1
|
Zou AE, Kongthong S, Mueller AA, Brenner MB. Fibroblasts in immune responses, inflammatory diseases and therapeutic implications. Nat Rev Rheumatol 2025:10.1038/s41584-025-01259-0. [PMID: 40369134 DOI: 10.1038/s41584-025-01259-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/16/2025]
Abstract
Once regarded as passive bystander cells of the tissue stroma, fibroblasts have emerged as active orchestrators of tissue homeostasis and disease. From regulating immunity and controlling tissue remodelling to governing cell growth and differentiation, fibroblasts assume myriad roles in guiding normal tissue development, maintenance and repair. By comparison, in chronic inflammatory diseases such as rheumatoid arthritis, fibroblasts recruit and sustain inflammatory leukocytes, become dominant producers of pro-inflammatory factors and catalyse tissue destruction. In other disease contexts, fibroblasts promote fibrosis and impair host control of cancer. Single-cell studies have uncovered striking transcriptional and functional heterogeneity exhibited by fibroblasts in both normal tissues and diseased tissues. In particular, advances in the understanding of fibroblast pathology in rheumatoid arthritis have shed light on pathogenic fibroblast states in other chronic diseases. The differentiation and activation of these fibroblast states is driven by diverse physical and chemical cues within the tissue microenvironment and by cell-intrinsic signalling and epigenetic mechanisms. These insights into fibroblast behaviour and regulation have illuminated therapeutic opportunities for the targeted deletion or modulation of pathogenic fibroblasts across many diseases.
Collapse
Affiliation(s)
- Angela E Zou
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suppawat Kongthong
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alisa A Mueller
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA and Palo Alto Veterans Affairs Health Care System, Palo Alto, CA, USA
| | - Michael B Brenner
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Kong M, Wang Z, Hao Y, Shi Y, Yang X, Djurist NR, Li Y. Role of the integrin‑β1/TGF‑β1 signaling pathway in the pathogenesis of pelvic organ prolapse: A study on vaginal wall tissue alterations and molecular dysfunction. Mol Med Rep 2025; 31:104. [PMID: 39981910 PMCID: PMC11876943 DOI: 10.3892/mmr.2025.13469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 12/12/2024] [Indexed: 02/22/2025] Open
Abstract
Pelvic organ prolapse (POP) is a prevalent condition among middle‑aged and older women, and is associated with the irregular production and breakdown of the extracellular matrix. Mechanical forces serve a key role in preserving the equilibrium between matrix synthesis and degradation, thereby supporting the structural integrity of pelvic floor tissues. The aim of the present study was to investigate alterations in the composition of vaginal wall tissues in individuals suffering from POP and to investigate the molecular mechanisms through which mechanical forces trigger fibroblast apoptosis and influence collagen expression via the integrin‑β1/TGF‑β1 signaling pathway. Masson's trichrome and Elastica van Gieson staining were used to examine the pathological alterations in the tissue associated with POP. Analysis of immunofluorescence, western blotting and reverse transcription‑quantitative PCR data was performed to assess changes in the levels of proteins and genes such as collagen, integrin‑β1, TGF‑β1, MMP‑1 and tissue inhibitor of metalloproteinase‑1 (TIMP‑1). Fibroblasts were incubated with an integrin‑β1 antagonist RGD peptide to mimic cellular injury induced by mechanical forces, and cell migration and apoptosis were analyzed using scratch assays and flow cytometry. Cytoskeletal alterations were detected via immunofluorescence staining, and western blot analysis was conducted to examine the expression levels of integrin‑β1, TGF‑β1, TIMP‑1, MMP‑1, collagen type I α1 chain (COL1A1) and collagen type III α1 chain (COL3A1) across various groups. Analysis revealed that in the POP group, the collagen fibers in the vaginal wall tissues were loose and irregularly arranged, the number of elastic fibers was reduced and the structure was degraded. Furthermore, stress fibers were incomplete and their functions were impaired, resulting in damage to the connective tissue structure of the pelvic floor. Integrin‑β1 was key for fibroblast migration, apoptosis and collagen synthesis. Additionally, the integrin‑β1/TGF‑β1 signaling pathway served a role in mediating fibroblast apoptosis, and influencing the synthesis and metabolism of COL1A1 and COL3A1 induced by mechanical forces. Understanding the underlying pathogenesis of pelvic floor organ prolapse could pave the way for future investigations into innovative prevention and treatment strategies.
Collapse
Affiliation(s)
- Min Kong
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Zhuo Wang
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yao Hao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- Key Laboratory of Fertility Maintenance, Ministry of Education, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yueyue Shi
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Xin Yang
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Ngenzi Richard Djurist
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yan Li
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
3
|
Ayhan S, Dursun A. ELFN1 is a new extracellular matrix (ECM)-associated protein. Life Sci 2024; 352:122900. [PMID: 38986898 DOI: 10.1016/j.lfs.2024.122900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
AIMS The ELFN1, discovered in 2007, is a single-pass transmembrane protein. Studies conducted thus far to elucidate the function of the Elfn1 have been limited only to animal studies. These studies have reported that ELFN1 is a universal binding partner of metabotropic glutamate receptors (mGluRs) in the central nervous system and its functional deficiency has been associated with the pathogenesis of neurological and neuropsychiatric diseases. In 2021, we described the first disease-associated human ELFN1 pathogenic gene mutation. Severe joint laxity, which was the most striking finding of this new disease and was clearly seen in the patients since early infancy, showed that the ELFN1 may have a possible function in the connective tissue besides the nervous system. Here, we present the first experimental evidence of the extracellular matrix (ECM)-related function of the ELFN1. MATERIALS AND METHODS Primary skin fibroblasts were isolated from the skin biopsies of ELFN1 mutated patients and healthy foreskin donors. For the clinical trial in a dish, in vitro ECM and DEM (decellularized ECM) models were created from skin fibroblasts. All the in vitro models were comparatively characterized and analyzed. KEY FINDINGS The mutation in the ELFN1 signal peptide region of patients resulted in a severe lack of ELFN1 expression and dramatically altered the characteristic morphology and behavior (growth, proliferation, and motility) of fibroblasts. SIGNIFICANCE We propose that ELFN1 is involved in the cell-ECM attachment, and its deficiency is critical enough to cause a loss of cell motility and soft ECM stiffness.
Collapse
Affiliation(s)
- Selda Ayhan
- Department of Pediatrics Metabolism, Institute of Child Health, Hacettepe University, Sıhhıye, Ankara 06100, Turkey.
| | - Ali Dursun
- Department of Pediatrics Metabolism, Faculty of Medicine, Hacettepe University, Sıhhıye, Ankara 06100, Turkey.
| |
Collapse
|
4
|
Mamun AA, Shao C, Geng P, Wang S, Xiao J. Recent advances in molecular mechanisms of skin wound healing and its treatments. Front Immunol 2024; 15:1395479. [PMID: 38835782 PMCID: PMC11148235 DOI: 10.3389/fimmu.2024.1395479] [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: 03/04/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The skin, being a multifaceted organ, performs a pivotal function in the complicated wound-healing procedure, which encompasses the triggering of several cellular entities and signaling cascades. Aberrations in the typical healing process of wounds may result in atypical scar development and the establishment of a persistent condition, rendering patients more vulnerable to infections. Chronic burns and wounds have a detrimental effect on the overall quality of life of patients, resulting in higher levels of physical discomfort and socio-economic complexities. The occurrence and frequency of prolonged wounds are on the rise as a result of aging people, hence contributing to escalated expenditures within the healthcare system. The clinical evaluation and treatment of chronic wounds continue to pose challenges despite the advancement of different therapeutic approaches. This is mainly owing to the prolonged treatment duration and intricate processes involved in wound healing. Many conventional methods, such as the administration of growth factors, the use of wound dressings, and the application of skin grafts, are used to ease the process of wound healing across diverse wound types. Nevertheless, these therapeutic approaches may only be practical for some wounds, highlighting the need to advance alternative treatment modalities. Novel wound care technologies, such as nanotherapeutics, stem cell treatment, and 3D bioprinting, aim to improve therapeutic efficacy, prioritize skin regeneration, and minimize adverse effects. This review provides an updated overview of recent advancements in chronic wound healing and therapeutic management using innovative approaches.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Chuxiao Shao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Peiwu Geng
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Shuanghu Wang
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Jian Xiao
- Central Laboratory of The Lishui Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Wound Healing, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Wang F, Do TT, Smith N, Orringer JS, Kang S, Voorhees JJ, Fisher GJ. Implications for cumulative and prolonged clinical improvement induced by cross-linked hyaluronic acid: An in vivo biochemical/microscopic study in humans. Exp Dermatol 2024; 33:e14998. [PMID: 38284186 DOI: 10.1111/exd.14998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 01/30/2024]
Abstract
In photoaged human skin, type I collagen fragmentation impairs dermal extracellular matrix (ECM) integrity, resulting in collapsed/contracted fibroblasts with reduced type I procollagen synthesis. Injections of cross-linked hyaluronic acid (CL-HA) reverse these deleterious changes. To investigate the time course and effects of biochemical changes induced by injected CL-HA, particularly whether fibroblast activation leads to accumulation/deposition of dermal collagen, we injected CL-HA into photoaged skin of human participants over 60 years-old and performed biochemical/microscopic analyses of skin samples. Beginning 1 week post-injection and lasting 6-9 months, fibroblasts exhibited activation, including increased immunostaining and gene expression of markers of type I collagen synthesis, such as heat shock protein 47 and components of the transforming growth factor-β pathway. At 1 week post-injection, multiphoton microscopy revealed elongation/stretching of fibroblasts, indicating enhanced dermal mechanical support. At 4 weeks, second-harmonic generation microscopy revealed thick collagen bundles densely packed around pools of injected CL-HA. At 12 months, accumulation of thick collagen bundles was observed and injected CL-HA remained present in substantial amounts. Thus, by occupying space in the dermal ECM, injected CL-HA rapidly and durably enhances mechanical support, stimulating fibroblast elongation and activation, which results in thick, densely packed type I collagen bundles accumulating as early as 4 weeks post-injection and continuing for at least a year. These observations indicate that early and prolonged clinical improvement following CL-HA injection results from space-filling and collagen deposition. As type I collagen has an estimated half-life of 15 years, our data provide the foundations for optimizing the timing/frequency of repeat CL-HA injections.
Collapse
Affiliation(s)
- Frank Wang
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Thy Thy Do
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah Smith
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeffrey S Orringer
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sewon Kang
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - John J Voorhees
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gary J Fisher
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Kundu AN, Dougan CE, Mahmoud S, Kilic A, Panagiotou A, Richbourg N, Irakoze N, Peyton SR. Tenascin-C Activation of Lung Fibroblasts in a 3D Synthetic Lung Extracellular Matrix Mimic. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301493. [PMID: 37227134 PMCID: PMC10528529 DOI: 10.1002/adma.202301493] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/26/2023] [Indexed: 05/26/2023]
Abstract
The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that match the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro. Here, a synthetic, bioactive hydrogel is synthesized that mimics the native lung modulus and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin-binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which enables quiescent culture of human lung fibroblasts (HLFs). Stimulation with transforming growth factor β1 (TGF-β1), metastatic breast cancer conditioned media (CM), or tenascin-C-derived integrin-binding peptide activated hydrogel-encapsulated HLFs demonstrates multiple environmental methods to activate HLFs in a lung ECM-mimicking hydrogel. This lung hydrogel platform is a tunable, synthetic approach to studying the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.
Collapse
Affiliation(s)
- Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Carey E. Dougan
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Samar Mahmoud
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Alara Kilic
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst
| | - Alexi Panagiotou
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Nathan Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
- Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003
| |
Collapse
|
7
|
Josyula A, Mozzer A, Szeto J, Ha Y, Richmond N, Chung SW, Rompicharla SVK, Narayan J, Ramesh S, Hanes J, Ensign L, Parikh K, Pitha I. Nanofiber-based glaucoma drainage implant improves surgical outcomes by modulating fibroblast behavior. Bioeng Transl Med 2023; 8:e10487. [PMID: 37206200 PMCID: PMC10189467 DOI: 10.1002/btm2.10487] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/01/2022] [Accepted: 12/19/2022] [Indexed: 11/02/2023] Open
Abstract
Biomaterials are implanted in millions of individuals worldwide each year. Both naturally derived and synthetic biomaterials induce a foreign body reaction that often culminates in fibrotic encapsulation and reduced functional lifespan. In ophthalmology, glaucoma drainage implants (GDIs) are implanted in the eye to reduce intraocular pressure (IOP) in order to prevent glaucoma progression and vision loss. Despite recent efforts towards miniaturization and surface chemistry modification, clinically available GDIs are susceptible to high rates of fibrosis and surgical failure. Here, we describe the development of synthetic, nanofiber-based GDIs with partially degradable inner cores. We evaluated GDIs with nanofiber or smooth surfaces to investigate the effect of surface topography on implant performance. We observed in vitro that nanofiber surfaces supported fibroblast integration and quiescence, even in the presence of pro-fibrotic signals, compared to smooth surfaces. In rabbit eyes, GDIs with a nanofiber architecture were biocompatible, prevented hypotony, and provided a volumetric aqueous outflow comparable to commercially available GDIs, though with significantly reduced fibrotic encapsulation and expression of key fibrotic markers in the surrounding tissue. We propose that the physical cues provided by the surface of the nanofiber-based GDIs mimic healthy extracellular matrix structure, mitigating fibroblast activation and potentially extending functional GDI lifespan.
Collapse
Affiliation(s)
- Aditya Josyula
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Ann Mozzer
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Julia Szeto
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Youlim Ha
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Nicole Richmond
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Seung Woo Chung
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Sri Vishnu Kiran Rompicharla
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Janani Narayan
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Samiksha Ramesh
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Justin Hanes
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Departments of Pharmacology and Molecular Sciences, Environmental Health Sciences, Oncology, and NeurosurgeryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Laura Ensign
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Departments of Pharmacology and Molecular Sciences, Infectious Diseases, Oncology, and Gynecology and ObstetricsJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kunal Parikh
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Center for Bioengineering Innovation & DesignJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Ian Pitha
- Center for NanomedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Ophthalmology, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Glaucoma Center of Excellence, Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
8
|
Kundu AN, Dougan CE, Mahmoud S, Kilic A, Panagiotou A, Irakoze N, Richbourg N, Peyton SR. Tenascin-C activation of lung fibroblasts in a 3D synthetic lung extracellular matrix mimic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529926. [PMID: 36865293 PMCID: PMC9980292 DOI: 10.1101/2023.02.24.529926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that contain the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro . Here, we developed a synthetic, bioactive hydrogel that mimics the native lung modulus, and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which promotes quiescence of human lung fibroblasts (HLFs). Stimulation with transforming growth factor β1 (TGF-β1), metastatic breast cancer conditioned media (CM), or tenascin-C activated these hydrogel-encapsulated HLFs in a manner reflective of their native in vivo responses. We propose this lung hydrogel platform as a tunable, synthetic approach to study the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.
Collapse
Affiliation(s)
- Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Carey E. Dougan
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Samar Mahmoud
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Alara Kilic
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst
| | - Alexi Panagiotou
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Nathan Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
- Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003
| |
Collapse
|
9
|
Berry CE, Downer M, Morgan AG, Griffin M, Liang NE, Kameni L, Laufey Parker JB, Guo J, Longaker MT, Wan DC. The effects of mechanical force on fibroblast behavior in cutaneous injury. Front Surg 2023; 10:1167067. [PMID: 37143767 PMCID: PMC10151708 DOI: 10.3389/fsurg.2023.1167067] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
Wound healing results in the formation of scar tissue which can be associated with functional impairment, psychological stress, and significant socioeconomic cost which exceeds 20 billion dollars annually in the United States alone. Pathologic scarring is often associated with exaggerated action of fibroblasts and subsequent excessive accumulation of extracellular matrix proteins which results in fibrotic thickening of the dermis. In skin wounds, fibroblasts transition to myofibroblasts which contract the wound and contribute to remodeling of the extracellular matrix. Mechanical stress on wounds has long been clinically observed to result in increased pathologic scar formation, and studies over the past decade have begun to uncover the cellular mechanisms that underly this phenomenon. In this article, we will review the investigations which have identified proteins involved in mechano-sensing, such as focal adhesion kinase, as well as other important pathway components that relay the transcriptional effects of mechanical forces, such as RhoA/ROCK, the hippo pathway, YAP/TAZ, and Piezo1. Additionally, we will discuss findings in animal models which show the inhibition of these pathways to promote wound healing, reduce contracture, mitigate scar formation, and restore normal extracellular matrix architecture. Recent advances in single cell RNA sequencing and spatial transcriptomics and the resulting ability to further characterize mechanoresponsive fibroblast subpopulations and the genes that define them will be summarized. Given the importance of mechanical signaling in scar formation, several clinical treatments focused on reducing tension on the wound have been developed and are described here. Finally, we will look toward future research which may reveal novel cellular pathways and deepen our understanding of the pathogenesis of pathologic scarring. The past decade of scientific inquiry has drawn many lines connecting these cellular mechanisms that may lead to a map for the development of transitional treatments for patients on the path to scarless healing.
Collapse
Affiliation(s)
- Charlotte E. Berry
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Mauricio Downer
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Annah G. Morgan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Norah E. Liang
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Lionel Kameni
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Jennifer B. Laufey Parker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Jason Guo
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Derrick C. Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Correspondence: Derrick C. Wan
| |
Collapse
|
10
|
Hosseini M, Brown J, Khosrotehrani K, Bayat A, Shafiee A. Skin biomechanics: a potential therapeutic intervention target to reduce scarring. BURNS & TRAUMA 2022; 10:tkac036. [PMID: 36017082 PMCID: PMC9398863 DOI: 10.1093/burnst/tkac036] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/27/2022] [Indexed: 12/19/2022]
Abstract
Pathological scarring imposes a major clinical and social burden worldwide. Human cutaneous wounds are responsive to mechanical forces and convert mechanical cues to biochemical signals that eventually promote scarring. To understand the mechanotransduction pathways in cutaneous scarring and develop new mechanotherapy approaches to achieve optimal scarring, the current study highlights the mechanical behavior of unwounded and scarred skin as well as intra- and extracellular mechanisms behind keloid and hypertrophic scars. Additionally, the therapeutic interventions that promote optimal scar healing by mechanical means at the molecular, cellular or tissue level are extensively reviewed. The current literature highlights the significant role of fibroblasts in wound contraction and scar formation via differentiation into myofibroblasts. Thus, understanding myofibroblasts and their responses to mechanical loading allows the development of new scar therapeutics. A review of the current clinical and preclinical studies suggests that existing treatment strategies only reduce scarring on a small scale after wound closure and result in poor functional and aesthetic outcomes. Therefore, the perspective of mechanotherapies needs to consider the application of both mechanical forces and biochemical cues to achieve optimal scarring. Moreover, early intervention is critical in wound management; thus, mechanoregulation should be conducted during the healing process to avoid scar maturation. Future studies should either consider combining mechanical loading (pressure) therapies with tension offloading approaches for scar management or developing more effective early therapies based on contraction-blocking biomaterials for the prevention of pathological scarring.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering (MMPE), Faculty of Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Jason Brown
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Ardeshir Bayat
- Centre for Dermatology Research, NIHR Manchester Biomedical Research Centre, Stopford Building, University of Manchester, Oxford Road, Manchester, M13 9PT, England, UK
| | - Abbas Shafiee
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
| |
Collapse
|
11
|
The Stiffness of Cardiac Fibroblast Substrates Exerts a Regulatory Influence on Collagen Metabolism via α2β1 Integrin, FAK and Src Kinases. Cells 2021; 10:cells10123506. [PMID: 34944014 PMCID: PMC8700012 DOI: 10.3390/cells10123506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 12/15/2022] Open
Abstract
Information about mechanical strain in the extracellular space is conducted along collagen fibers connected with integrins and then transmitted within cells. An aim of the study is to verify the hypothesis that the stiffness of cardiac human fibroblast substrates exerts a regulatory effect on collagen metabolism via integrin α2β1 and downstream signaling. The experiments were performed on human cardiac fibroblasts cultured on stiff or soft polyacrylamide gels. Extracellular and intracellular collagen content, metalloproteinase-1 (MMP-1), metalloproteinase-9 (MMP-9) and expression of the α1 chain of the procollagen type I gene (Col1A1) were elevated in cultures settled on soft substrate. The substrate stiffness did not modify tissue inhibitors of matrix metalloproteinase capacity (TIMPs 1–4). Integrin α2β1 inhibition (TC-I 15) or α2 subunit silencing resulted in augmentation of collagen content within the culture. Expression of Col1A1 and Col3A1 genes was increased in TC-I 15-treated fibroblasts. Total and phosphorylated levels of both FAK and Src kinases were elevated in fibroblasts cultured on stiff substrate. Inhibition of FAK (FAK kinase inhibitor 14) or Src kinase (AZM 47527) increased collagen content within the culture. The substrate stiffness exerted a regulatory influence on collagen metabolism via integrin α2β1 and its downstream signaling (FAK and Src kinases) in cardiac fibroblasts.
Collapse
|
12
|
Douillet C, Nicodeme M, Hermant L, Bergeron V, Guillemot F, Fricain JC, Oliveira H, Garcia M. From local to global matrix organization by fibroblasts: a 4D laser-assisted bioprinting approach. Biofabrication 2021; 14. [PMID: 34875632 DOI: 10.1088/1758-5090/ac40ed] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/07/2021] [Indexed: 11/11/2022]
Abstract
Fibroblasts and myofibroblasts play a central role in skin homeostasis through dermal organization and maintenance. Nonetheless, the dynamic interactions between (myo)fibroblasts and the extracellular matrix (ECM) remain poorly exploited in skin repair strategies. Indeed, there is still an unmet need for soft tissue models allowing to study the spatial-temporal remodeling properties of (myo)fibroblasts. In vivo, wound healing studies in animals are limited by species specificity. In vitro, most models rely on collagen gels reorganized by randomly distributed fibroblasts. But biofabrication technologies have significantly evolved over the past ten years. High-resolution bioprinting now allows to investigate various cellular micropatterns and the emergent tissue organizations over time. In order to harness the full dynamic properties of cells and active biomaterials, it is essential to consider "time" as the 4th dimension in soft tissue design. Following this 4D bioprinting approach, we aimed to develop a novel model that could replicate fibroblast dynamic remodeling in vitro. For this purpose, (myo)fibroblasts were patterned on collagen gels with laser-assisted bioprinting (LAB) to study the generated matrix deformations and reorganizations. First, distinct populations, mainly composed of fibroblasts or myofibroblasts, were established in vitro to account for the variety of fibroblastic remodeling properties. Then, LAB was used to organize both populations on collagen gels in even isotropic patterns with high resolution, high density and high viability. With maturation, bioprinted patterns of fibroblasts and myofibroblasts reorganized into dispersed or aggregated cells, respectively. Stress-release contraction assays revealed that these phenotype-specific pattern maturations were associated with distinct lattice tension states. The two populations were then patterned in anisotropic rows in order to direct the cell-generated deformations and to orient global matrix remodeling. Only maturation of anisotropic fibroblast patterns, but not myofibroblasts, resulted in collagen anisotropic reorganizations both at tissue-scale, with lattice contraction, and at microscale, with embedded microbead displacements. Following a 4D bioprinting approach, LAB patterning enabled to elicit and orient the dynamic matrix remodeling mechanisms of distinct fibroblastic populations and organizations on collagen. For future studies, this method provides a new versatile tool to investigate in vitro dermal organizations and properties, processes of remodeling in healing, and new treatment opportunities.
Collapse
Affiliation(s)
- Camille Douillet
- Bioingénierie tissulaire, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, Aquitaine, 33076, FRANCE
| | - Marc Nicodeme
- Poietis, 27 Allée Charles Darwin, Pessac, 33600, FRANCE
| | - Loïc Hermant
- Poietis, 27 Allée Charles Darwin, Pessac, 33600, FRANCE
| | | | | | - Jean-Christophe Fricain
- Bioingénierie tissulaire, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, 33076, FRANCE
| | - Hugo Oliveira
- Bioingénierie tissulaire, Université de Bordeaux, 146 rue Léo Saignat, Bordeaux, 33076, FRANCE
| | - Mikael Garcia
- Poietis, 27 Allée Charles Darwin, Pessac, 33600, FRANCE
| |
Collapse
|
13
|
Winter A, Salamonsen LA, Evans J. Modelling fibroid pathology: development and manipulation of a myometrial smooth muscle cell macromolecular crowding model to alter extracellular matrix deposition. Mol Hum Reprod 2021; 26:498-509. [PMID: 32449756 DOI: 10.1093/molehr/gaaa036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/31/2022] Open
Abstract
Current treatment options for uterine fibroids are limited to hormonal manipulation or surgical intervention. We aimed to develop an in vitro model to mirror collagen deposition and extracellular matrix (ECM) formation, the principal features of uterine fibroids, to enable testing of novel therapeutics. Macromolecular crowding with Ficoll 400 and Ficoll 70 in cultures of human uterine myometrial smooth muscle cells containing ascorbic acid, provided the basis for this model. These culture conditions mimic the 'crowded' nature of the in vivo extracellular environment by incorporating neutral, space-filling macromolecules into conventional cell cultures. This method of culture facilitates appropriate ECM deposition, thus closely representing the in vivo fibrotic phenotype of uterine fibroids. Macromolecular crowding in Ficoll cultures containing ascorbic acid reduced myometrial smooth muscle cell proliferation and promoted collagen production. Under these conditions, collagen was processed for extracellular deposition as demonstrated by C-propeptide cleavage from secreted procollagen. The fibrosis marker activin was increased relative to its natural inhibitor, follistatin, in crowded culture conditions while addition of exogenous follistatin reduced collagen (Col1A1) gene expression. This in vitro model represents a promising development for the testing of therapeutic interventions for uterine fibroids. However, it does not recapitulate the full in vivo pathology which can include specific genetic and epigenetic alterations that have not been identified in the myometrial smooth muscle (hTERT-HM) cell line. Following screening of potential therapeutics using the model, the most promising compounds will require further assessment in the context of individual subjects including those with genetic changes implicated in fibroid pathogenesis.
Collapse
Affiliation(s)
- Ann Winter
- Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Obstetrics & Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Lois A Salamonsen
- Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Jemma Evans
- Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
14
|
Mechanical and Immunological Regulation in Wound Healing and Skin Reconstruction. Int J Mol Sci 2021; 22:ijms22115474. [PMID: 34067386 PMCID: PMC8197020 DOI: 10.3390/ijms22115474] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/07/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022] Open
Abstract
In the past decade, a new frontier in scarless wound healing has arisen because of significant advances in the field of wound healing realised by incorporating emerging concepts from mechanobiology and immunology. The complete integumentary organ system (IOS) regeneration and scarless wound healing mechanism, which occurs in specific species, body sites and developmental stages, clearly shows that mechanical stress signals and immune responses play important roles in determining the wound healing mode. Advances in tissue engineering technology have led to the production of novel human skin equivalents and organoids that reproduce cell–cell interactions with tissue-scale tensional homeostasis, and enable us to evaluate skin tissue morphology, functionality, drug response and wound healing. This breakthrough in tissue engineering has the potential to accelerate the understanding of wound healing control mechanisms through complex mechanobiological and immunological interactions. In this review, we present an overview of recent studies of biomechanical and immunological wound healing and tissue remodelling mechanisms through comparisons of species- and developmental stage-dependent wound healing mechanisms. We also discuss the possibility of elucidating the control mechanism of wound healing involving mechanobiological and immunological interaction by using next-generation human skin equivalents.
Collapse
|
15
|
Rap1a Regulates Cardiac Fibroblast Contraction of 3D Diabetic Collagen Matrices by Increased Activation of the AGE/RAGE Cascade. Cells 2021; 10:cells10061286. [PMID: 34067282 PMCID: PMC8224555 DOI: 10.3390/cells10061286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease is a common diabetic complication that can arise when cardiac fibroblasts transition into myofibroblasts. Myofibroblast transition can be induced by advanced glycated end products (AGEs) present in the extracellular matrix (ECM) activating RAGE (receptor for advanced glycated end products) to elicit intracellular signaling. The levels of AGEs are higher under diabetic conditions due to the hyperglycemic conditions present in diabetics. AGE/RAGE signaling has been shown to alter protein expression and ROS production in cardiac fibroblasts, resulting in changes in cellular function, such as migration and contraction. Recently, a small GTPase, Rap1a, has been identified to overlap the AGE/RAGE signaling cascade and mediate changes in protein expression. While Rap1a has been shown to impact AGE/RAGE-induced protein expression, there are currently no data examining the impact Rap1a has on AGE/RAGE-induced cardiac fibroblast function. Therefore, we aimed to determine the impact of Rap1a on AGE/RAGE-mediated cardiac fibroblast contraction, as well as the influence isolated diabetic ECM has on facilitating these effects. In order to address this idea, genetically different cardiac fibroblasts were embedded in 3D collagen matrices consisting of collagen isolated from either non-diabetic of diabetic mice. Fibroblasts were treated with EPAC and/or exogenous AGEs, which was followed by assessment of matrix contraction, protein expression (α-SMA, SOD-1, and SOD-2), and hydrogen peroxide production. The results showed Rap1a overlaps the AGE/RAGE cascade to increase the myofibroblast population and generation of ROS production. The increase in myofibroblasts and oxidative stress appeared to contribute to increased matrix contraction, which was further exacerbated by diabetic conditions. Based off these results, we determined that Rap1a was essential in mediating the response of cardiac fibroblasts to AGEs within diabetic collagen.
Collapse
|
16
|
Meagher PB, Lee XA, Lee J, Visram A, Friedberg MK, Connelly KA. Cardiac Fibrosis: Key Role of Integrins in Cardiac Homeostasis and Remodeling. Cells 2021; 10:cells10040770. [PMID: 33807373 PMCID: PMC8066890 DOI: 10.3390/cells10040770] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiac fibrosis is a common finding that is associated with the progression of heart failure (HF) and impacts all chambers of the heart. Despite intense research, the treatment of HF has primarily focused upon strategies to prevent cardiomyocyte remodeling, and there are no targeted antifibrotic strategies available to reverse cardiac fibrosis. Cardiac fibrosis is defined as an accumulation of extracellular matrix (ECM) proteins which stiffen the myocardium resulting in the deterioration cardiac function. This occurs in response to a wide range of mechanical and biochemical signals. Integrins are transmembrane cell adhesion receptors, that integrate signaling between cardiac fibroblasts and cardiomyocytes with the ECM by the communication of mechanical stress signals. Integrins play an important role in the development of pathological ECM deposition. This review will discuss the role of integrins in mechano-transduced cardiac fibrosis in response to disease throughout the myocardium. This review will also demonstrate the important role of integrins as both initiators of the fibrotic response, and modulators of fibrosis through their effect on cardiac fibroblast physiology across the various heart chambers.
Collapse
Affiliation(s)
- Patrick B. Meagher
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Xavier Alexander Lee
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Joseph Lee
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Aylin Visram
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Mark K. Friedberg
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Labatt Family Heart Center and Department of Paediatrics, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Kim A. Connelly
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (P.B.M.); (X.A.L.); (J.L.); (A.V.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence: ; Tel.: +141-686-45201
| |
Collapse
|
17
|
He J, Fang B, Shan S, Xie Y, Wang C, Zhang Y, Zhang X, Li Q. Mechanical stretch promotes hypertrophic scar formation through mechanically activated cation channel Piezo1. Cell Death Dis 2021; 12:226. [PMID: 33649312 PMCID: PMC7921104 DOI: 10.1038/s41419-021-03481-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/27/2022]
Abstract
Hypertrophic scar (HS) formation is a skin fibroproliferative disease that occurs following a cutaneous injury, leading to functional and cosmetic impairment. To date, few therapeutic treatments exhibit satisfactory outcomes. The mechanical force has been shown to be a key regulator of HS formation, but the underlying mechanism is not completely understood. The Piezo1 channel has been identified as a novel mechanically activated cation channel (MAC) and is reportedly capable of regulating force-mediated cellular biological behaviors. However, the mechanotransduction role of Piezo1 in HS formation has not been investigated. In this work, we found that Piezo1 was overexpressed in myofibroblasts of human and rat HS tissues. In vitro, cyclic mechanical stretch (CMS) increased Piezo1 expression and Piezo1-mediated calcium influx in human dermal fibroblasts (HDFs). In addition, Piezo1 activity promoted HDFs proliferation, motility, and differentiation in response to CMS. More importantly, intradermal injection of GsMTx4, a Piezo1-blocking peptide, protected rats from stretch-induced HS formation. Together, Piezo1 was shown to participate in HS formation and could be a novel target for the development of promising therapies for HS formation.
Collapse
Affiliation(s)
- Jiahao He
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Bin Fang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Yun Xie
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China
| | - Chuandong Wang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), 200092, Shanghai, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), 200092, Shanghai, China.
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200011, Shanghai, China.
| |
Collapse
|
18
|
Dutta D, Markhoff J, Suter N, Rezwan K, Brüggemann D. Effect of Collagen Nanofibers and Silanization on the Interaction of HaCaT Keratinocytes and 3T3 Fibroblasts with Alumina Nanopores. ACS APPLIED BIO MATERIALS 2021; 4:1852-1862. [DOI: 10.1021/acsabm.0c01538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Deepanjalee Dutta
- Institute for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
| | - Jana Markhoff
- Institute for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
| | - Naiana Suter
- Institute for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
| | - Kurosch Rezwan
- Advanced Ceramics, University of Bremen, Am Biologischen Garten 2, 28359 Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, 28359 Bremen, Germany
| | - Dorothea Brüggemann
- Institute for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, 28359 Bremen, Germany
| |
Collapse
|
19
|
Pan P, Cheng J, Si Y, Chen W, Hou J, Zhao T, Gu Y, Lv L, Hong Z, Zhu Z, Chai Y, Guo Z, Chen X. A stop-flow comprehensive two-dimensional HK-2 and HK-2/CIKI cell membrane chromatography comparative analysis system for screening the active ingredients from Pyrrosia calvata (Bak.) Ching against crystal-induced kidney injury. J Pharm Biomed Anal 2020; 195:113825. [PMID: 33339641 DOI: 10.1016/j.jpba.2020.113825] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/06/2020] [Accepted: 12/02/2020] [Indexed: 12/29/2022]
Abstract
Crystal-induced kidney injury (CIKI) is the fundamental pathological change during nephrolithiasis, although the molecular mechanism is still unclear. Pyrrosia calvata (Bak.) Ching has been used in folk medicine to treat urolithiasis for years. To clarify the pharmacodynamic substances and the mechanism of its antiurolithiasis effects, in this study, a novel, stop-flow, comprehensive, two-dimensional (2D) HK-2 and HK-2/CIKI cell membrane chromatography (CMC) comparative analysis system was developed to screen for the potential active ingredients from Pyrrosia calvata (Bak.) Ching against CIKI. The comprehensive 2D CMC comparative analysis system showed satisfactory selectivity, and eight ingredients were screened and identified by this system. Among them, mangiferin exhibited higher affinity for the HK-2/CIKI CMC column than the HK-2 CMC column and was selected for further efficacy verification. Cell proliferation assays showed that mangiferin could protect HK-2 cell viability after stimulation with sodium oxalate (NaOX). Additionally, in a rodent model of CIKI, mangiferin decreased the deposition of calcium oxalate (CaOX) crystals in mouse kidneys, alleviated the pathological damage to kidney tissue, and inhibited the upregulation of OPN, MCP1, and CD44 expression caused by CaOX crystals. The established comprehensive 2D CMC comparative analysis system can be applied to screen active ingredients with disease specificity from traditional Chinese medicine (TCM) and is suitable for other cell models.
Collapse
Affiliation(s)
- Pengchao Pan
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Jin Cheng
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Yachen Si
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Wei Chen
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Jiebin Hou
- Department of Nephrology, The Second Medical Centre, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Tingting Zhao
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China
| | - Yanqiu Gu
- Department of Pharmacy, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, No. 280 Mohe Road, Shanghai, 201999, China
| | - Lei Lv
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Naval Medical University, No. 225 Changhai Road, Shanghai, 200438, China
| | - Zhanying Hong
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Zhenyu Zhu
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai, 200433, China
| | - Yifeng Chai
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai, 200433, China.
| | - Zhiyong Guo
- Department of Nephrology, Shanghai Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai, 200433, China.
| | - Xiaofei Chen
- School of Pharmacy, Naval Medical University, No. 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
20
|
Roofeh D, Lescoat A, Khanna D. Emerging drugs for the treatment of scleroderma: a review of recent phase 2 and 3 trials. Expert Opin Emerg Drugs 2020; 25:455-466. [PMID: 33054463 PMCID: PMC7770026 DOI: 10.1080/14728214.2020.1836156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Systemic sclerosis (SSc) has the highest case-specific mortality of all connective tissue diseases. Its underlying disease mechanism affects several organs and remains incompletely understood. Ongoing work clarifying its etiopathogenesis is helping to develop targeted therapy. AREAS COVERED Several clinical trials have evaluated the safety and efficacy of agents targeting different mechanisms of this disease. This review article reviews those mechanisms and surveys four key recent phase II or III clinical trials that are contributing to the landscape of SSc therapy. The reported trials primarily focus on patients with systemic sclerosis in the early phase of disease. EXPERT OPINION Traditional therapies for SSc center on immunosuppressive and cytotoxic agents. A new cadre of therapies is borne from improved understandings of SSc pathobiology and target the inflammatory-fibrotic pathways. Scleroderma trials have entered the initial phase of personalized medicine, recognizing molecular subsets that will improve upon cohort enrichment and maximize the measurable benefit of future therapies.
Collapse
Affiliation(s)
| | - Alain Lescoat
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | | |
Collapse
|
21
|
Ali SM. In vivo confocal Raman spectroscopic imaging of the human skin extracellular matrix degradation due to accumulated intrinsic and extrinsic aging. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2020; 37:140-152. [DOI: 10.1111/phpp.12623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/07/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Syed Mehmood Ali
- Department of Biomedical Engineering College of Engineering Imam Abdulrahman Bin Faisal University Dammam Saudi Arabia
| |
Collapse
|
22
|
Tissue-scale tensional homeostasis in skin regulates structure and physiological function. Commun Biol 2020; 3:637. [PMID: 33127987 PMCID: PMC7603398 DOI: 10.1038/s42003-020-01365-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Tensional homeostasis is crucial for organ and tissue development, including the establishment of morphological and functional properties. Skin plays essential roles in waterproofing, cushioning and protecting deeper tissues by forming internal tension-distribution patterns, which involves aligning various cells, appendages and extracellular matrices (ECMs). The balance of traction force is thought to contribute to the formation of strong and pliable physical structures that maintain their integrity and flexibility. Here, by using a human skin equivalent (HSE), the horizontal tension-force balance of the dermal layer was found to clearly improve HSE characteristics, such as the physical relationship between cells and the ECM. The tension also promoted skin homeostasis through the activation of mechano-sensitive molecules such as ROCK and MRTF-A, and these results compared favourably to what was observed in tension-released models. Tension-induced HSE will contribute to analyze skin physiological functions regulated by tensional homeostasis as an alternative animal model.
Collapse
|
23
|
Gałdyszyńska M, Bobrowska J, Lekka M, Radwańska P, Piera L, Szymański J, Drobnik J. The stiffness-controlled release of interleukin-6 by cardiac fibroblasts is dependent on integrin α2β1. J Cell Mol Med 2020; 24:13853-13862. [PMID: 33124775 PMCID: PMC7754059 DOI: 10.1111/jcmm.15974] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiac fibroblasts are able to sense the rigidity of their environment. The present study examines whether the stiffness of the substrate in cardiac fibroblast culture can influence the release of interleukin‐6 (IL‐6), interleukin‐11 (IL‐11) and soluble receptor of IL‐6 (sIL‐6R). It also examines the roles of integrin α2β1 activation and intracellular signalling in these processes. Cardiac fibroblasts were cultured on polyacrylamide gels and grafted to collagen, with an elasticity of E = 2.23 ± 0.8 kPa (soft gel) and E = 8.28 ± 1.06 kPa (stiff gel, measured by Atomic Force Microscope). Flow cytometry and ELISA demonstrated that the fibroblasts cultured on the soft gel demonstrated higher expression of the α2 integrin subunit and increased α2β1 integrin count and released higher levels of IL‐6 and sIL‐6R than those on the stiff gel. Substrate elasticity did not modify fibroblast IL‐11 content. The silencing of the α2 integrin subunit decreased the release of IL‐6. Similar effects were induced by TC‐I 15 (an α2β1 integrin inhibitor). The IL‐6 levels in the serum and heart were markedly lower in α2 integrin‐deficient mice B6.Cg‐Itga2tm1.1Tkun/tm1.1Tkun than wild type. Inhibition of Src kinase by AZM 475271 modifies the IL‐6 level. sIL‐6R secretion is not dependent on α2β1 integrin. Conclusion: The elastic properties of the substrate influence the release of IL‐6 by cardiac fibroblasts, and this effect is dependent on α2β1 integrin and kinase Src activation.
Collapse
Affiliation(s)
- Małgorzata Gałdyszyńska
- Laboratory of Connective Tissue Metabolism, Department of Pathophysiology, Medical University of Lodz, Lodz, Poland
| | | | | | - Paulina Radwańska
- Laboratory of Connective Tissue Metabolism, Department of Pathophysiology, Medical University of Lodz, Lodz, Poland
| | - Lucyna Piera
- Laboratory of Connective Tissue Metabolism, Department of Pathophysiology, Medical University of Lodz, Lodz, Poland
| | - Jacek Szymański
- Central Scientific Laboratory, Medical University of Lodz, Lodz, Poland
| | - Jacek Drobnik
- Laboratory of Connective Tissue Metabolism, Department of Pathophysiology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
24
|
Tunable bioactivity and mechanics of collagen-based tissue engineering constructs: A comparison of EDC-NHS, genipin and TG2 crosslinkers. Biomaterials 2020; 254:120109. [PMID: 32480093 PMCID: PMC7298615 DOI: 10.1016/j.biomaterials.2020.120109] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 02/05/2023]
Abstract
Due to its ubiquity and versatility in the human body, collagen is an ideal base material for tissue-engineering constructs. Chemical crosslinking treatments allow precise control of the biochemical and mechanical properties through macromolecular modifications to the structure of collagen. In this work, three key facets regarding the collagen crosslinking process are explored. Firstly, a comparison is drawn between the carbodiimide-succinimide (EDC-NHS) system and two emerging crosslinkers utilising alternate chemistries: genipin and tissue transglutaminase (TG2). By characterising the chemical changes upon treatment, the effect of EDC-NHS, genipin and TG2 crosslinking mechanisms on the chemical structure of collagen, and thus the mechanical properties conferred to the substrate is explored. Secondly, the relative importance of mechanical and biochemical cues on cellular phenomena are investigated, including cell viability, integrin-specific attachment, spreading and proliferation. Here, we observe that for human dermal fibroblasts, long-term, stable proliferation is preconditioned by the availability of suitable binding sites, irrespective of the substrate modulus post-crosslinking. Finally, as seen in the graphical abstract we show that by choosing the appropriate crosslinker chemistries, a materials selection map can be drawn for collagen films, encompassing both a range of tensile modulus and fibroblast proliferation which can be modified independently. Thus, in addition to a range of parameters that can be modified in collagen constructs, we demonstrate a route to obtaining tunable bioactivity and mechanics in collagen constructs is uncovered, that is exclusively driven by the crosslinking process.
Collapse
|
25
|
Monemian Esfahani A, Rosenbohm J, Reddy K, Jin X, Bouzid T, Riehl B, Kim E, Lim JY, Yang R. Tissue Regeneration from Mechanical Stretching of Cell-Cell Adhesion. Tissue Eng Part C Methods 2019; 25:631-640. [PMID: 31407627 PMCID: PMC6859692 DOI: 10.1089/ten.tec.2019.0098] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/05/2019] [Indexed: 01/09/2023] Open
Abstract
Cell-cell adhesion complexes are macromolecular adhesive organelles that integrate cells into tissues. This mechanochemical coupling in cell-cell adhesion is required for a large number of cell behaviors, and perturbations of the cell-cell adhesion structure or related mechanotransduction pathways can lead to critical pathological conditions such as skin and heart diseases, arthritis, and cancer. Mechanical stretching has been a widely used method to stimulate the mechanotransduction process originating from the cell-cell adhesion and cell-extracellular matrix (ECM) complexes. These studies aimed to reveal the biophysical processes governing cell proliferation, wound healing, gene expression regulation, and cell differentiation in various tissues, including cardiac, muscle, vascular, and bone. This review explores techniques in mechanical stretching in two-dimensional settings with different stretching regimens on different cell types. The mechanotransduction responses from these different cell types will be discussed with an emphasis on their biophysical transformations during mechanical stretching and the cross talk between the cell-cell and cell-ECM adhesion complexes. Therapeutic aspects of mechanical stretching are reviewed considering these cellular responses after the application of mechanical forces, with a focus on wound healing and tissue regeneration. Impact Statement Mechanical stretching has been proposed as a therapeutic option for tissue regeneration and wound healing. It has been accepted that mechanotransduction processes elicited by mechanical stretching govern cellular response and behavior, and these studies have predominantly focused on the cell-extracellular matrix (ECM) sites. This review serves the mechanobiology community by shifting the focus of mechanical stretching effects from cell-ECM adhesions to the less examined cell-cell adhesions, which we believe play an equally important role in orchestrating the response pathways.
Collapse
Affiliation(s)
- Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Keerthana Reddy
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Tasneem Bouzid
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Brandon Riehl
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
26
|
Leroux R, Ringenbach C, Marchand T, Peschard O, Mondon P, Criton P. A new matrikine-derived peptide up-regulates longevity genes for improving extracellular matrix architecture and connections of dermal cell with its matrix. Int J Cosmet Sci 2019; 42:53-59. [PMID: 31596957 DOI: 10.1111/ics.12584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/30/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Skin extracellular matrix (ECM) is a dense and well-organized structure produced by fibroblasts. This ECM transduces environmental mechano-signals to cell nucleus through the integrin-actin complex, thus triggering ECM protein syntheses. The aim of this study was to discover a novel peptide, structurally related to dermal matrikines, that promotes syntheses of ECM components. METHODS AND RESULTS Screening tests with 120 peptides were carried out by using normal dermal human fibroblasts (HF). As a result, one candidate of interest was isolated, the N-Prolyl Palmitoyl Tripeptide-56 Acetate (PP56), which increases collagen and fibronectin productions at gene and/or protein levels. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), a recent and innovative analytical technology, in addition to more traditional techniques, it was showed that two metabolic pathways were significantly modulated: one for collagen production and one for actin. Moreover, this peptide up-regulated the transcription of Forkhead Box O (FOXO) and sestrin messenger RNAs (mRNA), leading to production of proteins involved into longevity and more recently in collagen production. RESULTS Results indicated that this peptide is a potential candidate to improve ECM density and organization in a new way.
Collapse
Affiliation(s)
- R Leroux
- SEDERMA, 29 rue du Chemin Vert, 78612, Le Perray-en-Yvelines cedex, France
| | - C Ringenbach
- SEDERMA, 29 rue du Chemin Vert, 78612, Le Perray-en-Yvelines cedex, France
| | - T Marchand
- SEDERMA, 29 rue du Chemin Vert, 78612, Le Perray-en-Yvelines cedex, France
| | - O Peschard
- SEDERMA, 29 rue du Chemin Vert, 78612, Le Perray-en-Yvelines cedex, France
| | - P Mondon
- SEDERMA, 29 rue du Chemin Vert, 78612, Le Perray-en-Yvelines cedex, France
| | - P Criton
- SEDERMA, 29 rue du Chemin Vert, 78612, Le Perray-en-Yvelines cedex, France
| |
Collapse
|
27
|
The Non-Fibrillar Side of Fibrosis: Contribution of the Basement Membrane, Proteoglycans, and Glycoproteins to Myocardial Fibrosis. J Cardiovasc Dev Dis 2019; 6:jcdd6040035. [PMID: 31547598 PMCID: PMC6956278 DOI: 10.3390/jcdd6040035] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) provides structural support and a microenvironmentfor soluble extracellular molecules. ECM is comprised of numerous proteins which can be broadly classified as fibrillar (collagen types I and III) and non-fibrillar (basement membrane, proteoglycans, and glycoproteins). The basement membrane provides an interface between the cardiomyocytes and the fibrillar ECM, while proteoglycans sequester soluble growth factors and cytokines. Myocardial fibrosis was originally only linked to accumulation of fibrillar collagens, but is now recognized as the expansion of the ECM including the non-fibrillar ECM proteins. Myocardial fibrosis can be reparative to replace the lost myocardium (e.g., ischemic injury or myocardial infarction), or can be reactive resulting from pathological activity of fibroblasts (e.g., dilated or hypertrophic cardiomyopathy). Contribution of fibrillar collagens to fibrosis is well studied, but the role of the non-fibrillar ECM proteins has remained less explored. In this article, we provide an overview of the contribution of the non-fibrillar components of the extracellular space of the heart to highlight the potential significance of these molecules in fibrosis, with direct evidence for some, although not all of these molecules in their direct contribution to fibrosis.
Collapse
|
28
|
Iwahashi H, Kawashima Y, Masaki H. Interleukin-1 alpha derived from ultraviolet B-exposed keratinocytes is associated with a decrease of endocytic collagen receptor Endo180. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2019; 36:34-41. [PMID: 31376337 DOI: 10.1111/phpp.12502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Endo180 contributes to the remodeling of the collagen fibers that comprise the dermal matrix due to the internalization of extracellular collagen fragments. In the sun-exposed elder skin, an accumulation of collagen fragments was observed in the dermal matrix which was associated with a reduction in Endo180 in the dermal fibroblasts. This suggests that the loss of Endo180 results in the accumulation of collagen fragments in the surrounding fibroblasts and causes interference with dermal matrix remodeling via collagen fibers. The purpose of the study was to identify a mechanism by which ultraviolet B (UVB) exposure induces a loss of Endo 180 with a specific focus on the crosstalk between keratinocytes and fibroblasts. METHODS Endo180 from normal human dermal fibroblasts, which were cultured with a conditioned medium (CM) of UVB-exposed keratinocytes, was examined using mRNA expression, protein levels and collagen internalization by quantitative RT-PCR, ELISA, and flow cytometry, respectively. RESULTS Although UVB irradiation to fibroblasts failed to reduce Endo180, the CM of UVB-exposed keratinocytes reduced Endo180 in the fibroblasts. Collagen internalization into the fibroblasts was decreased and was associated with a loss of Endo180. Among cytokines secreted from UVB-exposed keratinocytes, IL-1α solely reduced Endo180, and the reduction induced by the CM of UVB-exposed keratinocytes was abolished by the presence of IL-1RA. CONCLUSIONS These results indicate that a substance secreted from UVB-exposed keratinocytes regulates Endo180 expression and that IL-1α may play an important role in the maintenance of Endo180.
Collapse
Affiliation(s)
- Hiroyasu Iwahashi
- Research Center, Maruzen Pharmaceuticals Co., Ltd., Fukuyama, Hiroshima, Japan
| | - Yoshihito Kawashima
- Research Center, Maruzen Pharmaceuticals Co., Ltd., Fukuyama, Hiroshima, Japan
| | - Hitoshi Masaki
- Laboratory of photoaging Research, School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Tokyo, Japan
| |
Collapse
|
29
|
Cañedo-Dorantes L, Cañedo-Ayala M. Skin Acute Wound Healing: A Comprehensive Review. Int J Inflam 2019; 2019:3706315. [PMID: 31275545 PMCID: PMC6582859 DOI: 10.1155/2019/3706315] [Citation(s) in RCA: 270] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 04/22/2019] [Indexed: 02/07/2023] Open
Abstract
Experimental work of the last two decades has revealed the general steps of the wound healing process. This complex network has been organized in three sequential and overlapping steps. The first step of the inflammatory phase is an immediate response to injury; primary sensory neurons sense injury and send danger signals to the brain, to stop bleeding and start inflammation. The following target of the inflammatory phase, led by the peripheral blood mononuclear cells, is to eliminate the pathogens and clean the wound. Once this is completed, the inflammatory phase is resolved and homeostasis is restored. The aim of the proliferative phase, the second phase, is to repair wound damage and begin tissue remodeling. Fibroplasia, reepithelialization, angiogenesis, and peripheral nerve repair are the central actions of this phase. Lastly, the objective of the final phase is to complete tissue remodeling and restore skin integrity. This review provides present day information regarding the status of the participant cells, extracellular matrix, cytokines, chemokines, and growth factors, as well as their interactions with the microenvironment during the wound healing process.
Collapse
Affiliation(s)
- Luis Cañedo-Dorantes
- Research Division, Faculty of Medicine, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | | |
Collapse
|
30
|
Chu SY, Chou CH, Huang HD, Yen MH, Hong HC, Chao PH, Wang YH, Chen PY, Nian SX, Chen YR, Liou LY, Liu YC, Chen HM, Lin FM, Chang YT, Chen CC, Lee OK. Mechanical stretch induces hair regeneration through the alternative activation of macrophages. Nat Commun 2019; 10:1524. [PMID: 30944305 PMCID: PMC6447615 DOI: 10.1038/s41467-019-09402-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/04/2019] [Indexed: 12/28/2022] Open
Abstract
Tissues and cells in organism are continuously exposed to complex mechanical cues from the environment. Mechanical stimulations affect cell proliferation, differentiation, and migration, as well as determining tissue homeostasis and repair. By using a specially designed skin-stretching device, we discover that hair stem cells proliferate in response to stretch and hair regeneration occurs only when applying proper strain for an appropriate duration. A counterbalance between WNT and BMP-2 and the subsequent two-step mechanism are identified through molecular and genetic analyses. Macrophages are first recruited by chemokines produced by stretch and polarized to M2 phenotype. Growth factors such as HGF and IGF-1, released by M2 macrophages, then activate stem cells and facilitate hair regeneration. A hierarchical control system is revealed, from mechanical and chemical signals to cell behaviors and tissue responses, elucidating avenues of regenerative medicine and disease control by demonstrating the potential to manipulate cellular processes through simple mechanical stimulation.
Collapse
Affiliation(s)
- Szu-Ying Chu
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 112, Taiwan
- Department of Dermatology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chih-Hung Chou
- Department of Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, School of Life and Health Sciences, School of Sciences and Engineering, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Meng-Hua Yen
- Department of Electronic Engineering, National Chin-Yi University of Technology, Taichung, 411, Taiwan
| | - Hsiao-Chin Hong
- Department of Biological Science and Technology, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Po-Han Chao
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Yu-Hsuan Wang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Po-Yu Chen
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Dermatology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shi-Xin Nian
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Dermatology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Yu-Ru Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Li-Ying Liou
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Dermatology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Yu-Chen Liu
- Department of Biological Science and Technology, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Hui-Mei Chen
- Department of Biological Science and Technology, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Feng-Mao Lin
- Department of Biological Science and Technology, Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, 300, Taiwan
| | - Yun-Ting Chang
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Dermatology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Chih-Chiang Chen
- Department of Dermatology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
- Department of Dermatology, National Yang-Ming University, Taipei, 112, Taiwan.
| | - Oscar K Lee
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China.
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, China.
| |
Collapse
|
31
|
Abstract
The 4-and-a-half LIM domain protein 2 (FHL2) is a multifunctional adaptor protein that can interact with cell surface receptors, cytosolic adaptor and structural proteins, kinases, and nuclear transcription factors. It is involved in numerous functional activities, including the epithelial-mesenchymal transition, cell proliferation, apoptosis, adhesion, migration, structural stability, and gene expression. Despite this, FHL2-knockout (KO) mice are viable and fertile with no obvious abnormalities, rather suggesting a high capacity for fine-tuning adjustment and functional redundancy of FHL2. Indeed, challenging FHL2-KO cells or mice provided numerous evidences for the great functional significance of FHL2. In recent years, several reviews have been published describing the high capacity of FHL2 to bind diverse proteins as well as the versatile functions of FHL2, emphasizing in particular its role in cardiovascular diseases and carcinogenesis. Here, we view the function of FHL2 from a different perspective. We summarize the published data demonstrating the impact of FHL2 on wound healing and inflammation. FHL2 seems to be involved in numerous steps of these extremely complex and multidirectional but tightly regulated tissue remodeling processes, supporting tissue repair and coordinating inflammation. Deficiency of FHL2 not only slows down ongoing wound healing but also often turns it into a chronic condition.-Wixler, V. The role of FHL2 in wound healing and inflammation.
Collapse
Affiliation(s)
- Viktor Wixler
- Centre for Molecular Biology of Inflammation, Institute of Molecular Virology, Westfaelische Wilhelms University Muenster, Muenster, Germany
| |
Collapse
|
32
|
Tension enhances cell proliferation and collagen synthesis by upregulating expressions of integrin αvβ3 in human keloid-derived mesenchymal stem cells. Life Sci 2018; 219:272-282. [PMID: 30597173 DOI: 10.1016/j.lfs.2018.12.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/19/2018] [Accepted: 12/25/2018] [Indexed: 12/21/2022]
Abstract
AIMS Keloids are a dermal fibrotic disease whose etiology remains totally unknown and for which there is no successful treatment. Mechanical tension, in addition, is closely associated with the germination and development of keloids. In this study, we investigated the influence of human keloid-derived mesenchymal stem cells (KD-MSCs) on cell proliferation, collagen synthesis, and expressions of integrin αvβ3 under tension. MAIN METHODS KD-MSCs and human normal skin-derived mesenchymal stem cells (NS-MSCs) were isolated and cultured in stem cell medium with a gradual increase in the serum concentration. Cell proliferation and collagen synthesis were detected by Cell Counting Kit-8 (CCK-8) assay and hydroxyproline content analysis under tension respectively. We investigated the messenger RNA expressions of nine integrin subunits, including integrin units α2, α3, α5, αv, α8, α10, α11, β1, and β3, in KD-MSCs stimulated with tension. Identification of differentially expressed genes was performed by Western blot analysis and immunocytochemistry staining. KEY FINDINGS We obtained high-purity KD-MSCs and NS-MSCs using the culture method of decreasing serum concentration gradient gradually. Furthermore, we found that tension enhances cell proliferation and collagen synthesis and promotes expressions of integrin αvβ3 in KD-MSCs. In addition, blocking experiments showed that increased integrin αvβ3 expression affects cell proliferation and collagen synthesis of KD-MSCs under tension. SIGNIFICANCE Our results suggest that integrin αvβ3 receptor may be sensitive molecules of mechanical tension and could contribute to the occurrence and development of keloids. It could lead to novel targets for therapeutic intervention, treatment, and prevention of recurrence for keloid disorders.
Collapse
|
33
|
Thi Thanh Hai N, Thuy LTT, Shiota A, Kadono C, Daikoku A, Hoang DV, Dat NQ, Sato-Matsubara M, Yoshizato K, Kawada N. Selective overexpression of cytoglobin in stellate cells attenuates thioacetamide-induced liver fibrosis in mice. Sci Rep 2018; 8:17860. [PMID: 30552362 PMCID: PMC6294752 DOI: 10.1038/s41598-018-36215-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023] Open
Abstract
Cytoglobin (CYGB), discovered in hepatic stellate cells (HSCs), is known to possess a radical scavenger function, but its pathophysiological roles remain unclear. Here, for the first time, we generated a new transgenic (TG) mouse line in which both Cygb and mCherry reporter gene expression were under the control of the native Cygb gene promoter. We demonstrated that the expression of Cygb-mCherry was related to endogenous Cygb in adult tissues by tracing mCherry fluorescence together with DNA, mRNA, and protein analyses. Administration of a single dose (50 mg/kg) of thioacetamide (TAA) in Cygb-TG mice resulted in lower levels of alanine transaminase and oxidative stress than those in WT mice. After 10 weeks of TAA administration, Cygb-TG livers exhibited reduced neutrophil accumulation, cytokine expression and fibrosis but high levels of quiescent HSCs. Primary HSCs isolated from Cygb-TG mice (HSCCygb-TG) exhibited significantly decreased mRNA levels of α-smooth muscle actin (αSMA), collagen 1α1, and transforming growth factor β-3 after 4 days in culture relative to WT cells. HSCsCygb-TG were resistant to H2O2-induced αSMA expression. Thus, cell-specific overexpression of Cygb attenuates HSC activation and protects mice against TAA-induced liver fibrosis presumably by maintaining HSC quiescence. Cygb is a potential new target for antifibrotic approaches.
Collapse
Affiliation(s)
- Nguyen Thi Thanh Hai
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
- Department of Biochemistry, Hanoi Medical University, Hanoi, Vietnam
| | - Le Thi Thanh Thuy
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | | | - Chiho Kadono
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Atsuko Daikoku
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Dinh Viet Hoang
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Ninh Quoc Dat
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Misako Sato-Matsubara
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Katsutoshi Yoshizato
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
- PhoenixBio Co. Ltd., Hiroshima, Japan
- Endowed Laboratory of Synthetic Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Norifumi Kawada
- Departments of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan.
| |
Collapse
|
34
|
Foster DS, Jones RE, Ransom RC, Longaker MT, Norton JA. The evolving relationship of wound healing and tumor stroma. JCI Insight 2018; 3:99911. [PMID: 30232274 DOI: 10.1172/jci.insight.99911] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The stroma in solid tumors contains a variety of cellular phenotypes and signaling pathways associated with wound healing, leading to the concept that a tumor behaves as a wound that does not heal. Similarities between tumors and healing wounds include fibroblast recruitment and activation, extracellular matrix (ECM) component deposition, infiltration of immune cells, neovascularization, and cellular lineage plasticity. However, unlike a wound that heals, the edges of a tumor are constantly expanding. Cell migration occurs both inward and outward as the tumor proliferates and invades adjacent tissues, often disregarding organ boundaries. The focus of our review is cancer associated fibroblast (CAF) cellular heterogeneity and plasticity and the acellular matrix components that accompany these cells. We explore how similarities and differences between healing wounds and tumor stroma continue to evolve as research progresses, shedding light on possible therapeutic targets that can result in innovative stromal-based treatments for cancer.
Collapse
Affiliation(s)
- Deshka S Foster
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and.,Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - R Ellen Jones
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and
| | - Ryan C Ransom
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and.,Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jeffrey A Norton
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Division of Plastic and Reconstructive Surgery, and.,Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
35
|
Sonoki A, Okano Y, Yoshitake Y. Dermal fibroblasts can activate matrix metalloproteinase-1 independent of keratinocytes via plasmin in a 3D collagen model. Exp Dermatol 2018; 27:520-525. [PMID: 29498767 DOI: 10.1111/exd.13522] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2018] [Indexed: 11/30/2022]
Abstract
Photoaging of the skin is marked by obvious wrinkles and mainly depends on degradation of the extracellular matrix (ECM) in the dermis. Matrix metalloproteinase (MMP)-1 is one of the most important factors involved in degradation of the ECM; however, its mechanism of activation is not fully understood. It has been thought that MMP-1 is expressed by dermal fibroblasts as an inactive precursor protein that is activated by proteinases produced by keratinocytes in the epidermis. In this study, we constructed a 3D model of the dermis using collagen-embedded fibroblasts with or without ultraviolet (UV)-A exposure to mimic photoaging in the dermis. Collagen lattices embedded with UV-A-irradiated fibroblasts miniaturized and collagen was degraded to a greater extent than collagen lattices embedded with non-irradiated fibroblasts. The results demonstrate that fibroblasts in this 3D model express activated MMP-1 in the absence of keratinocytes. Moreover, the results confirm that activation of MMP-1 depends on increased plasmin activity in this model and lattice miniaturization was inhibited by the plasmin inhibitor tranexamic acid. Our results suggest that plasmin acts as an activator of MMP-1 and the inhibition of plasmin prevents collagen degradation.
Collapse
Affiliation(s)
- Aska Sonoki
- OPPEN COSMETICS CO., LTD., Kusatsu-shi, Shiga, Japan
| | - Yuri Okano
- School of Bioscience and Biotechnology, Tokyo University of Technology, Hachiouji, Tokyo, Japan
| | | |
Collapse
|
36
|
Moore AL, Marshall CD, Barnes LA, Murphy MP, Ransom RC, Longaker MT. Scarless wound healing: Transitioning from fetal research to regenerative healing. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:10.1002/wdev.309. [PMID: 29316315 PMCID: PMC6485243 DOI: 10.1002/wdev.309] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 09/07/2017] [Accepted: 10/27/2017] [Indexed: 01/08/2023]
Abstract
Since the discovery of scarless fetal skin wound healing, research in the field has expanded significantly with the hopes of advancing the finding to adult human patients. There are several differences between fetal and adult skin that have been exploited to facilitate scarless healing in adults including growth factors, cytokines, and extracellular matrix substitutes. However, no one therapy, pathway, or cell subtype is sufficient to support scarless wound healing in adult skin. More recently, products that contain or mimic fetal and adult uninjured dermis were introduced to the wound healing market with promising clinical outcomes. Through our review of the major experimental targets of fetal wound healing, we hope to encourage research in areas that may have a significant clinical impact. Additionally, we will investigate therapies currently in clinical use and evaluate whether they represent a legitimate advance in regenerative medicine or a vulnerary agent. WIREs Dev Biol 2018, 7:e309. doi: 10.1002/wdev.309 This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Plant Development > Cell Growth and Differentiation Adult Stem Cells, Tissue Renewal, and Regeneration > Environmental Control of Stem Cells.
Collapse
Affiliation(s)
- Alessandra L. Moore
- Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Clement D. Marshall
- Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Leandra A. Barnes
- Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Matthew P. Murphy
- Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Ryan C. Ransom
- Department of Surgery, Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Michael T. Longaker
- Department of Surgery, Stanford University School of Medicine, Stanford, California
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| |
Collapse
|
37
|
Barnes LA, Marshall CD, Leavitt T, Hu MS, Moore AL, Gonzalez JG, Longaker MT, Gurtner GC. Mechanical Forces in Cutaneous Wound Healing: Emerging Therapies to Minimize Scar Formation. Adv Wound Care (New Rochelle) 2018; 7:47-56. [PMID: 29392093 PMCID: PMC5792236 DOI: 10.1089/wound.2016.0709] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/15/2016] [Indexed: 12/25/2022] Open
Abstract
Significance: Excessive scarring is major clinical and financial burden in the United States. Improved therapies are necessary to reduce scarring, especially in patients affected by hypertrophic and keloid scars. Recent Advances: Advances in our understanding of mechanical forces in the wound environment enable us to target mechanical forces to minimize scar formation. Fetal wounds experience much lower resting stress when compared with adult wounds, and they heal without scars. Therapies that modulate mechanical forces in the wound environment are able to reduce scar size. Critical Issues: Increased mechanical stresses in the wound environment induce hypertrophic scarring via activation of mechanotransduction pathways. Mechanical stimulation modulates integrin, Wingless-type, protein kinase B, and focal adhesion kinase, resulting in cell proliferation and, ultimately, fibrosis. Therefore, the development of therapies that reduce mechanical forces in the wound environment would decrease the risk of developing excessive scars. Future Directions: The development of novel mechanotherapies is necessary to minimize scar formation and advance adult wound healing toward the scarless ideal. Mechanotransduction pathways are potential targets to reduce excessive scar formation, and thus, continued studies on therapies that utilize mechanical offloading and mechanomodulation are needed.
Collapse
Affiliation(s)
- Leandra A. Barnes
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Clement D. Marshall
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Tripp Leavitt
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael S. Hu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
- Department of Surgery, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | | | - Jennifer G. Gonzalez
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael T. Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
38
|
Kaufman G, Whitescarver RA, Nunes L, Palmer XL, Skrtic D, Tutak W. Effects of protein-coated nanofibers on conformation of gingival fibroblast spheroids: potential utility for connective tissue regeneration. ACTA ACUST UNITED AC 2018; 13:025006. [PMID: 29364821 DOI: 10.1088/1748-605x/aa91d9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Deep wounds in the gingiva caused by trauma or surgery require a rapid and robust healing of connective tissues. We propose utilizing gas-brushed nanofibers coated with collagen and fibrin for that purpose. Our hypotheses are that protein-coated nanofibers will: (i) attract and mobilize cells in various spatial orientations, and (ii) regulate the expression levels of specific extracellular matrix (ECM)-associated proteins, determining the initial conformational nature of dense and soft connective tissues. Gingival fibroblast monolayers and 3D spheroids were cultured on ECM substrate and covered with gas-blown poly-(DL-lactide-co-glycolide) (PLGA) nanofibers (uncoated/coated with collagen and fibrin). Cell attraction and rearrangement was followed by F-actin staining and confocal microscopy. Thicknesses of the cell layers, developed within the nanofibers, were quantified by ImageJ software. The expression of collagen1α1 chain (Col1α1), fibronectin, and metalloproteinase 2 (MMP2) encoding genes was determined by quantitative reverse transcription analysis. Collagen- and fibrin- coated nanofibers induced cell migration toward fibers and supported cellular growth within the scaffolds. Both proteins affected the spatial rearrangement of fibroblasts by favoring packed cell clusters or intermittent cell spreading. These cell arrangements resembled the structural characteristic of dense and soft connective tissues, respectively. Within three days of incubation, fibroblast spheroids interacted with the fibers, and grew robustly by increasing their thickness compared to monolayers. While the ECM key components, such as fibronectin and MMP2 encoding genes, were expressed in both protein groups, Col1α1 was predominantly expressed in bundled fibroblasts grown on collagen fibers. This enhanced expression of collagen1 is typical for dense connective tissue. Based on results of this study, our gas-blown, collagen- and fibrin-coated PLGA nanofibers are viable candidates for engineering soft and dense connective tissues with the required structural characteristics and functions needed for wound healing applications. Rapid regeneration of these layers should enhance healing of open wounds in a harsh oral environment.
Collapse
Affiliation(s)
- Gili Kaufman
- Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, United States of America
| | | | | | | | | | | |
Collapse
|
39
|
Kumar D, Yalamanchali S, New J, Parsel S, New N, Holcomb A, Gunewardena S, Tawfik O, Lominska C, Kimler BF, Anant S, Kakarala K, Tsue T, Shnayder Y, Sykes K, Padhye S, Thomas SM. Development and Characterization of an In Vitro Model for Radiation-Induced Fibrosis. Radiat Res 2018; 189:326-336. [PMID: 29351058 DOI: 10.1667/rr14926.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radiation-induced fibrosis (RIF) is a major side effect of radiotherapy in cancer patients with no effective therapeutic options. RIF involves excess deposition and aberrant remodeling of the extracellular matrix (ECM) leading to stiffness in tissues and organ failure. Development of preclinical models of RIF is crucial to elucidate the molecular mechanisms regulating fibrosis and to develop therapeutic approaches. In addition to radiation, the main molecular perpetrators of fibrotic reactions are cytokines, including transforming growth factor-β (TGF-β). We hypothesized that human oral fibroblasts would develop an in vitro fibrotic reaction in response to radiation and TGF-β. We demonstrate here that fibroblasts exposed to radiation followed by TGF-β exhibit a fibrotic phenotype with increased collagen deposition, cell proliferation, migration and invasion. In this in vitro model of RIF (RIFiv), the early biological processes involved in fibrosis are demonstrated, along with increased levels of several molecules including collagen 1α1, collagen XIα1, integrin-α2 and cyclin D1 mRNA in irradiated cells. A clinically relevant antifibrotic agent, pentoxifylline, and a curcumin analogue both mitigated collagen deposition in irradiated fibroblast cultures. In summary, we have established an in vitro model for RIF that facilitates the elucidation of molecular mechanisms in radiation-induced fibrosis and the development of effective therapeutic approaches.
Collapse
Affiliation(s)
- Dhruv Kumar
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Sreeya Yalamanchali
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Jacob New
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas.,b Department of Anatomy and Cell Biology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Sean Parsel
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Natalie New
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Andrew Holcomb
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Sumedha Gunewardena
- c Department of Molecular and Integrative Physiology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Ossama Tawfik
- d Department of Pathology and Laboratory Medicine, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Chris Lominska
- e Department of Radiation Oncology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Bruce F Kimler
- e Department of Radiation Oncology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Shrikant Anant
- f Department of Surgery, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas.,g Department of Cancer Biology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Kiran Kakarala
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Terance Tsue
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Yelizaveta Shnayder
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Kevin Sykes
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| | - Subhash Padhye
- h Interdisciplinary Science and Technology Research Academy, University of Pune, Pune, India
| | - Sufi Mary Thomas
- Department of a Otolaryngology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas.,b Department of Anatomy and Cell Biology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas.,g Department of Cancer Biology, University of Kansas Medical Center and University of Kansas Cancer Center, Kansas City, Kansas
| |
Collapse
|
40
|
Inducible knockdown of procollagen I protects mice from liver fibrosis and leads to dysregulated matrix genes and attenuated inflammation. Matrix Biol 2017; 66:34-49. [PMID: 29122677 DOI: 10.1016/j.matbio.2017.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/01/2017] [Accepted: 11/02/2017] [Indexed: 02/06/2023]
Abstract
Organ fibrosis is characterized by a chronic wound-healing response, with excess deposition of extracellular matrix components. Here, collagen type I represents the most abundant scar component and a primary target for antifibrotic therapies. Liver fibrosis can progress to cirrhosis and primary liver cancer, which are the major causes of liver related morbidity and mortality. However, a (pro-)collagen type I specific therapy remains difficult and its therapeutic abrogation may incur unwanted side effects. We therefore designed tetracycline-regulated procollagen alpha1(I) short hairpin (sh)RNA expressing mice that permit a highly efficient inducible knockdown of the procollagen alpha1(I) gene in activated (myo-)fibroblasts, to study the effect of induced procollagen type I deficiency. Transgenic mice were generated using recombinase-mediated integration in embryonic stem cells or zinc-finger nuclease-aided genomic targeting combined with miR30-shRNA technology. Liver fibrosis was induced in transgenic mice by carbon tetrachloride, either without or with doxycycline supplementation. Doxycycline treated mice showed an 80-90% suppression of procollagen alpha1(I) transcription and a 40-50% reduction in hepatic collagen accumulation. Procollagen alpha1(I) knockdown also downregulated procollagens type III, IV and VI and other fibrosis related parameters. Moreover, this was associated with an attenuation of chronic inflammation, suggesting that collagen type I serves not only as major scar component, but also as modulator of other collagens and promoter of chronic inflammation.
Collapse
|
41
|
Cha BH, Shin SR, Leijten J, Li YC, Singh S, Liu JC, Annabi N, Abdi R, Dokmeci MR, Vrana NE, Ghaemmaghami AM, Khademhosseini A. Integrin-Mediated Interactions Control Macrophage Polarization in 3D Hydrogels. Adv Healthc Mater 2017; 6:10.1002/adhm.201700289. [PMID: 28782184 PMCID: PMC5677560 DOI: 10.1002/adhm.201700289] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/17/2017] [Indexed: 12/23/2022]
Abstract
Adverse immune reactions prevent clinical translation of numerous implantable devices and materials. Although inflammation is an essential part of tissue regeneration, chronic inflammation ultimately leads to implant failure. In particular, macrophage polarity steers the microenvironment toward inflammation or wound healing via the induction of M1 and M2 macrophages, respectively. Here, this paper demonstrates that macrophage polarity within biomaterials can be controlled through integrin-mediated interactions between human monocytic THP-1 cells and collagen-derived matrix. Surface marker, gene expression, biochemical, and cytokine profiling consistently indicate that THP-1 cells within a biomaterial lacking cell attachment motifs yield proinflammatory M1 macrophages, whereas biomaterials with attachment sites in the presence of interleukin-4 (IL-4) induce an anti-inflammatory M2-like phenotype and propagate the effect of IL-4 in induction of M2-like macrophages. Importantly, integrin α2β1 plays a pivotal role as its inhibition blocks the induction of M2 macrophages. The influence of the microenvironment of the biomaterial over macrophage polarity is further confirmed by its ability to modulate the effect of IL-4 and lipopolysaccharide, which are potent inducers of M2 or M1 phenotypes, respectively. Thus, this study represents a novel, versatile, and effective strategy to steer macrophage polarity through integrin-mediated 3D microenvironment for biomaterial-based programming.
Collapse
Affiliation(s)
- Byung-Hyun Cha
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Su Ryon Shin
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500, AE, Enschede, The Netherlands
| | - Yi-Chen Li
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Sonali Singh
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Julie C Liu
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Davidson School of Chemical Engineering and Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Nasim Annabi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Reza Abdi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Transplant Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital, Boston, MA, 02115, USA
| | - Mehmet R Dokmeci
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Nihal Engin Vrana
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Fundamental Research Unit, Protip Medical, 8 Place de l'Hôpital, 67000, Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S 1121, "Biomatériaux et Bioingénierie", 11 rue Humann, 67085, Strasbourg Cedex, France
| | - Amir M Ghaemmaghami
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, 143-701, Republic of Korea
- Nanotechnology Center, King Abdulaziz University, Jeddah, 21569, Saudi Arabia
| |
Collapse
|
42
|
Kaisers W, Boukamp P, Stark HJ, Schwender H, Tigges J, Krutmann J, Schaal H. Age, gender and UV-exposition related effects on gene expression in in vivo aged short term cultivated human dermal fibroblasts. PLoS One 2017; 12:e0175657. [PMID: 28475575 PMCID: PMC5419556 DOI: 10.1371/journal.pone.0175657] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 03/29/2017] [Indexed: 12/15/2022] Open
Abstract
Ageing, the progressive functional decline of virtually all tissues, affects numerous living organisms. Main phenotypic alterations of human skin during the ageing process include reduced skin thickness and elasticity which are related to extracellular matrix proteins. Dermal fibroblasts, the main source of extracellular fibrillar proteins, exhibit complex alterations during in vivo ageing and any of these are likely to be accompanied or caused by changes in gene expression. We investigated gene expression of short term cultivated in vivo aged human dermal fibroblasts using RNA-seq. Therefore, fibroblast samples derived from unaffected skin were obtained from 30 human donors. The donors were grouped by gender and age (Young: 19 to 25 years, Middle: 36 to 45 years, Old: 60 to 66 years). Two samples were taken from each donor, one from a sun-exposed and one from a sun-unexposed site. In our data, no consistently changed gene expression associated with donor age can be asserted. Instead, highly correlated expression of a small number of genes associated with transforming growth factor beta signalling was observed. Also, known gene expression alterations of in vivo aged dermal fibroblasts seem to be non-detectable in cultured fibroblasts.
Collapse
Affiliation(s)
- Wolfgang Kaisers
- Center for Bioinformatics and Biostatistics, BMFZ, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Petra Boukamp
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Holger Schwender
- Center for Bioinformatics and Biostatistics, BMFZ, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
- Mathematical Institute, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Jean Krutmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Heiner Schaal
- Institut für Virologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
43
|
Ma Y, Iyer RP, Jung M, Czubryt MP, Lindsey ML. Cardiac Fibroblast Activation Post-Myocardial Infarction: Current Knowledge Gaps. Trends Pharmacol Sci 2017; 38:448-458. [PMID: 28365093 DOI: 10.1016/j.tips.2017.03.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 03/05/2017] [Accepted: 03/06/2017] [Indexed: 12/21/2022]
Abstract
In response to myocardial infarction (MI), the wound healing response of the left ventricle (LV) comprises overlapping inflammatory, proliferative, and maturation phases, and the cardiac fibroblast is a key cell type involved in each phase. It has recently been appreciated that, early post-MI, fibroblasts transform to a proinflammatory phenotype and secrete cytokines and chemokines as well as matrix metalloproteinases (MMPs). Later post-MI, fibroblasts are activated to anti-inflammatory and proreparative phenotypes and generate anti-inflammatory and proangiogenic factors and extracellular matrix (ECM) components that form the infarct scar. Additional studies are needed to systematically examine how fibroblast activation shifts over the timeframe of the MI response and how modulation at different activation stages could alter wound healing and LV remodeling in distinct ways. This review summarizes current fibroblast knowledge as the foundation for a discussion of existing knowledge gaps.
Collapse
Affiliation(s)
- Yonggang Ma
- Mississippi Center for Heart Research, Department of Biophysics and Physiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Rugmani Padmanabhan Iyer
- Mississippi Center for Heart Research, Department of Biophysics and Physiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mira Jung
- Mississippi Center for Heart Research, Department of Biophysics and Physiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael P Czubryt
- St Boniface Hospital Albrechtsen Research Centre Institute of Cardiovascular Sciences, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Merry L Lindsey
- Mississippi Center for Heart Research, Department of Biophysics and Physiology, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
44
|
Yamaba H, Haba M, Kunita M, Sakaida T, Tanaka H, Yashiro Y, Nakata S. Morphological change of skin fibroblasts induced by UV Irradiation is involved in photoaging. Exp Dermatol 2016; 25 Suppl 3:45-51. [DOI: 10.1111/exd.13084] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Hiroyuki Yamaba
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Manami Haba
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Mayumi Kunita
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Tsutomu Sakaida
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Hiroshi Tanaka
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Youichi Yashiro
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| | - Satoru Nakata
- Research Laboratories; Nippon Menard Cosmetic Co., Ltd.; Nagoya Japan
| |
Collapse
|
45
|
Manuyakorn W, Smart DE, Noto A, Bucchieri F, Haitchi HM, Holgate ST, Howarth PH, Davies DE. Mechanical Strain Causes Adaptive Change in Bronchial Fibroblasts Enhancing Profibrotic and Inflammatory Responses. PLoS One 2016; 11:e0153926. [PMID: 27101406 PMCID: PMC4839664 DOI: 10.1371/journal.pone.0153926] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/06/2016] [Indexed: 01/27/2023] Open
Abstract
Asthma is characterized by periodic episodes of bronchoconstriction and reversible airway obstruction; these symptoms are attributable to a number of factors including increased mass and reactivity of bronchial smooth muscle and extracellular matrix (ECM) in asthmatic airways. Literature has suggested changes in cell responses and signaling can be elicited via modulation of mechanical stress acting upon them, potentially affecting the microenvironment of the cell. In this study, we hypothesized that mechanical strain directly affects the (myo)fibroblast phenotype in asthma. Therefore, we characterized responses of bronchial fibroblasts, from 6 normal and 11 asthmatic non-smoking volunteers, exposed to cyclical mechanical strain using flexible silastic membranes. Samples were analyzed for proteoglycans, α-smooth muscle actin (αSMA), collagens I and III, matrix metalloproteinase (MMP) 2 & 9 and interleukin-8 (IL-8) by qRT-PCR, Western blot, zymography and ELISA. Mechanical strain caused a decrease in αSMA mRNA but no change in either αSMA protein or proteoglycan expression. In contrast the inflammatory mediator IL-8, MMPs and interstitial collagens were increased at both the transcriptional and protein level. The results demonstrate an adaptive response of bronchial fibroblasts to mechanical strain, irrespective of donor. The adaptation involves cytoskeletal rearrangement, matrix remodelling and inflammatory cytokine release. These results suggest that mechanical strain could contribute to disease progression in asthma by promoting inflammation and remodelling responses.
Collapse
Affiliation(s)
- Wiparat Manuyakorn
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | - David E. Smart
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
- * E-mail:
| | - Antonio Noto
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
- BIONEC Department, University of Palermo, Palermo, Italy
- Istituto Euro-Mediterraneo di Scienza e Tecnologia, IEMEST, Palermo, Italy
| | - Fabio Bucchieri
- BIONEC Department, University of Palermo, Palermo, Italy
- Istituto Euro-Mediterraneo di Scienza e Tecnologia, IEMEST, Palermo, Italy
| | - Hans Michael Haitchi
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
- National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Stephen T. Holgate
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
- National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Peter H. Howarth
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
- National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Donna E. Davies
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
- National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
46
|
Krishna CV, Singh J, Thangavel C, Rattan S. Role of microRNAs in gastrointestinal smooth muscle fibrosis and dysfunction: novel molecular perspectives on the pathophysiology and therapeutic targeting. Am J Physiol Gastrointest Liver Physiol 2016; 310:G449-59. [PMID: 26822916 PMCID: PMC4824177 DOI: 10.1152/ajpgi.00445.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/20/2016] [Indexed: 01/31/2023]
Abstract
MicroRNAs (miRNAs) belong to a group of short noncoding RNA molecules with important roles in cellular biology. miRNAs regulate gene expression by repressing translation or degrading the target mRNA. Recently, a growing body of evidence suggests that miRNAs are implicated in many diseases and could be potential biomarkers. Fibrosis and/smooth muscle (SM) dysfunction contributes to the morbidity and mortality associated with several diseases of the gastrointestinal tract (GIT). Currently available therapeutic modalities are unsuccessful in efficiently blocking or reversing fibrosis and/or SM dysfunction. Recent understanding of the role of miRNAs in signaling pathway of fibrogenesis and SM phenotype switch has provided a new insight into translational research. However, much is still unknown about the molecular targets and therapeutic potential of miRNAs in the GIT. This review discusses miRNA biology, pathophysiology of fibrosis, and aging- associated SM dysfunction in relation to the deregulation of miRNAs in the GIT. We also highlight the role of selected miRNAs associated with fibrosis and SM dysfunction-related diseases of the GIT.
Collapse
Affiliation(s)
| | - Jagmohan Singh
- 2Department of Medicine, Division of Gastroenterology & Hepatology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Chellappagounder Thangavel
- 3Department of Radiation Oncology, Sidney Kimmel Cancer Center (TC), Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Satish Rattan
- 2Department of Medicine, Division of Gastroenterology & Hepatology, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania; and
| |
Collapse
|
47
|
Jiang Y, Wang X, Li Y, Mu S, Zhou S, Liu Y, Zhang B. GGsTOP increases migration of human periodontal ligament cells in vitro via reactive oxygen species pathway. Mol Med Rep 2016; 13:3813-20. [PMID: 27035100 PMCID: PMC4838071 DOI: 10.3892/mmr.2016.5038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 03/07/2016] [Indexed: 01/01/2023] Open
Abstract
GGsTOP is a novel and selective inhibitor of gamma-glutamyl transferase (GGT), a cell-surface enzyme that has a key role in glutathione homeostasis and the maintenance of cellular reactive oxygen species (ROS). ROS are essential for wound healing. However, little is known about the molecular mechanisms underlying the inhibition of GGT by GGsTOP in human periodontal ligament cells (hPLCs). The present study assessed GGT expression in mouse periodontal ligament tissues, GGT activity in hPLCs, and the potential physiological effect of GGsTOP on hPLC migration. Immunohistochemical analysis confirmed that GGT was widely expressed in mouse periodontal ligament tissue. Treatment with GGsTOP was associated with greater proliferation and migration of hPLCs, and higher levels of cellular ROS compared with untreated hPLCs. However, the increase in intracellular ROS was attenuated in hPLCs co-cultured with the anti-oxidant N-acetylcysteine (NAC), a precursor of glutathione. The higher ROS levels associated with GGsTOP treatment were in parallel with increases in the levels of type I collagen and alpha smooth muscle actin, which was inhibited in hPLCs co-cultured with NAC. Thus, GGsTOP may promote hPLC migration and participate in the maintenance of the periodontal ligament apparatus via the ROS pathway.
Collapse
Affiliation(s)
- Ying Jiang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiang Wang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ying Li
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Sen Mu
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Shuang Zhou
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yi Liu
- College of Stomatology, Tongji University, Shanghai 200072, P.R. China
| | - Bin Zhang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
48
|
Roux SL, Borbely G, Słoniecka M, Backman LJ, Danielson P. Transforming Growth Factor Beta 1 Modulates the Functional Expression of the Neurokinin-1 Receptor in Human Keratocytes. Curr Eye Res 2015; 41:1035-1043. [PMID: 26673553 PMCID: PMC4989870 DOI: 10.3109/02713683.2015.1088954] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Purpose: Transforming growth factor beta 1 (TGF-β1) is a cytokine involved in a variety of processes, such as differentiation of fibroblasts into myofibroblasts. TGF-β1 has also been shown to delay the internalization of the neurokinin-1 receptor (NK-1 R) after its activation by its ligand, the neuropeptide substance P (SP). NK-1 R comprises two naturally occurring variants, a full-length and a truncated form, triggering different cellular responses. SP has been shown to affect important events in the cornea – such as stimulating epithelial cell proliferation – processes that are involved in corneal wound healing and thus in maintaining the transparency of the corneal stroma. An impaired signaling through NK-1 R could thus impact the visual quality. We hypothesize that TGF-β1 modulates the expression pattern of NK-1 R in human corneal stroma cells, keratocytes. The purpose of this study was to test that hypothesis. Methods: Cultures of primary keratocytes were set up with cells derived from healthy human corneas, obtained from donated transplantation graft leftovers, and characterized by immunocytochemistry and Western blot. Immunocytochemistry for TGF-β receptors and NK-1 R was performed. Gene expression was assessed with real-time polymerase chain reaction (qPCR). Results: Expression of TGF-β receptors was confirmed in keratocytes in vitro. Treating the cells with TGF-β1 significantly reduced the gene expression of NK-1 R. Furthermore, immunocytochemistry for NK-1 R demonstrated that it is specifically the expression of the full-length isotype of the receptor that is reduced after treatment with TGF-β1, which was also confirmed with qPCR using a specific probe for the full-length receptor. Conclusions: TGF-β1 down-regulates the gene expression of the full-length variant of NK-1 R in human keratocytes, which might impact its signaling pathway and thus explain the known delay in internalization after activation by SP seen with TGF-β1 treatment.
Collapse
Affiliation(s)
- Sandrine Le Roux
- a Department of Integrative Medical Biology , Umeå University , Umeå , Sweden
| | - Gabor Borbely
- a Department of Integrative Medical Biology , Umeå University , Umeå , Sweden
| | - Marta Słoniecka
- a Department of Integrative Medical Biology , Umeå University , Umeå , Sweden.,b Department of Clinical Sciences, Ophthalmology , Umeå University , Umeå , Sweden
| | - Ludvig J Backman
- a Department of Integrative Medical Biology , Umeå University , Umeå , Sweden
| | - Patrik Danielson
- a Department of Integrative Medical Biology , Umeå University , Umeå , Sweden.,b Department of Clinical Sciences, Ophthalmology , Umeå University , Umeå , Sweden
| |
Collapse
|
49
|
Cremers NAJ, Suttorp M, Gerritsen MM, Wong RJ, van Run-van Breda C, van Dam GM, Brouwer KM, Kuijpers-Jagtman AM, Carels CEL, Lundvig DMS, Wagener FADTG. Mechanical Stress Changes the Complex Interplay Between HO-1, Inflammation and Fibrosis, During Excisional Wound Repair. Front Med (Lausanne) 2015; 2:86. [PMID: 26697429 PMCID: PMC4678194 DOI: 10.3389/fmed.2015.00086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/24/2015] [Indexed: 01/11/2023] Open
Abstract
Mechanical stress following surgery or injury can promote pathological wound healing and fibrosis, and lead to functional loss and esthetic problems. Splinted excisional wounds can be used as a model for inducing mechanical stress. The cytoprotective enzyme heme oxygenase-1 (HO-1) is thought to orchestrate the defense against inflammatory and oxidative insults that drive fibrosis. Here, we investigated the activation of the HO-1 system in a splinted and non-splinted full-thickness excisional wound model using HO-1-luc transgenic mice. Effects of splinting on wound closure, HO-1 promoter activity, and markers of inflammation and fibrosis were assessed. After seven days, splinted wounds were more than three times larger than non-splinted wounds, demonstrating a delay in wound closure. HO-1 promoter activity rapidly decreased following removal of the (epi)dermis, but was induced in both splinted and non-splinted wounds during skin repair. Splinting induced more HO-1 gene expression in 7-day wounds; however, HO-1 protein expression remained lower in the epidermis, likely due to lower numbers of keratinocytes in the re-epithelialization tissue. Higher numbers of F4/80-positive macrophages, αSMA-positive myofibroblasts, and increased levels of the inflammatory genes IL-1β, TNF-α, and COX-2 were present in 7-day splinted wounds. Surprisingly, mRNA expression of newly formed collagen (type III) was lower in 7-day wounds after splinting, whereas, VEGF and MMP-9 were increased. In summary, these data demonstrate that splinting delays cutaneous wound closure and HO-1 protein induction. The pro-inflammatory environment following splinting may facilitate higher myofibroblast numbers and increase the risk of fibrosis and scar formation. Therefore, inducing HO-1 activity against mechanical stress-induced inflammation and fibrosis may be an interesting strategy to prevent negative effects of surgery on growth and function in patients with orofacial clefts or in patients with burns.
Collapse
Affiliation(s)
- Niels A J Cremers
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands ; Experimental Rheumatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Maarten Suttorp
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Marlous M Gerritsen
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine , Stanford, CA , USA
| | - Coby van Run-van Breda
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen , Groningen , Netherlands
| | - Katrien M Brouwer
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, MOVE Research Institute Amsterdam , Amsterdam , Netherlands ; Association of Dutch Burn Centers , Beverwijk , Netherlands
| | - Anne Marie Kuijpers-Jagtman
- Department of Orthodontics and Craniofacial Biology, Cleft Palate Craniofacial Center, Radboud University Medical Center , Nijmegen , Netherlands
| | - Carine E L Carels
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Ditte M S Lundvig
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| | - Frank A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , Netherlands
| |
Collapse
|
50
|
Dupont S. Role of YAP/TAZ in cell-matrix adhesion-mediated signalling and mechanotransduction. Exp Cell Res 2015; 343:42-53. [PMID: 26524510 DOI: 10.1016/j.yexcr.2015.10.034] [Citation(s) in RCA: 347] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 10/29/2015] [Indexed: 12/12/2022]
Abstract
Signalling from the extracellular matrix (ECM) is a fundamental cellular input that sustains proliferation, opposes cell death and regulates differentiation. Through integrins, cells perceive both the chemical composition and physical properties of the ECM. In particular, cell behaviour is profoundly influenced by the mechanical elasticity or stiffness of the ECM, which regulates the ability of cells to develop forces through their contractile actomyosin cytoskeleton and to mature focal adhesions. This mechanosensing ability affects fundamental cellular functions, such that alterations of ECM stiffness is nowadays considered not a simple consequence of pathology, but a causative input driving aberrant cell behaviours. We here discuss recent advances on how mechanical signals intersect nuclear transcription and in particular the activity of YAP/TAZ transcriptional coactivators, known downstream transducers of the Hippo pathway and important effectors of ECM mechanical cues.
Collapse
Affiliation(s)
- Sirio Dupont
- Department of Molecular Medicine, University of Padua Medical School, via Bassi 58/B, 35131 Padua, Italy.
| |
Collapse
|