], Aradului St. 119, Timisoara, 300645, Romania.
| | - Ahmed Hamed Arisha
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine, Badr University in Cairo, Badr City, 11829, Egypt.
- Department of Physiology and Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Tarek Khamis
- Department of Pharmacology and Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
18
|
Karunasagara S, Taghizadeh A, Kim SH, Kim SJ, Kim YJ, Taghizadeh M, Kim MY, Oh KY, Lee JH, Kim HS, Hyun J, Kim HW. Tissue Mechanics and Hedgehog Signaling Crosstalk as a Key Epithelial-Stromal Interplay in Cancer Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400063. [PMID: 38976559 PMCID: PMC11425211 DOI: 10.1002/advs.202400063] [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: 01/03/2024] [Revised: 05/30/2024] [Indexed: 07/10/2024]
Abstract
Epithelial-stromal interplay through chemomechanical cues from cells and matrix propels cancer progression. Elevated tissue stiffness in potentially malignant tissues suggests a link between matrix stiffness and enhanced tumor growth. In this study, employing chronic oral/esophageal injury and cancer models, it is demonstrated that epithelial-stromal interplay through matrix stiffness and Hedgehog (Hh) signaling is key in compounding cancer development. Epithelial cells actively interact with fibroblasts, exchanging mechanoresponsive signals during the precancerous stage. Specifically, epithelial cells release Sonic Hh, activating fibroblasts to produce matrix proteins and remodeling enzymes, resulting in tissue stiffening. Subsequently, basal epithelial cells adjacent to the stiffened tissue become proliferative and undergo epithelial-to-mesenchymal transition, acquiring migratory and invasive properties, thereby promoting invasive tumor growth. Notably, transcriptomic programs of oncogenic GLI2, mechano-activated by actin cytoskeletal tension, govern this process, elucidating the crucial role of non-canonical GLI2 activation in orchestrating the proliferation and mesenchymal transition of epithelial cells. Furthermore, pharmacological intervention targeting tissue stiffening proves highly effective in slowing cancer progression. These findings underscore the impact of epithelial-stromal interplay through chemo-mechanical (Hh-stiffness) signaling in cancer development, and suggest that targeting tissue stiffness holds promise as a strategy to disrupt chemo-mechanical feedback, enabling effective cancer treatment.
Collapse
Affiliation(s)
- Shanika Karunasagara
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Ali Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Sang-Hyun Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Chemistry, College of Science & Technology, Dankook University, Cheonan, 31116, Republic of Korea
| | - So Jung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Yong-Jae Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Mohsen Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Moon-Young Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Kyu-Young Oh
- Department of Oral Pathology, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hye Sung Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeongeun Hyun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science & BK21 Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea
| |
Collapse
|
19
|
Nguyen H, Hsu CC, Meeson A, Oldershaw R, Richardson G, Czosseck A, Lundy DJ. Differentiation, Metabolism, and Cardioprotective Secretory Functions of Human Cardiac Stromal Cells from Ischemic and Endocarditis Patients. Stem Cells Dev 2024; 33:484-495. [PMID: 38940748 DOI: 10.1089/scd.2024.0103] [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: 06/29/2024] Open
Abstract
This study investigates the characteristics of cardiac mesenchymal stem cell-like cells (CMSCLCs) isolated from the right atrial appendage of human donors with ischemia and a young patient with endocarditis (NE-CMSCLCs). Typical CMSCLCs from ischemic heart patients were derived from coronary artery bypass grafting procedures and compared against bone marrow mesenchymal stromal cells (BM-MSCs). NE-CMSCLCs had a normal immunophenotype, but exhibited enhanced osteogenic differentiation potential, rapid proliferation, reduced senescence, reduced glycolysis, and lower reactive oxygen species generation after oxidative stress compared with typical ischemic CMSCLCs. These differences suggest a unique functional status of NE-CMSCLCs, influenced by the donor health condition. Despite large variances in their paracrine secretome, NE-CMSCLCs retained therapeutic potential, as indicated by their ability to protect hypoxia/reoxygenation-injured human cardiomyocytes, albeit less effectively than typical CMSCLCs. This research describes a unique cell phenotype and underscores the importance of donor health status in the therapeutic efficacy of autologous cardiac cell therapy.
Collapse
Affiliation(s)
- Helen Nguyen
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chuan-Chih Hsu
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Cardiovascular Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Annette Meeson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rachel Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Gavin Richardson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andreas Czosseck
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
| | - David J Lundy
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei, Taiwan
- International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- Center for Cell Therapy, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
20
|
Moro-López M, Farré R, Otero J, Sunyer R. Trusting the forces of our cell lines. Cells Dev 2024; 179:203931. [PMID: 38852676 DOI: 10.1016/j.cdev.2024.203931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/03/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Cells isolated from their native tissues and cultured in vitro face different selection pressures than those cultured in vivo. These pressures induce a profound transformation that reshapes the cell, alters its genome, and transforms the way it senses and generates forces. In this perspective, we focus on the evidence that cells cultured on conventional polystyrene substrates display a fundamentally different mechanobiology than their in vivo counterparts. We explore the role of adhesion reinforcement in this transformation and to what extent it is reversible. We argue that this mechanoadaptation is often understood as a mechanical memory. We propose some strategies to mitigate the effects of on-plastic culture on mechanobiology, such as organoid-inspired protocols or mechanical priming. While isolating cells from their native tissues and culturing them on artificial substrates has revolutionized biomedical research, it has also transformed cellular forces. Only by understanding and controlling them, we can improve their truthfulness and validity.
Collapse
Affiliation(s)
- Marina Moro-López
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-RES), Barcelona, Spain; Institut Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain
| | - Jorge Otero
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-RES), Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Raimon Sunyer
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería (CIBER-BBN), Barcelona, Spain.
| |
Collapse
|
21
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
22
|
Younesi FS, Miller AE, Barker TH, Rossi FMV, Hinz B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol 2024; 25:617-638. [PMID: 38589640 DOI: 10.1038/s41580-024-00716-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
The term 'fibroblast' often serves as a catch-all for a diverse array of mesenchymal cells, including perivascular cells, stromal progenitor cells and bona fide fibroblasts. Although phenotypically similar, these subpopulations are functionally distinct, maintaining tissue integrity and serving as local progenitor reservoirs. In response to tissue injury, these cells undergo a dynamic fibroblast-myofibroblast transition, marked by extracellular matrix secretion and contraction of actomyosin-based stress fibres. Importantly, whereas transient activation into myofibroblasts aids in tissue repair, persistent activation triggers pathological fibrosis. In this Review, we discuss the roles of mechanical cues, such as tissue stiffness and strain, alongside cell signalling pathways and extracellular matrix ligands in modulating myofibroblast activation and survival. We also highlight the role of epigenetic modifications and myofibroblast memory in physiological and pathological processes. Finally, we discuss potential strategies for therapeutically interfering with these factors and the associated signal transduction pathways to improve the outcome of dysregulated healing.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Andrew E Miller
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, USA
| | - Fabio M V Rossi
- School of Biomedical Engineering and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada.
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
23
|
Poomani MS, Regurajan R, Perumal R, Ramachandran A, Mariappan I, Muthan K, Subramanian V. Differentiation of placenta-derived MSCs cultured in human platelet lysate: a xenofree supplement. 3 Biotech 2024; 14:116. [PMID: 38524240 PMCID: PMC10959853 DOI: 10.1007/s13205-024-03966-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 02/22/2024] [Indexed: 03/26/2024] Open
Abstract
In the last few decades, mesenchymal stem cells (MSCs)-based regenerative therapies in clinical applications have gradually become a hot topic due to their long-term self-renewal and multilineage differentiation ability. In this scenario, placenta (p) has been considered as a good source of MSCs. As a tissue of fetal origin with abundant number of stem cells compared to other sources, their non-invasive acquisition, strong immunosuppression, and lack of ethical concerns make placenta an indispensable source of MSC in stem cell research and therapy. The mesenchymal stem cells were derived from human term placenta (p-MSCs) in xenofree condition using platelet lysate (PL) as a suitable alternative to fetal bovine serum (FBS). Upon isolation, p-MSCs showed plastic adherence with spindle-shaped, fibroblast-like morphology under microscope. p-MSCs flourished well in PL-containing media. Immunophenotyping showed classical MSC markers (> 90%) and lack expression of hematopoietic and HLA-DR (< 1%). Surprisingly, differentiation study showed differentiation of p-MSCs to mature adipocytes in both induced cells and control (spontaneous differentiation), as observed via oil red staining. This is in line with gene expression data where both control and induced cells were positive for visfatin and leptin. Thus, we propose that p-MSCs can be used for clinical applications in the treatment of various chronic and degenerative diseases.
Collapse
Affiliation(s)
- Merlin Sobia Poomani
- Genetic Engineering and Regenerative Biology Lab, Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | - Rathika Regurajan
- Centre for Marine Science and Technology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | | | | | - Iyyadurai Mariappan
- Genetic Engineering and Regenerative Biology Lab, Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | - Krishnaveni Muthan
- Department of Animal Science, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| | - Venkatesh Subramanian
- Genetic Engineering and Regenerative Biology Lab, Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012 India
| |
Collapse
|
24
|
Sirisereephap K, Tamura H, Lim JH, Surboyo MDC, Isono T, Hiyoshi T, Rosenkranz AL, Sato-Yamada Y, Domon H, Ikeda A, Hirose T, Sunazuka T, Yoshiba N, Okada H, Terao Y, Maeda T, Tabeta K, Chavakis T, Hajishengallis G, Maekawa T. A novel macrolide-Del-1 axis to regenerate bone in old age. iScience 2024; 27:108798. [PMID: 38261928 PMCID: PMC10797555 DOI: 10.1016/j.isci.2024.108798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/24/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
Aging is associated with increased susceptibility to chronic inflammatory bone loss disorders, such as periodontitis, in large part due to the impaired regenerative potential of aging tissues. DEL-1 exerts osteogenic activity and promotes bone regeneration. However, DEL-1 expression declines with age. Here we show that systemically administered macrolide antibiotics and a non-antibiotic erythromycin derivative, EM-523, restore DEL-1 expression in 18-month-old ("aged") mice while promoting regeneration of bone lost due to naturally occurring age-related periodontitis. These compounds failed to induce bone regeneration in age-matched DEL-1-deficient mice. Consequently, these drugs promoted DEL-1-dependent functions, including alkaline phosphatase activity and osteogenic gene expression in the periodontal tissue while inhibiting osteoclastogenesis, leading to net bone growth. Macrolide-treated aged mice exhibited increased skeletal bone mass, suggesting that this treatment may be pertinent to systemic bone loss disorders. In conclusion, we identified a macrolide-DEL-1 axis that can regenerate bone lost due to aging-related disease.
Collapse
Affiliation(s)
- Kridtapat Sirisereephap
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Hikaru Tamura
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meircurius Dwi Condro Surboyo
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
- Faculty of Dentistry, Universitas Airlangga, Surabaya 60132, Indonesia
| | - Toshihito Isono
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Takumi Hiyoshi
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Andrea L. Rosenkranz
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Yurie Sato-Yamada
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Akari Ikeda
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoyasu Hirose
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Toshiaki Sunazuka
- Ōmura Satoshi Memorial Institute, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
- Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Nagako Yoshiba
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Hiroyuki Okada
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Koichi Tabeta
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tomoki Maekawa
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan
| |
Collapse
|
25
|
Strippoli R, Niayesh-Mehr R, Adelipour M, Khosravi A, Cordani M, Zarrabi A, Allameh A. Contribution of Autophagy to Epithelial Mesenchymal Transition Induction during Cancer Progression. Cancers (Basel) 2024; 16:807. [PMID: 38398197 PMCID: PMC10886827 DOI: 10.3390/cancers16040807] [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: 12/15/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Epithelial Mesenchymal Transition (EMT) is a dedifferentiation process implicated in many physio-pathological conditions including tumor transformation. EMT is regulated by several extracellular mediators and under certain conditions it can be reversible. Autophagy is a conserved catabolic process in which intracellular components such as protein/DNA aggregates and abnormal organelles are degraded in specific lysosomes. In cancer, autophagy plays a controversial role, acting in different conditions as both a tumor suppressor and a tumor-promoting mechanism. Experimental evidence shows that deep interrelations exist between EMT and autophagy-related pathways. Although this interplay has already been analyzed in previous studies, understanding mechanisms and the translational implications of autophagy/EMT need further study. The role of autophagy in EMT is not limited to morphological changes, but activation of autophagy could be important to DNA repair/damage system, cell adhesion molecules, and cell proliferation and differentiation processes. Based on this, both autophagy and EMT and related pathways are now considered as targets for cancer therapy. In this review article, the contribution of autophagy to EMT and progression of cancer is discussed. This article also describes the multiple connections between EMT and autophagy and their implication in cancer treatment.
Collapse
Affiliation(s)
- Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy;
- National Institute for Infectious Diseases “Lazzaro Spallanzani”, I.R.C.C.S., 00149 Rome, Italy
| | - Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| | - Maryam Adelipour
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran;
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye;
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain;
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye;
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Science, Tarbiat Modares University, Tehran P.O. Box 14115-331, Iran;
| |
Collapse
|
26
|
Zhu L, Gou W, Ou L, Liu B, Liu M, Feng H. Role and new insights of microfibrillar-associated protein 4 in fibrotic diseases. APMIS 2024; 132:55-67. [PMID: 37957836 DOI: 10.1111/apm.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023]
Abstract
Fibrosis is one of the most worrisome complications of chronic inflammatory diseases, leading to tissue damage, organ failure, and ultimately, death. The most notable pathological characteristic of fibrosis is the excessive accumulation of extracellular matrix (ECM) components such as collagen and fibronectin adjacent to foci of inflammation or damage. The human microfibrillar-associated protein 4 (MFAP4), an important member of the superfamily of fibrinogen-related proteins, is considered to have an extremely important role in ECM transformation of fibrogenesis. This review summarizes the structure, characteristics, and physiological functions of MFAP4 and the importance of MFAP4 in various fibrotic diseases. Meanwhile, we elaborated the underlying actions and mechanisms of MFAP4 in the development of fibrosis, suggesting that a better understand of MFAP4 broadens novel perspective for early screening, diagnosis, prognostic risk assessment, and treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Long Zhu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Wenqun Gou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
- Changsha Stomatological Hospital, Changsha, China
| | - Lijia Ou
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Binjie Liu
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Manyi Liu
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| | - Hui Feng
- Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Changsha, China
- Xiangya Stomatological Hospital, Changsha, China
- Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
27
|
Fung M, Armstrong JJ, Zhang R, Vinokurtseva A, Liu H, Hutnik C. Development and Verification of a Novel Three-Dimensional Aqueous Outflow Model for High-Throughput Drug Screening. Bioengineering (Basel) 2024; 11:142. [PMID: 38391628 PMCID: PMC10885921 DOI: 10.3390/bioengineering11020142] [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: 12/30/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Distal outflow bleb-forming procedures in ophthalmic surgery expose subconjunctival tissue to inflammatory cytokines present in the aqueous humor, resulting in impaired outflow and, consequently, increased intraocular pressure. Clinically, this manifests as an increased risk of surgical failure often necessitating revision. This study (1) introduces a novel high-throughput screening platform for testing potential anti-fibrotic compounds and (2) assesses the clinical viability of modulating the transforming growth factor beta-SMAD2/3 pathway as a key contributor to post-operative outflow reduction, using the signal transduction inhibitor verteporfin. Human Tenon's capsule fibroblasts (HTCFs) were cultured within a 3D collagen matrix in a microfluidic system modelling aqueous humor drainage. The perfusate was augmented with transforming growth factor beta 1 (TGFβ1), and afferent pressure to the tissue-mimetic was continuously monitored to detect treatment-related pressure elevations. Co-treatment with verteporfin was employed to evaluate its capacity to counteract TGFβ1 induced pressure changes. Immunofluorescent studies were conducted on the tissue-mimetic to corroborate the pressure data with cellular changes. Introduction of TGFβ1 induced treatment-related afferent pressure increase in the tissue-mimetic. HTCFs treated with TGFβ1 displayed visibly enlarged cytoskeletons and stress fiber formation, consistent with myofibroblast transformation. Importantly, verteporfin effectively mitigated these changes, reducing both afferent pressure increases and cytoskeletal alterations. In summary, this study models the pathological filtration bleb response to TGFβ1, while demonstrating verteporfin's effectiveness in ameliorating both functional and cellular changes caused by TGFβ1. These demonstrate modulation of the aforementioned pathway as a potential avenue for addressing post-operative changes and reductions in filtration bleb outflow capacity. Furthermore, the establishment of a high-throughput screening platform offers a valuable pre-animal testing tool for investigating potential compounds to facilitate surgical wound healing.
Collapse
Affiliation(s)
- Matthew Fung
- Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - James J Armstrong
- Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Ophthalmology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Richard Zhang
- Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Anastasiya Vinokurtseva
- Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Ophthalmology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Hong Liu
- Department of Ophthalmology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Cindy Hutnik
- Department of Ophthalmology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Ophthalmology, Ivey Eye Institute, St. Joseph's Health Center, London, ON N6A 4V2, Canada
| |
Collapse
|
28
|
Yang Y, Alves T, Miao M, Wu Y, Li G, Lou J, Hasturk H, Van Dyke T, Kantarci A, Wu D. Single-Cell Transcriptomic Analysis of Dental Pulp and Periodontal Ligament Stem Cells. J Dent Res 2024; 103:71-80. [PMID: 37982164 PMCID: PMC10850875 DOI: 10.1177/00220345231205283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
The regeneration of periodontal, periapical, and pulpal tissues is a complex process requiring the direct involvement of cells derived from pluripotent stem cells in the periodontal ligament and dental pulp. Dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) are spatially distinct with the potential to differentiate into similar functional and phenotypic cells. We aimed to identify the cell heterogeneity of DPSCs and PDLSCs and explore the differentiation potentials of their specialized organ-specific functions using single-cell transcriptomic analysis. Our results revealed 7 distinct clusters, with cluster 3 showing the highest potential for differentiation. Clusters 0 to 2 displayed features similar to fibroblasts. The trajectory route of the cell state transition from cluster 3 to clusters 0, 1, and 2 indicated the distinct nature of cell differentiation. PDLSCs had a higher proportion of cells (78.6%) at the G1 phase, while DPSCs had a higher proportion of cells at the S and G2/M phases (36.1%), mirroring the lower cell proliferation capacity of PDLSCs than DPSCs. Our study suggested the heterogeneity of stemness across PDLSCs and DPSCs, the similarities of these 2 stem cell compartments to be potentially integrated for regenerative strategies, and the distinct features between them potentially particularized for organ-specific functions of the dental pulp and periodontal ligament for a targeted regenerative dental tissue repair and other regeneration therapies.
Collapse
Affiliation(s)
- Y. Yang
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - T. Alves
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M.Z. Miao
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Y.C. Wu
- The Forsyth Institute, Cambridge, MA, USA
| | - G. Li
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| | - J. Lou
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - H. Hasturk
- The Forsyth Institute, Cambridge, MA, USA
| | | | | | - D. Wu
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
29
|
Saxena N, Chakraborty S, Dutta S, Bhardwaj G, Karnik N, Shetty O, Jadhav S, Zafar H, Sen S. Stiffness-dependent MSC homing and differentiation into CAFs - implications for breast cancer invasion. J Cell Sci 2024; 137:jcs261145. [PMID: 38108421 DOI: 10.1242/jcs.261145] [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: 03/09/2023] [Accepted: 12/03/2023] [Indexed: 12/19/2023] Open
Abstract
Cellular heterogeneity and extracellular matrix (ECM) stiffening have been shown to be drivers of breast cancer invasiveness. Here, we examine how stiffness-dependent crosstalk between cancer cells and mesenchymal stem cells (MSCs) within an evolving tumor microenvironment regulates cancer invasion. By analyzing previously published single-cell RNA sequencing datasets, we establish the existence of a subpopulation of cells in primary tumors, secondary sites and circulatory tumor cell clusters of highly aggressive triple-negative breast cancer (TNBC) that co-express MSC and cancer-associated fibroblast (CAF) markers. By using hydrogels with stiffnesses of 0.5, 2 and 5 kPa to mimic different stages of ECM stiffening, we show that conditioned medium from MDA-MB-231 TNBC cells cultured on 2 kPa gels, which mimic the pre-metastatic stroma, drives efficient MSC chemotaxis and induces stable differentiation of MSC-derived CAFs in a TGFβ (TGFB1)- and contractility-dependent manner. In addition to enhancing cancer cell proliferation, MSC-derived CAFs on 2 kPa gels maximally boost local invasion and confer resistance to flow-induced shear stresses. Collectively, our results suggest that homing of MSCs at the pre-metastatic stage and their differentiation into CAFs actively drives breast cancer invasion and metastasis in TNBC.
Collapse
Affiliation(s)
- Neha Saxena
- Department of Chemical Engineering, IIT Bombay,Mumbai 400076, India
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Soura Chakraborty
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
| | - Sarbajeet Dutta
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Garvit Bhardwaj
- Department of Electrical Engineering, IIT Kanpur, Kanpur 208016, India
| | - Nupur Karnik
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Omshree Shetty
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Sameer Jadhav
- Department of Chemical Engineering, IIT Bombay,Mumbai 400076, India
| | - Hamim Zafar
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Department of Computer Science and Engineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine , IIT Kanpur, Kanpur 208016, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| |
Collapse
|
30
|
Marinkovic M, Tran ON, Wang H, Abdul-Azees P, Dean DD, Chen XD, Yeh CK. Extracellular matrix turnover in salivary gland disorders and regenerative therapies: Obstacles and opportunities. J Oral Biol Craniofac Res 2023; 13:693-703. [PMID: 37719063 PMCID: PMC10502366 DOI: 10.1016/j.jobcr.2023.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/28/2023] [Indexed: 09/19/2023] Open
Abstract
Salivary gland (SG) extracellular matrix (ECM) has a major influence on tissue development, homeostasis, and tissue regeneration after injury. During aging, disease, and physical insult, normal remodeling of the SG microenvironment (i.e. ECM) becomes dysregulated, leading to alterations in matrix composition which disrupt tissue architecture/structure, alter cell activity, and negatively impact gland function. Matrix metalloproteinases (MMPs) are a large and diverse family of metalloendopeptidases which play a major role in matrix degradation and are intimately involved in regulating development and cell function; dysregulation of these enzymes leads to the production of a fibrotic matrix. In the SG this altered fibrotic ECM (or cell microenvironment) negatively impacts normal cell function and the effectiveness of gene and stem cell therapies which serve as a foundation for many SG regenerative therapies. For this reason, prospective regenerative strategies should prioritize the maintenance and/or restoration of a healthy SG ECM. Mesenchymal stem cells (MSCs) have great potential for mitigating damage to the SG microenvironment by ameliorating inflammation, reducing fibrosis, and repairing the damaged milieu of extracellular regulatory cues, including the matrix. This review addresses our current understanding of the impact of aging and disease on the SG microenvironment and suggests critical deficiencies and opportunities in ECM-targeted therapeutic interventions.
Collapse
Affiliation(s)
- Milos Marinkovic
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, 78229-4404, USA
| | - Olivia N. Tran
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Hanzhou Wang
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Parveez Abdul-Azees
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, 78229-4404, USA
| | - David D. Dean
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Xiao-Dong Chen
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, 78229-4404, USA
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, 78229-4404, USA
| |
Collapse
|
31
|
Ezzo M, Hinz B. Novel approaches to target fibroblast mechanotransduction in fibroproliferative diseases. Pharmacol Ther 2023; 250:108528. [PMID: 37708995 DOI: 10.1016/j.pharmthera.2023.108528] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/09/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The ability of cells to sense and respond to changes in mechanical environment is vital in conditions of organ injury when the architecture of normal tissues is disturbed or lost. Among the various cellular players that respond to injury, fibroblasts take center stage in re-establishing tissue integrity by secreting and organizing extracellular matrix into stabilizing scar tissue. Activation, activity, survival, and death of scar-forming fibroblasts are tightly controlled by mechanical environment and proper mechanotransduction ensures that fibroblast activities cease after completion of the tissue repair process. Conversely, dysregulated mechanotransduction often results in fibroblast over-activation or persistence beyond the state of normal repair. The resulting pathological accumulation of extracellular matrix is called fibrosis, a condition that has been associated with over 40% of all deaths in the industrialized countries. Consequently, elements in fibroblast mechanotransduction are scrutinized for their suitability as anti-fibrotic therapeutic targets. We review the current knowledge on mechanically relevant factors in the fibroblast extracellular environment, cell-matrix and cell-cell adhesion structures, stretch-activated membrane channels, stress-regulated cytoskeletal structures, and co-transcription factors. We critically discuss the targetability of these elements in therapeutic approaches and their progress in pre-clinical and/or clinical trials to treat organ fibrosis.
Collapse
Affiliation(s)
- Maya Ezzo
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Boris Hinz
- Keenan Research Institute for Biomedical Science of the St. Michael's Hospital, and Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
32
|
Huang F, Li Y, Guan L, Hu Y, Zeng M. MiR-30a inhibits silica dust-induced epithelial-mesenchymal transition by targeting Snail. Toxicol In Vitro 2023; 92:105657. [PMID: 37543170 DOI: 10.1016/j.tiv.2023.105657] [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: 03/25/2023] [Revised: 06/21/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023]
Abstract
The mechanism of action of MicroRNA-30a(miR-30a) and Snail, a transcription factor, in silica(SiO2) dust-induced pulmonary EMT and secondary pulmonary fibrosis remains elusive. In this study, the cellular EMT model induced by the stimulation of A549 cells with SiO2 was established. A549 cells were transfected with miR-30a mimic and miR-30a inhibitor and the SNAIL gene was silenced to examine the mechanism of miR-30a targeting Snail to regulate silica dust-induced EMT. The results showed that 50 μg/mL SiO2 stained A549 cells for 24 h could induce EMT in A549 cells. Exposure of A549 cells to SiO2 dust decreased miR-30a expression, as well as mRNA and protein expression levels of E-cad. Conversely, SiO2 exposure increased mRNA and protein expression levels of α-SMA, vimentin, and Snail. The miR-30a mimic upregulated mRNA and protein expression levels of E-cadherin in SiO2-induced A549 cells, while downregulating mRNA and protein expression levels of α-SMA, vimentin and Snail. MiR-30a inhibitors have the opposite effect. Silencing the SNAIL gene, followed by SiO2 dust-induced stimulation of A549 cells, could enhance mRNA and protein expression levels of E-cad, whereas those of α-SMA and vimentin were reduced. Altogether, we found that miR-30a directly targeted Snail and inhibited its expression, thereby delaying silica induced pulmonary EMT.
Collapse
Affiliation(s)
- Fangcai Huang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, China
| | - Yupei Li
- The First Affiliated Hospital of Xi'An Jiaotong University, China
| | - Lan Guan
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, China
| | - Yuming Hu
- Hunan Provincial Center For Disease Control And Prevention, China.
| | - Ming Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
33
|
Ding Z, Shi R, Hu W, Tian L, Sun R, Wu Y, Zhang X. Cancer-associated fibroblasts in hematologic malignancies: elucidating roles and spotlighting therapeutic targets. Front Oncol 2023; 13:1193978. [PMID: 37746306 PMCID: PMC10511871 DOI: 10.3389/fonc.2023.1193978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Hematologic malignancies comprise a diverse range of blood, bone marrow, and organ-related disorders that present significant challenges due to drug resistance, relapse, and treatment failure. Cancer-associated fibroblasts (CAFs) represent a critical component of the tumor microenvironment (TME) and have recently emerged as potential therapeutic targets. In this comprehensive review, we summarize the latest findings on the roles of CAFs in various hematologic malignancies, including acute leukemia, multiple myeloma, chronic lymphocytic leukemia, myeloproliferative neoplasms, and lymphoma. We also explore their involvement in tumor progression, drug resistance, and the various signaling pathways implicated in their activation and function. While the underlying mechanisms and the existence of multiple CAF subtypes pose challenges, targeting CAFs and their associated pathways offers a promising avenue for the development of innovative treatments to improve patient outcomes in hematologic malignancies.
Collapse
Affiliation(s)
- Ziyang Ding
- The Second Clinical School of Nanjing Medical University, Nanjing, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weikang Hu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Tian
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Sun
- Department of Radiation Oncology, Jinling Hospital, Nanjing, China
| | - Yang Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoyan Zhang
- Department of Hematology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
McElhinney K, Irnaten M, O’Brien C. p53 and Myofibroblast Apoptosis in Organ Fibrosis. Int J Mol Sci 2023; 24:ijms24076737. [PMID: 37047710 PMCID: PMC10095465 DOI: 10.3390/ijms24076737] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
Organ fibrosis represents a dysregulated, maladaptive wound repair response that results in progressive disruption of normal tissue architecture leading to detrimental deterioration in physiological function, and significant morbidity/mortality. Fibrosis is thought to contribute to nearly 50% of all deaths in the Western world with current treatment modalities effective in slowing disease progression but not effective in restoring organ function or reversing fibrotic changes. When physiological wound repair is complete, myofibroblasts are programmed to undergo cell death and self-clearance, however, in fibrosis there is a characteristic absence of myofibroblast apoptosis. It has been shown that in fibrosis, myofibroblasts adopt an apoptotic-resistant, highly proliferative phenotype leading to persistent myofibroblast activation and perpetuation of the fibrotic disease process. Recently, this pathological adaptation has been linked to dysregulated expression of tumour suppressor gene p53. In this review, we discuss p53 dysregulation and apoptotic failure in myofibroblasts and demonstrate its consistent link to fibrotic disease development in all types of organ fibrosis. An enhanced understanding of the role of p53 dysregulation and myofibroblast apoptosis may aid in future novel therapeutic and/or diagnostic strategies in organ fibrosis.
Collapse
Affiliation(s)
- Kealan McElhinney
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Mustapha Irnaten
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Colm O’Brien
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| |
Collapse
|
35
|
Samsonraj RM, Law SF, Chandra A, Pignolo RJ. An unbiased proteomics approach to identify the senescence-associated secretory phenotype of human bone marrow-derived mesenchymal stem cells. Bone Rep 2023; 18:101674. [PMID: 36994454 PMCID: PMC10041468 DOI: 10.1016/j.bonr.2023.101674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/17/2023] [Indexed: 03/28/2023] Open
Abstract
Mesenchymal stem cells (MSCs) derived from bone marrow can support skeletal tissue repair and regeneration owing to their self-renewing capacity, differentiation ability, and trophic functions. Bone marrow-derived MSCs undergo dramatic changes with aging, including the senescence-associated secretory phenotype (SASP) which may largely contribute to age-related changes in bone tissue leading to osteoporosis. A mass spectrometry-based proteomics approach was used to investigate the MSC SASP. Replicative senescence was achieved by exhaustive in vitro sub-cultivation and confirmed by standard proliferation criteria. Conditioned media from non-senescent and senescent MSCs underwent mass spectrometry. Proteomics and bioinformatics analyses enabled the identification of 95 proteins expressed uniquely in senescent MSCs. Protein ontology analysis revealed the enrichment of proteins linked to the extracellular matrix, exosomes, cell adhesion, and calcium ion binding. The proteomic analysis was independently validated by taking ten identified proteins with relevance to bone aging and confirming their increased abundance in conditioned media from replicatively senescent versus non-senescent MSCs (ACTα2, LTF, SOD1, IL-6, LTBP2, PXDN, SERPINE 1, COL1α1, THBS1, OPG). These target proteins were used to further investigate changes in the MSC SASP profile in response to other inducers of senescence, ionizing radiation (IR) and H2O2. Similar secreted protein expression profiles with replicatively senescent cells were seen with H2O2 treatment except for LTF and PXDN, which were increased by IR treatment. With both IR and H2O2 treatment there was a decrease in THBS1. In vivo investigation of these secreted proteins with aging was shown by significant changes in the abundance of OPG, COL1α1, IL-6, ACTα2, SERPINE 1, and THBS1 in the plasma of aged rats. This unbiased, comprehensive analysis of the changes in the MSC secretome with senescence defines the unique protein signature of the SASP in these cells and provides a better understanding of the aging bone microenvironment.
Identified the senescence-associated secretory phenotype of mesenchymal stem cells. Investigated protein expression under different senescence induction conditions. Showed significant changes in in vivo abundance of target proteins in aging rats.
Collapse
Affiliation(s)
| | - Susan F. Law
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Abhishek Chandra
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Robert J. Pignolo
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Corresponding author at: Robert and Arlene Kogod Professor of Geriatric Medicine, Department of Medicine, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
36
|
Esophageal wound healing by aligned smooth muscle cell-laden nanofibrous patch. Mater Today Bio 2023; 19:100564. [PMID: 36747583 PMCID: PMC9898453 DOI: 10.1016/j.mtbio.2023.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
The esophagus exhibits peristalsis via contraction of circularly and longitudinally aligned smooth muscles, and esophageal replacement is required if there is a critical-sized wound. In this study, we proposed to reconstruct esophageal tissues using cell electrospinning (CE), an advanced technique for encapsulating living cells into fibers that allows control of the direction of fiber deposition. After treatment with transforming growth factor-β, mesenchymal stem cell-derived smooth muscle cells (SMCs) were utilized for cell electrospinning or three-dimensional bioprinting to compare the effects of aligned micropatterns on cell morphology. CE resulted in SMCs with uniaxially arranged and elongated cell morphology with upregulated expression levels of SMC-specific markers, including connexin 43, smooth muscle protein 22 alpha (SM22α), desmin, and smoothelin. When SMC-laden nanofibrous patches were transplanted into a rat esophageal defect model, the SMC patch promoted regeneration of esophageal wounds with an increased number of newly formed blood vessels and enhanced the SMC-specific markers of SM22α and vimentin. Taken together, CE with its advantages, such as guidance of highly elongated, aligned cell morphology and accelerated SMC differentiation, can be an efficient strategy to reconstruct smooth muscle tissues and treat esophageal perforation.
Collapse
|
37
|
Schuster R, Younesi F, Ezzo M, Hinz B. The Role of Myofibroblasts in Physiological and Pathological Tissue Repair. Cold Spring Harb Perspect Biol 2023; 15:a041231. [PMID: 36123034 PMCID: PMC9808581 DOI: 10.1101/cshperspect.a041231] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Myofibroblasts are the construction workers of wound healing and repair damaged tissues by producing and organizing collagen/extracellular matrix (ECM) into scar tissue. Scar tissue effectively and quickly restores the mechanical integrity of lost tissue architecture but comes at the price of lost tissue functionality. Fibrotic diseases caused by excessive or persistent myofibroblast activity can lead to organ failure. This review defines myofibroblast terminology, phenotypic characteristics, and functions. We will focus on the central role of the cell, ECM, and tissue mechanics in regulating tissue repair by controlling myofibroblast action. Additionally, we will discuss how therapies based on mechanical intervention potentially ameliorate wound healing outcomes. Although myofibroblast physiology and pathology affect all organs, we will emphasize cutaneous wound healing and hypertrophic scarring as paradigms for normal tissue repair versus fibrosis. A central message of this review is that myofibroblasts can be activated from multiple cell sources, varying with local environment and type of injury, to either restore tissue integrity and organ function or create an inappropriate mechanical environment.
Collapse
Affiliation(s)
- Ronen Schuster
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
| | - Fereshteh Younesi
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Maya Ezzo
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
38
|
Li J, Li K, Tian Y, Zhao P, Liu X, Li M, Bai Y. Effective-compounds of Jinshui Huanxian formula ameliorates fibroblast activation in pulmonary fibrosis by inhibiting the activation of mTOR signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154604. [PMID: 36610143 DOI: 10.1016/j.phymed.2022.154604] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/26/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Jinshui Huanxian formula (JHF) ameliorates idiopathic pulmonary fibrosis patients. Active compounds, including icariin, isoliquiritigenin, nobiletin, peimine, and paeoniflorin, deriving from JHF were combined as effective-component compatibility ECC of JHF II (ECC-JHF II), which is an effective therapeutic strategy for pulmonary fibrosis (PF) induced by bleomycin (BLM) in rats. PURPOSE This study aimed to explore the underlying mechanism of ECC-JHF II on pulmonary fibrosis. METHODS A model of PF in rats was established through intratracheal instillation of BLM. Pulmonary function, pathological changes, and collagen deposition were examined. The gene and protein expressions in fibroblast activation were detected by quantitative real-time PCR and western blotting respectively. RESULTS ECC-JHF II significantly improved BLM-induced PF in rats, manifested as decreased collagen deposition, reduced pathological damage and improved pulmonary function. Furthermore, ECC-JHF II inhibited fibroblast activation by reducing the expression of α-smooth muscle actin (α-SMA) and fibronectin. We analyzed the targets of ECC-JHF II and differentially expressed genes (DEGs) of fibroblast activation induced by transforming growth factor-β1 (TGF-β1) and found that ECC-JHF II might regulate fibroblast activation by EGFR, PI3K-Akt or mTOR signaling pathway. In vitro experiments, we also found that ECC-JHF II suppressed the mTOR pathway, such as downregulating the phosphorylation levels of p70S6K in fibroblast activation induced by TGF-β1. After activating mTOR signaling, the inhibition of ECC-JHF II on fibroblast activation was blocked. These results suggested that ECC-JHF II potently ameliorated pulmonary fibrosis in rats and effectively suppressed fibroblast activation by interfering with mTOR signaling. CONCLUSION We combined transcriptomics with the network analysis to predict the mechanism underlying ECC-JHF II suppression of fibroblast activation. In summary, ECC-JHF II improved BLM-induced pulmonary fibrosis, which might be associated with the suppression of fibroblast activation by inhibiting the mTOR signaling.
Collapse
Affiliation(s)
- Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan 450046, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Kangchen Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan 450046, China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Minyan Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan 450046, China
| | - Yunping Bai
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450000, China
| |
Collapse
|
39
|
Yu J, Li P, Duan Z, Liu X. Effect of Qiling Jiaogulan Powder on Pulmonary Fibrosis and Pulmonary Arteriole Remodeling in Low-Temperature-Exposed Broilers. Animals (Basel) 2022; 13:ani13010005. [PMID: 36611616 PMCID: PMC9817788 DOI: 10.3390/ani13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Chinese herbal medicine plays an important role in regulating the nutritional metabolism of poultry and maintaining or improving normal physiological functions and animal health. The present study investigated the effects of dietary supplementation with Qiling Jiaogulan Powder (QLJP) on pulmonary fibrosis and pulmonary arteriole remodeling in low temperature-exposed broilers. Seven-day-old Ross 308 broilers (n = 240) were reared adaptively to 14 days of age. The broilers were randomly divided into six groups: A control group (basal diet and normal feeding temperature); model group (basal diet); low-, medium- and high-dose QLJP groups (basal diet supplemented with 1 g/kg, 2 g/kg, 4 g/kg QLJP); and L-Arg group (basal diet supplemented with 10 g/kg L-arginine). Additionally, all the broilers, except the broilers in the control group, from the age of 14 days old, had a house temperature continuously lowered by 2 °C each day until it reached 12 °C at 21 days of age, and the low temperature was maintained until the end of the experiment. There were four replicates per group and 10 birds per replicate. The results showed that the structure of the lung tissue was clearer and basically intact in the broilers in the QLJP groups, with a small number of collagen fibers formed, and the content of hydroxyproline (HYP) was significantly reduced. QLJP improved pulmonary arteriole lesions, such as tunica media thickening, intimal hyperplasia, arterial wall hypertrophy, and lumen narrowing. QLJP reduced the relative media thickness (%) and relative medial area (%) of the pulmonary arteriole, and significantly decreased the expression level of the alpha-smooth muscle actin (α-SMA) protein in pulmonary arteriole, which alleviated pulmonary arteriole remodeling. The quantitative real-time PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA) results showed that QLJP treatment significantly reduced the gene and protein levels of transforming growth factor-beta l (TGF-β1) and Smad2 in the lung and downregulated the gene and protein levels of collagen type I alpha 1 (COL1A1) and matrix metalloproteinase 2 (MMP2). In conclusion, the results of our study suggested that dietary supplementation with QLJP improved pulmonary fibrosis and pulmonary arteriole remodeling by inhibiting the expression of genes related to the TGF-β1/Smad2 signaling pathway and inhibited the occurrence and development of pulmonary arterial hypertension in low-temperature-exposed broilers.
Collapse
Affiliation(s)
- Juan Yu
- School of Life Sciences and Basic Medicine, Xinxiang University, Xinxiang 453003, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030800, China
| | - Peng Li
- School of Life Sciences and Basic Medicine, Xinxiang University, Xinxiang 453003, China
| | - Zhibian Duan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030800, China
| | - Xingyou Liu
- School of Life Sciences and Basic Medicine, Xinxiang University, Xinxiang 453003, China
- Correspondence:
| |
Collapse
|
40
|
Hartanto MM, Prajoko YW, Putra A, Amalina ND. The Combination of Mesenchymal Stem Cells and Bovine Colostrum in Reducing α-SMA Expression and NLR Levels in Wistar Rats After 50% Fibrotic Liver Resection. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.10557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Liver fibrogenesis will produce α-smooth muscle actin (α-SMA) expression and a continuous inflammatory process, seen through the neutrophil lymphocyte ratio (NLR). The combination of mesenchymal stem cells and bovine colostrum is a novel strategy for repairing hepatic fibrosis tissue. To assess the combination of mesenchymal stem cells and bovine colostrum to reduce α-SMA expression and NLR levels in Wistar rats after 50% fibrotic liver resection.
Methods: Thirty-six Wistar male rats were randomly divided into 6 groups (sham, control, colostrum, MSCs, and colostrum and MSCs combination). Rats were injected with CCl4 for 8 weeks to induce liver fibrosis then underwent liver resection. NLR levels was determined using Hematology Analyzer, α-SMA expression of myofibroblast was analyzed by immunofluorescence staining.
Results: A significant reduction in NLR levels on day 3 in the treatment group I (1.10), treatment II (0.83), treatment III (0.93) compared to the control group. A significant reduction in NLR levels on day 10 in the treatment group I (0.76), treatment II (0.64), treatment III (0.54) compared to the control group. A significant decrease in α-SMA in treatment group I (0.134), treatment II (0.68), treatment III (0.42) compared to the control group.
Conclusion: In this study, it was found that α-SMA expression, NLR levels on the 3rd and 10th day of administration were reduced in group receiving combination of mesenchymal stem cells and bovine colostrum in the liver of post-resection Wistar rats by 50%.
Collapse
|
41
|
Zarubova J, Hasani-Sadrabadi MM, Norris SCP, Majedi FS, Xiao C, Kasko AM, Li S. Cell-Taxi: Mesenchymal Cells Carry and Transport Clusters of Cancer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203515. [PMID: 36307906 PMCID: PMC9772300 DOI: 10.1002/smll.202203515] [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: 06/10/2022] [Revised: 09/09/2022] [Indexed: 06/16/2023]
Abstract
Cell clusters that collectively migrate from primary tumors appear to be far more potent in forming distant metastases than single cancer cells. A better understanding of the collective cell migration phenomenon and the involvement of various cell types during this process is needed. Here, an in vitro platform based on inverted-pyramidal microwells to follow and quantify the collective migration of hundreds of tumor cell clusters at once is developed. These results indicate that mesenchymal stromal cells (MSCs) or cancer-associated fibroblasts (CAFs) in the heterotypic tumor cell clusters may facilitate metastatic dissemination by transporting low-motile cancer cells in a Rac-dependent manner and that extracellular vesicles secreted by mesenchymal cells only play a minor role in this process. Furthermore, in vivo studies show that cancer cell spheroids containing MSCs or CAFs have faster spreading rates. These findings highlight the active role of co-traveling stromal cells in the collective migration of tumor cell clusters and may help in developing better-targeted therapies.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, 14220, Czech Republic
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Sam C P Norris
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Fatemeh Sadat Majedi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Crystal Xiao
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Andrea M Kasko
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Song Li
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| |
Collapse
|
42
|
Ambrosio MR, Mosca G, Migliaccio T, Liguoro D, Nele G, Schonauer F, D’Andrea F, Liotti F, Prevete N, Melillo RM, Reale C, Ambrosino C, Miele C, Beguinot F, D’Esposito V, Formisano P. Glucose Enhances Pro-Tumorigenic Functions of Mammary Adipose-Derived Mesenchymal Stromal/Stem Cells on Breast Cancer Cell Lines. Cancers (Basel) 2022; 14:5421. [PMID: 36358839 PMCID: PMC9655059 DOI: 10.3390/cancers14215421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 10/13/2023] Open
Abstract
Adiposity and diabetes affect breast cancer (BC) progression. We addressed whether glucose may affect the interaction between mammary adipose tissue-derived mesenchymal stromal/stem cells (MAT-MSCs) and BC cells. Two-dimensional co-cultures and spheroids were established in 25 mM or 5.5 mM glucose (High Glucose-HG or Low Glucose-LG) by using MAT-MSCs and MCF7 or MDA-MB231 BC cells. Gene expression was measured by qPCR, while protein levels were measured by cytofluorimetry and ELISA. CD44high/CD24low BC stem-like sub-population was quantified by cytofluorimetry. An in vivo zebrafish model was assessed by injecting spheroid-derived labeled cells. MAT-MSCs co-cultured with BC cells showed an inflammatory/senescent phenotype with increased abundance of IL-6, IL-8, VEGF and p16INK4a, accompanied by altered levels of CDKN2A and LMNB1. BC cells reduced multipotency and increased fibrotic features modulating OCT4, SOX2, NANOG, αSMA and FAP in MAT-MSCs. Of note, these co-culture-mediated changes in MAT-MSCs were partially reverted in LG. Only in HG, MAT-MSCs increased CD44high/CD24low MCF7 sub-population and promoted their ability to form mammospheres. Injection in zebrafish embryos of HG spheroid-derived MCF7 and MAT-MSCs was followed by a significant cellular migration and caudal dissemination. Thus, MAT-MSCs enhance the aggressiveness of BC cells in a HG environment.
Collapse
Affiliation(s)
- Maria Rosaria Ambrosio
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Giusy Mosca
- Department of Translational Medicine, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Teresa Migliaccio
- Department of Translational Medicine, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Domenico Liguoro
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Gisella Nele
- Department of Public Health, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Fabrizio Schonauer
- Department of Public Health, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Francesco D’Andrea
- Department of Public Health, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Federica Liotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Nella Prevete
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
- Department of Translational Medicine, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Rosa Marina Melillo
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Carla Reale
- Institute of Genetic Research “G. Salvatore” Biogem, Via Camporeale, 83031 Ariano Irpino, Italy
| | - Concetta Ambrosino
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
- Institute of Genetic Research “G. Salvatore” Biogem, Via Camporeale, 83031 Ariano Irpino, Italy
- Department of Science and Technology, University of Sannio, Via De Sanctis, 82100 Benevento, Italy
| | - Claudia Miele
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Francesco Beguinot
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
- Department of Translational Medicine, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Vittoria D’Esposito
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
| | - Pietro Formisano
- URT “Genomic of Diabetes”, Institute for Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (IEOS-CNR), Via Pansini 5, 80131 Naples, Italy
- Department of Translational Medicine, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
43
|
Ebrahimighaei R, Sala-Newby GB, Hudson C, Kimura TE, Hathway T, Hawkins J, McNeill MC, Richardson R, Newby AC, Bond M. Combined role for YAP-TEAD and YAP-RUNX2 signalling in substrate-stiffness regulation of cardiac fibroblast proliferation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119329. [PMID: 35905788 PMCID: PMC7616274 DOI: 10.1016/j.bbamcr.2022.119329] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 06/15/2023]
Abstract
Cardiac fibrosis is associated with increased stiffness of the myocardial extracellular matrix (ECM) in part mediated by increased cardiac fibroblast proliferation However, our understanding of the mechanisms regulating cardiac fibroblast proliferation are incomplete. Here we characterise a novel mechanism involving a combined activation of Yes-associated protein (YAP) targets RUNX Family Transcription Factor 2 (RUNX2) and TEA Domain Transcription Factor (TEAD). We demonstrate that cardiac fibroblast proliferation is enhanced by interaction with a stiff ECM compared to a soft ECM. This is associated with activation of the transcriptional co-factor, YAP. We demonstrate that this stiffness induced activation of YAP enhances the transcriptional activity of both TEAD and RUNX2 transcription factors. Inhibition of either TEAD or RUNX2, using gene silencing, expression of dominant-negative mutants or pharmacological inhibition, reduces cardiac fibroblast proliferation. Using mutants of YAP, defective in TEAD or RUNX2 activation ability, we demonstrate a dual role of YAP-mediated activation of TEAD and RUNX2 for substrate stiffness induced cardiac fibroblast proliferation. Our data highlights a previously unrecognised role of YAP mediated RUNX2 activation for cardiac fibroblast proliferation in response to increased ECM stiffness.
Collapse
Affiliation(s)
- Reza Ebrahimighaei
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Graciela B Sala-Newby
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Claire Hudson
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Tomomi E Kimura
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Tom Hathway
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Joseph Hawkins
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Madeleine C McNeill
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Rebecca Richardson
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Andrew C Newby
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Mark Bond
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| |
Collapse
|
44
|
Koivunotko E, Snirvi J, Merivaara A, Harjumäki R, Rautiainen S, Kelloniemi M, Kuismanen K, Miettinen S, Yliperttula M, Koivuniemi R. Angiogenic Potential of Human Adipose-Derived Mesenchymal Stromal Cells in Nanofibrillated Cellulose Hydrogel. Biomedicines 2022; 10:2584. [PMID: 36289846 PMCID: PMC9599553 DOI: 10.3390/biomedicines10102584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Adipose-derived mesenchymal stromal cells (ASCs) hold great potential for cellular therapies by having immunomodulatory behavior and tissue regenerative properties. Due to the capability of ASCs to differentiate into endothelial cells (ECs) and other angiogenic cell types, such as pericytes, ASCs are a highly valuable source for stimulating angiogenesis. However, cellular therapies in tissue engineering have faced challenges in poor survival of the cells after transplantation, which is why a protective biomaterial scaffold is required. In this work, we studied the potential of nanofibrillated cellulose (NFC) hydrogel to be utilized as a suitable matrix for three-dimensional (3D) cell culturing of human-derived ASCs (hASCs) and studied their angiogenic properties and differentiation potential in ECs and pericytes. In addition, we tested the effect of hASC-conditioned medium and stimulation with angiopoietin-1 (Ang-1) on human umbilical vein endothelial cells (HUVECs) to induce blood vessel-type tube formation in NFC hydrogel. The hASCs were successfully 3D cell cultured in NFC hydrogel as they formed spheroids and had high cell viability with angiogenic features. Most importantly, they showed angiogenic potential by having pericyte-like characteristics when differentiated in EC medium, and their conditioned medium improved HUVEC viability and tube formation, which recalls the active paracrine properties. This study recommends NFC hydrogel for future use as an animal-free biomaterial scaffold for hASCs in therapeutic angiogenesis and other cell therapy purposes.
Collapse
Affiliation(s)
- Elle Koivunotko
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Jasmi Snirvi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Arto Merivaara
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Riina Harjumäki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Swarna Rautiainen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Minna Kelloniemi
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, 33520 Tampere, Finland
| | - Kirsi Kuismanen
- Department of Obstetrics and Gynecology, Tampere University Hospital, 33520 Tampere, Finland
| | - Susanna Miettinen
- Faculty of Medicine and Health Technologies, University of Tampere, 33520 Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, 33520 Tampere, Finland
| | - Marjo Yliperttula
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| | - Raili Koivuniemi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790 Helsinki, Finland
| |
Collapse
|
45
|
Ahangar P, Strudwick XL, Cowin AJ. Wound Healing from an Actin Cytoskeletal Perspective. Cold Spring Harb Perspect Biol 2022; 14:a041235. [PMID: 35074864 PMCID: PMC9341468 DOI: 10.1101/cshperspect.a041235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Wound healing requires a complex cascade of highly controlled and conserved cellular and molecular processes. These involve numerous cell types and extracellular matrix molecules regulated by the actin cytoskeleton. This microscopic network of filaments is present within the cytoplasm of all cells and provides the shape and mechanical support required for cell movement and proliferation. Here, an overview of the processes of wound healing are described from the perspective of the cell in relation to the actin cytoskeleton. Key points of discussion include the role of actin, its binding proteins, signaling pathways, and events that play significant roles in the phases of wound healing. The identification of cytoskeletal targets that can be used to manipulate and improve wound healing is included as an emerging area of focus that may inform future therapeutic approaches to improve healing of complex wounds.
Collapse
Affiliation(s)
- Parinaz Ahangar
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| | - Allison J Cowin
- Future Industries Institute, UniSA STEM, University of South Australia, South Australia, Adelaide 5000, Australia
| |
Collapse
|
46
|
Lecoutre S, Lambert M, Drygalski K, Dugail I, Maqdasy S, Hautefeuille M, Clément K. Importance of the Microenvironment and Mechanosensing in Adipose Tissue Biology. Cells 2022; 11:cells11152310. [PMID: 35954152 PMCID: PMC9367348 DOI: 10.3390/cells11152310] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
The expansion of adipose tissue is an adaptive mechanism that increases nutrient buffering capacity in response to an overall positive energy balance. Over the course of expansion, the adipose microenvironment undergoes continual remodeling to maintain its structural and functional integrity. However, in the long run, adipose tissue remodeling, typically characterized by adipocyte hypertrophy, immune cells infiltration, fibrosis and changes in vascular architecture, generates mechanical stress on adipose cells. This mechanical stimulus is then transduced into a biochemical signal that alters adipose function through mechanotransduction. In this review, we describe the physical changes occurring during adipose tissue remodeling, and how they regulate adipose cell physiology and promote obesity-associated dysfunction in adipose tissue.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Mélanie Lambert
- Labex Inflamex, Université Sorbonne Paris Nord, INSERM, F-93000 Bobigny, France;
| | - Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet Hospital, C2-94, 14186 Stockholm, Sweden;
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR 7622), IBPS, Sorbonne Université, F-75005 Paris, France;
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, F-75013 Paris, France
- Correspondence: or
| |
Collapse
|
47
|
Feng D, Gao P, Henley N, Dubuissez M, Chen N, Laurin LP, Royal V, Pichette V, Gerarduzzi C. SMOC2 promotes an epithelial-mesenchymal transition and a pro-metastatic phenotype in epithelial cells of renal cell carcinoma origin. Cell Death Dis 2022; 13:639. [PMID: 35869056 PMCID: PMC9307531 DOI: 10.1038/s41419-022-05059-2] [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: 11/23/2020] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 01/21/2023]
Abstract
Renal Cell Carcinoma (RCC) is the most common form of all renal cancer cases, and well-known for its highly aggressive metastatic behavior. SMOC2 is a recently described non-structural component of the extracellular matrix (ECM) that is highly expressed during tissue remodeling processes with emerging roles in cancers, yet its role in RCC remains elusive. Using gene expression profiles from patient samples, we identified SMOC2 as being significantly expressed in RCC tissue compared to normal renal tissue, which correlated with shorter RCC patient survival. Specifically, de novo protein synthesis of SMOC2 was shown to be much higher in the tubular epithelial cells of patients with biopsy-proven RCC. More importantly, we provide evidence of SMOC2 triggering kidney epithelial cells into an epithelial-to-mesenchymal transition (EMT), a phenotype known to promote metastasis. We found that SMOC2 induced mesenchymal-like morphology and activities in both RCC and non-RCC kidney epithelial cell lines. Mechanistically, treatment of RCC cell lines ACHN and 786-O with SMOC2 (recombinant and enforced expression) caused a significant increase in EMT-markers, -matrix production, -proliferation, and -migration, which were inhibited by targeting SMOC2 by siRNA. We further characterized SMOC2 activation of EMT to occur through the integrin β3, FAK and paxillin pathway. The proliferation and metastatic potential of SMOC2 overexpressing ACHN and 786-O cell lines were validated in vivo by their significantly higher tumor growth in kidneys and systemic dissemination into other organs when compared to their respective controls. In principle, understanding the impact that SMOC2 has on EMT may lead to more evidence-based treatments and biomarkers for RCC metastasis.
Collapse
Affiliation(s)
- Daniel Feng
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Peng Gao
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Nathalie Henley
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Marion Dubuissez
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Nan Chen
- Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Louis-Philippe Laurin
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Virginie Royal
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
| | - Vincent Pichette
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Casimiro Gerarduzzi
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l'Université de Montréal, Montréal, Québec, Canada.
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
48
|
Ando R, Sakai A, Iida T, Kataoka K, Mizutani Y, Enomoto A. Good and Bad Stroma in Pancreatic Cancer: Relevance of Functional States of Cancer-Associated Fibroblasts. Cancers (Basel) 2022; 14:cancers14143315. [PMID: 35884375 PMCID: PMC9317763 DOI: 10.3390/cancers14143315] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent progress in research on the biology of cancer-associated fibroblasts (CAFs) in pancreatic ductal adenocarcinoma (PDAC) indicates their diverse states and plasticity, which may lead to good and bad stroma, suppressing and promoting cancer progression, respectively. The characteristics of the stroma differ spatially, even within the same tumors, based on the balance between cancer-restraining CAF and cancer-promoting CAF proliferation at the site. These heterogeneous CAFs also influence the sensitivity of PDAC to anticancer therapeutics. Further preclinical and clinical studies will advance our understanding of the roles of CAFs in disease progression and aid the development of therapeutics that modulate or ameliorate the tumor microenvironment in PDAC. Abstract A well-known feature of human pancreatic ductal adenocarcinoma (PDAC) is the extensive proliferation of cancer-associated fibroblasts (CAFs) and highly fibrotic stroma. Recent evidence, based mainly on single-cell analyses, has identified various subsets of CAFs in PDAC mouse models. However, we do not know how these CAF subsets are involved in the progression and drug resistance of human PDAC. Additionally, it remains unclear whether these diverse CAFs have distinct origins and are indicators of genuinely distinct CAF lineages or reflect different states of the same CAFs depending on the tumor microenvironment. Interestingly, recent preclinical studies have started to characterize the nature of cancer-restraining CAFs and have identified their markers Meflin and collagen type I alpha 1. These studies have led to the development of strategies to induce changes in CAF phenotypes using chemical reagents or recombinant viruses, and some of them have been tested in clinical studies. These strategies have the unique potential to convert the so-called bad stroma to good stroma and may also have therapeutic implications for non-cancer diseases such as fibrotic diseases. Together with recently developed sophisticated strategies that specifically target distinct CAF subsets via adoptive cell transfer therapy, vaccination, and antibody–drug conjugates, any future findings arising from these clinical efforts may expand our understanding of the significance of CAF diversity in human PDAC.
Collapse
Affiliation(s)
- Ryota Ando
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (R.A.); (A.S.); (T.I.); (K.K.); (Y.M.)
| | - Akihiro Sakai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (R.A.); (A.S.); (T.I.); (K.K.); (Y.M.)
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (R.A.); (A.S.); (T.I.); (K.K.); (Y.M.)
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kunio Kataoka
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (R.A.); (A.S.); (T.I.); (K.K.); (Y.M.)
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (R.A.); (A.S.); (T.I.); (K.K.); (Y.M.)
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (R.A.); (A.S.); (T.I.); (K.K.); (Y.M.)
- Correspondence: ; Tel.: +81-52-744-2093
| |
Collapse
|
49
|
Peng Q, Shan D, Cui K, Li K, Zhu B, Wu H, Wang B, Wong S, Norton V, Dong Y, Lu YW, Zhou C, Chen H. The Role of Endothelial-to-Mesenchymal Transition in Cardiovascular Disease. Cells 2022; 11:1834. [PMID: 35681530 PMCID: PMC9180466 DOI: 10.3390/cells11111834] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Endothelial-to-mesenchymal transition (EndoMT) is the process of endothelial cells progressively losing endothelial-specific markers and gaining mesenchymal phenotypes. In the normal physiological condition, EndoMT plays a fundamental role in forming the cardiac valves of the developing heart. However, EndoMT contributes to the development of various cardiovascular diseases (CVD), such as atherosclerosis, valve diseases, fibrosis, and pulmonary arterial hypertension (PAH). Therefore, a deeper understanding of the cellular and molecular mechanisms underlying EndoMT in CVD should provide urgently needed insights into reversing this condition. This review summarizes a 30-year span of relevant literature, delineating the EndoMT process in particular, key signaling pathways, and the underlying regulatory networks involved in CVD.
Collapse
Affiliation(s)
- Qianman Peng
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dan Shan
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn Li
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Beibei Wang
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Scott Wong
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vikram Norton
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yunzhou Dong
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yao Wei Lu
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Changcheng Zhou
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
50
|
Li Z, Zhu Z, Liu Y, Liu Y, Zhao H. Function and regulation of GPX4 in the development and progression of fibrotic disease. J Cell Physiol 2022; 237:2808-2824. [PMID: 35605092 DOI: 10.1002/jcp.30780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023]
Abstract
Fibrosis is a common feature of fibrotic diseases that poses a serious threat to global health due to high morbidity and mortality in developing countries. There exist some chemical compounds and biomolecules associated with the development of fibrosis, including cytokines, hormones, and enzymes. Among them, glutathione peroxidase 4 (GPX4), as a selenoprotein antioxidant enzyme, is widely found in the embryo, testis, brain, liver, heart, and photoreceptor cells. Moreover, it is shown that GPX4 elicits diverse biological functions by suppressing phospholipid hydroperoxide at the expense of decreased glutathione (GSH), including loss of neurons, autophagy, cell repair, inflammation, ferroptosis, apoptosis, and oxidative stress. Interestingly, these processes are intimately related to the occurrence of fibrotic disease. Recently, GPX4 has been reported to exhibit a decline in fibrotic disease and inhibit fibrosis, suggesting that alterations of GPX4 can change the course or dictate the outcome of fibrotic disease. In this review, we summarize the role and underlying mechanisms of GPX4 in fibrosis diseases such as lung fibrosis, liver fibrosis, kidney fibrosis, cardiac fibrosis, and myelofibrosis.
Collapse
Affiliation(s)
- Zhaobing Li
- Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunnan, China
| | - Zigui Zhu
- Department of Intensive Care Units, The Affiliated Nanhua Hospital, Hengyang Medical school, University of South China, Hengyang, Hunnan, China
| | - Yulu Liu
- Department of Intensive Care Units, The Affiliated Nanhua Hospital, Hengyang Medical school, University of South China, Hengyang, Hunnan, China
| | - Yannan Liu
- School of Nursing, Hunan University of Medicine, Huaihua, Hunan, China
| | - Hong Zhao
- School of Nursing, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|