1
|
Kuhn PM, Chen S, Venkatraman A, Abadir PM, Walston JD, Kokkoli E. Co-Delivery of Valsartan and Metformin from a Thermosensitive Hydrogel-Nanoparticle System Promotes Collagen Production in Proliferating and Senescent Primary Human Dermal Fibroblasts. Biomacromolecules 2024; 25:5702-5717. [PMID: 39186039 DOI: 10.1021/acs.biomac.3c01461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Aging negatively impacts skin health, notably through the senescent cell phenotype, which reduces collagen production and leads to thinner, more fragile skin prone to injuries and chronic wounds. We designed a drug delivery system that addresses these age-related issues using a hybrid hydrogel-nanoparticle system that utilizes a poly(δ-valerolactone-co-lactide)-b-poly(ethylene-glycol)-b-poly(δ-valerolactone-co-lactide) (PVLA-PEG-PVLA) hydrogel. This hydrogel allows for the local, extended release of therapeutics targeting both proliferating and senescent cells. The PVLA-PEG-PVLA hydrogel entrapped valsartan, and metformin-loaded liposomes functionalized with a fibronectin-mimetic peptide, PR_b. Metformin acts as a senomorphic, reversing aspects of cellular senescence, and valsartan, an angiotensin receptor blocker, promotes collagen production. This combination treatment partially reversed the senescent phenotype and improved collagen production in senescent dermal fibroblasts from both young and old adults. Our codelivery hydrogel-nanoparticle system offers a promising treatment for improving age-related dermal pathologies.
Collapse
Affiliation(s)
- Paul M Kuhn
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Siwei Chen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Aditya Venkatraman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Peter M Abadir
- Division of Geriatrics and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, United States
| | - Jeremy D Walston
- Division of Geriatrics and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, United States
| | - Efrosini Kokkoli
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
2
|
Cross-Linked Gamma Polyglutamic Acid/Human Hair Keratin Electrospun Nanofibrous Scaffolds with Excellent Biocompatibility and Biodegradability. Polymers (Basel) 2022; 14:polym14245505. [PMID: 36559871 PMCID: PMC9781754 DOI: 10.3390/polym14245505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Recently, human hair keratin has been widely studied and applied in clinical fields due to its good histocompatibility, biocompatibility, and biodegradability. However, the regenerated keratin from human hair cannot be electrospun alone because of its low molecular weight. Herein, gamma polyglutamic acid (γ-PGA) was first selected to fabricate smooth and uniform γ-PGA/keratin composite scaffolds with excellent biocompatibility and biodegradability by electrospinning technology and a chemical cross-linking method in this study. The effect of electrospinning parameters on the structure and morphology, the mechanism of chemical cross-linking, biocompatibility in vitro cell culture experiments, and biodegradability in phosphate-buffered saline buffer solution and trypsin solution of the γ-PGA/keratin electrospun nanofibrous scaffolds (ENS) was studied. The results show that the cross-linked γ-PGA/keratin ENSs had excellent water stability and biodegradability. The γ-PGA/keratin ENSs showed better biocompatibility in promoting cell adhesion and cell growth compared with the γ-PGA ENSs. It is expected that γ-PGA/keratin ENSs will be easily and significantly used in tissue engineering to repair or regenerate materials.
Collapse
|
3
|
Jung SM, Park KS, Kim KJ. Integrative analysis of lung molecular signatures reveals key drivers of systemic sclerosis-associated interstitial lung disease. Ann Rheum Dis 2022; 81:108-116. [PMID: 34380701 DOI: 10.1136/annrheumdis-2021-220493] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/25/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Interstitial lung disease is a significant comorbidity and the leading cause of mortality in patients with systemic sclerosis. Transcriptomic data of systemic sclerosis-associated interstitial lung disease (SSc-ILD) were analysed to evaluate the salient molecular and cellular signatures in comparison with those in related pulmonary diseases and to identify the key driver genes and target molecules in the disease module. METHODS A transcriptomic dataset of lung tissues from patients with SSc-ILD (n=52), idiopathic pulmonary fibrosis (IPF) (n=549), non-specific interstitial pneumonia (n=49) and pulmonary arterial hypertension (n=81) and from normal healthy controls (n=331) was subjected to filtration of differentially expressed genes, functional enrichment analysis, network-based key driver analysis and kernel-based diffusion scoring. The association of enriched pathways with clinical parameters was evaluated in patients with SSc-ILD. RESULTS SSc-ILD shared key pathogenic pathways with other fibrosing pulmonary diseases but was distinguishable in some pathological processes. SSc-ILD showed general similarity with IPF in molecular and cellular signatures but stronger signals for myofibroblasts, which in SSc-ILD were in a senescent and apoptosis-resistant state. The p53 signalling pathway was the most enriched signature in lung tissues and lung fibroblasts of SSc-ILD, and was significantly correlated with carbon monoxide diffusing capacity of lung, cellular senescence and apoptosis. EEF2, EFF2K, PHKG2, VCAM1, PRKACB, ITGA4, CDK1, CDK2, FN1 and HDAC1 were key regulators with high diffusion scores in the disease module. CONCLUSIONS Integrative transcriptomic analysis of lung tissues revealed key signatures of fibrosis in SSc-ILD. A network-based Bayesian approach provides deep insights into key regulatory genes and molecular targets applicable to treating SSc-ILD.
Collapse
Affiliation(s)
- Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyung-Su Park
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ki-Jo Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
4
|
Mathur A, Kharbanda OP, Koul V, Dinda AK, Anwar MF, Singh S. Fabrication and evaluation of antimicrobial biomimetic nanofibre coating for improved dental implant bio-seal: An in vitro study. J Periodontol 2021; 93:1578-1588. [PMID: 34855256 DOI: 10.1002/jper.21-0255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/26/2021] [Accepted: 09/26/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND A weak implant-soft tissue interface may lead to bacterial ingression, breakdown of underlying tissues, and eventually implant failure. This study proposes a surface modification technique of titanium alloy (Ti), using a nano-biopolymer scaffold to enhance soft tissue attachment in dental implants. METHODS Gelatin (20% w/v) embedded with 10±2 nm silver nanoparticles (AgNPs) was electrospun to form a Gelatin Electrospun Mat (GEM) scaffold, bonded to Ti alloy surface using chemical surface functionalization. Antimicrobial activity of AgNPs was tested against representative Gram-positive (S. aureus) and Gram-negative bacteria (E. coli) at 4, 24, and 48 hours (h) and after embedding in scaffold at 48 h. Cytotoxicity analysis (MTT assay) was carried out using the 3T3 mouse fibroblast cell line at 24 and 72 h for two groups: Control (unmodified Ti disc) and Experimental (GEM embedded with AgNPs); and further validated by scanning electron microscopy (SEM). RESULTS The AgNPs-embedded GEM showed good antimicrobial activity at 48 h, with the AgNPs showing complete (99.99%) inhibition of bacterial colony counts at 24 h and 48 h. Cell viability and proliferation over the GEM modified Ti discs were seen to be significantly increased (p < 0.05) at 72 h as compared to control. SEM images revealed intimate spreading of fibroblasts, with differentiated cell morphology and pseudopodial processes, indicative of enhanced fibroblastic adhesion, growth, and differentiation over the scaffold. CONCLUSION Results show good antifouling properties and biocompatibility of the fabricated coating, making it a promising strategy to reduce post-operative infections and peri-implant diseases in Ti dental implants. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Apoorva Mathur
- Centre for Dental Education and Research (CDER, ), All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Om Prakash Kharbanda
- Dr. CG Pandit, National Chair of ICMR, Room 206, Department of Plastic Surgery, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Veena Koul
- PhD Professor Emeritus, Centre for Biomedical Engineering, IIT Delhi, India
| | - Amit Kumar Dinda
- ICMR Emeritus Scientist, Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | | - Suchita Singh
- Scientist C (Clinical Operations), Div. of ECD, ITRC, Indian Council for Medical Research, New Delhi, India
| |
Collapse
|
5
|
Keratin-Alginate Sponges Support Healing of Partial-Thickness Burns. Int J Mol Sci 2021; 22:ijms22168594. [PMID: 34445299 PMCID: PMC8395243 DOI: 10.3390/ijms22168594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/27/2021] [Accepted: 08/07/2021] [Indexed: 01/07/2023] Open
Abstract
Deep partial-thickness burns damage most of the dermis and can cause severe pain, scarring, and mortality if left untreated. This study serves to evaluate the effectiveness of crosslinked keratin–alginate composite sponges as dermal substitutes for deep partial-thickness burns. Crosslinked keratin–alginate sponges were tested for the ability to support human dermal fibroblasts in vitro and to support the closure and healing of partial-thickness burn wounds in Sus scrofa pigs. Keratin–alginate composite sponges supported the enhanced proliferation of human dermal fibroblasts compared to alginate-only sponges and exhibited decreased contraction in vitro when compared to keratin only sponges. As dermal substitutes in vivo, the sponges supported the expression of keratin 14, alpha-smooth muscle actin, and collagen IV within wound sites, comparable to collagen sponges. Keratin–alginate composite sponges supported the regeneration of basement membranes in the wounds more than in collagen-treated wounds and non-grafted controls, suggesting the subsequent development of pathological scar tissues may be minimized. Results from this study indicate that crosslinked keratin–alginate sponges are suitable alternative dermal substitutes for clinical applications in wound healing and skin regeneration.
Collapse
|
6
|
Gerbo ML, Powers RL. Case report of
natalizumab‐related
ocular cicatricial pemphigoid. Dermatol Ther 2021. [DOI: 10.1111/dth.14620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Michael L. Gerbo
- West Virginia University School of Medicine Morgantown West Virginia USA
| | - Roxann L. Powers
- West Virginia University School of Medicine Morgantown West Virginia USA
| |
Collapse
|
7
|
DiPersio CM, Van De Water L. Integrin Regulation of CAF Differentiation and Function. Cancers (Basel) 2019; 11:cancers11050715. [PMID: 31137641 PMCID: PMC6563118 DOI: 10.3390/cancers11050715] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/14/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
Extensive remodeling of the extracellular matrix, together with paracrine communication between tumor cells and stromal cells, contribute to an “activated” tumor microenvironment that supports malignant growth and progression. These stromal cells include inflammatory cells, endothelial cells, and cancer-associated fibroblasts (CAFs). Integrins are expressed on all tumor and stromal cell types where they regulate both cell adhesion and bidirectional signal transduction across the cell membrane. In this capacity, integrins control pro-tumorigenic cell autonomous functions such as growth and survival, as well as paracrine crosstalk between tumor cells and stromal cells. The myofibroblast-like properties of cancer-associated fibroblasts (CAFs), such as robust contractility and extracellular matrix (ECM) deposition, allow them to generate both chemical and mechanical signals that support invasive tumor growth. In this review, we discuss the roles of integrins in regulating the ability of CAFs to generate and respond to extracellular cues in the tumor microenvironment. Since functions of specific integrins in CAFs are only beginning to emerge, we take advantage of a more extensive literature on how integrins regulate wound myofibroblast differentiation and function, as some of these integrin functions are likely to extrapolate to CAFs within the tumor microenvironment. In addition, we discuss the roles that integrins play in controlling paracrine signals that emanate from epithelial/tumor cells to stimulate fibroblasts/CAFs.
Collapse
|
8
|
Klingberg F, Chau G, Walraven M, Boo S, Koehler A, Chow ML, Olsen AL, Im M, Lodyga M, Wells RG, White ES, Hinz B. The fibronectin ED-A domain enhances recruitment of latent TGF-β-binding protein-1 to the fibroblast matrix. J Cell Sci 2018; 131:jcs201293. [PMID: 29361522 PMCID: PMC5897715 DOI: 10.1242/jcs.201293] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/09/2018] [Indexed: 12/18/2022] Open
Abstract
Dysregulated secretion and extracellular activation of TGF-β1 stimulates myofibroblasts to accumulate disordered and stiff extracellular matrix (ECM) leading to fibrosis. Fibronectin immobilizes latent TGF-β-binding protein-1 (LTBP-1) and thus stores TGF-β1 in the ECM. Because the ED-A fibronectin splice variant is prominently expressed during fibrosis and supports myofibroblast activation, we investigated whether ED-A promotes LTBP-1-fibronectin interactions. Using stiffness-tuneable substrates for human dermal fibroblast cultures, we showed that high ECM stiffness promotes expression and colocalization of LTBP-1 and ED-A-containing fibronectin. When rescuing fibronectin-depleted fibroblasts with specific fibronectin splice variants, LTBP-1 bound more efficiently to ED-A-containing fibronectin than to ED-B-containing fibronectin and fibronectin lacking splice domains. Function blocking of the ED-A domain using antibodies and competitive peptides resulted in reduced LTBP-1 binding to ED-A-containing fibronectin, reduced LTBP-1 incorporation into the fibroblast ECM and reduced TGF-β1 activation. Similar results were obtained by blocking the heparin-binding stretch FNIII12-13-14 (HepII), adjacent to the ED-A domain in fibronectin. Collectively, our results suggest that the ED-A domain enhances association of the latent TGF-β1 by promoting weak direct binding to LTBP-1 and by enhancing heparin-mediated protein interactions through HepII in fibronectin.
Collapse
Affiliation(s)
- Franco Klingberg
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Grace Chau
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Marielle Walraven
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Stellar Boo
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Anne Koehler
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Melissa L Chow
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Abby L Olsen
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., BRB, Philadelphia, PA 19104, USA
| | - Michelle Im
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Monika Lodyga
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| | - Rebecca G Wells
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 421 Curie Blvd., BRB, Philadelphia, PA 19104, USA
| | - Eric S White
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, 150 College St., FG234, ON M5S3E2, Canada
| |
Collapse
|
9
|
Li X, Qian H, Ono F, Tsuchisaka A, Krol RP, Ohara K, Hayakawa T, Matsueda S, Sasada T, Ohata C, Furumura M, Hamada T, Hashimoto T. Human dermal fibroblast migration induced by fibronectin in autocrine and paracrine manners. Exp Dermatol 2016; 23:682-4. [PMID: 24828603 DOI: 10.1111/exd.12447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2014] [Indexed: 11/27/2022]
Abstract
Although fibronectin (FN) is known as a chemoattractant for human dermal fibroblasts (HDFs), it is unclear whether HDF migration is stimulated by FN produced by HDFs (autocrine manner) or by keratinocytes (paracrine manner). In this study, we investigated HDF migration by Boyden chamber assay using conditioned media from HDFs and HaCaT cells (keratinocyte cell line). Immunoblotting and enzyme-linked immunosorbent assay revealed that FN existed in both conditioned media. Boyden chamber assay showed both conditioned media stimulated HDF migration, which was inhibited by anti-FN antibody. Antibodies to both integrin β1and β3 subunits inhibited HDF migration induced by HDF-conditioned medium almost completely and that by HaCaT cell-conditioned medium with 50-60%. These results suggested that HDF migration was stimulated by FN in both autocrine and paracrine manners. However, the mechanisms of HDF migration by FN, particularly the role of integrin β1 and β3 subunits, were slightly different between autocrine and paracrine manners.
Collapse
Affiliation(s)
- Xiaoguang Li
- Department of Dermatology, Kurume University School of Medicine, and Kurume University Institute of Cutaneous Cell Biology, Kurume, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kufaishi H, Alarab M, Drutz H, Lye S, Shynlova O. Comparative Characterization of Vaginal Cells Derived From Premenopausal Women With and Without Severe Pelvic Organ Prolapse. Reprod Sci 2016; 23:931-43. [PMID: 26763525 DOI: 10.1177/1933719115625840] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND This study tested a hypothesis that primary human vaginal cells derived from tissue of premenopausal women with severe pelvic organ prolapse (POP-HVCs) would display differential functional characteristics as compared to vaginal cells derived from asymptomatic women with normal pelvic floor support (control-HVCs). METHODS Vaginal tissue biopsies were collected from premenopausal patients with POP (n = 8) and asymptomatic controls (n = 7) during vaginal hysterectomy or repair. Primary vaginal cells were isolated by enzymatic digestion and characterized by immunocytochemistry. Cell attachment and proliferation on different matrices (collagen I, collagen II, collagen IV, fibronectin, laminin, tenascin, and vitronectin) were compared between POP-HVCs and control-HVCs. RNA was extracted, and the expression of 84 genes was screened using Human Extracellular Matrix and Adhesion Molecules RT(2) Profiler PCR array. The expression of selected genes was verified by quantitative reverse transcription-polymerase chain reaction. RESULTS (1) Control-HVCs attached to collagen IV more efficiently than POP-HVCs; (2) control-HVCs and POP-HVCs show a similar proliferation rate when plated on proNectin and collagen I; (3) when seeded on collagen I, resting POP-HVCs expressed significantly (P < .05) increased transcript levels of collagen VII, multiple matrix metalloproteinases (MMP3, MMP7, MMP10, MMP12, MMP13, and MMP14), integrins (ITGA1, ITGA4, ITGA6, ITGA8, ITGB1, ITGB2, and ITGB3), and cell adhesion molecules as compared to control-HVCs. Collagen XV and tissue inhibitors of MMPs (TIMP1 and TIMP2) as well as genes involved in the biogenesis and maturation of collagen and elastin fibers (LOX, LOXL1-LOXL3, BMP1, and ADAMTS2) were significantly downregulated in POP-HVCs versus control-HVCs (P < .05). CONCLUSIONS Resting primary POP-HVCs in vitro show altered cellular characteristics as compared to control-HVCs, which may influence their dynamic responses to external mechanical or hormonal stimuli.
Collapse
Affiliation(s)
- Hala Kufaishi
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - May Alarab
- Division of Urogynecology and Reconstructive Pelvic Surgery, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Toronto, Ontario, Canada Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| | - Harold Drutz
- Division of Urogynecology and Reconstructive Pelvic Surgery, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Toronto, Ontario, Canada Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| | - Stephen Lye
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| | - Oksana Shynlova
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Hahn JM, Glaser K, McFarland KL, Aronow BJ, Boyce ST, Supp DM. Keloid-derived keratinocytes exhibit an abnormal gene expression profile consistent with a distinct causal role in keloid pathology. Wound Repair Regen 2014; 21:530-44. [PMID: 23815228 DOI: 10.1111/wrr.12060] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 03/18/2013] [Indexed: 12/17/2022]
Abstract
Keloids are disfiguring scars that extend beyond the original wound borders and resist treatment. Keloids exhibit excessive extracellular matrix deposition, although the underlying mechanisms remain unclear. To better understand the molecular basis of keloid scarring, here we define the genomic profiles of keloid fibroblasts and keratinocytes. In both cell types, keloid-derived cells exhibit differential expression of genes encompassing a diverse set of functional categories. Strikingly, keloid keratinocytes exhibited decreased expression of a set of transcription factor, cell adhesion, and intermediate filament genes essential for normal epidermal morphology. Conversely, they exhibit elevated expression of genes associated with wound healing, cellular motility, and vascular development. A substantial number of genes involved in epithelial-mesenchymal transition were also up-regulated in keloid keratinocytes, implicating this process in keloid pathology. Furthermore, keloid keratinocytes displayed significantly higher migration rates than normal keratinocytes in vitro and reduced expression of desmosomal proteins in vivo. Previous studies suggested that keratinocytes contribute to keloid scarring by regulating extracellular matrix production in fibroblasts. Our current results show fundamental abnormalities in keloid keratinocytes, suggesting they have a profoundly more direct role in keloid scarring than previously appreciated. Therefore, development of novel therapies should target both fibroblast and keratinocyte populations for increased efficacy.
Collapse
Affiliation(s)
- Jennifer M Hahn
- Research Department, Shriners Hospitals for Children-Cincinnati, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | |
Collapse
|
12
|
Sow WT, Lui YS, Ng KW. Electrospun human keratin matrices as templates for tissue regeneration. Nanomedicine (Lond) 2013; 8:531-41. [DOI: 10.2217/nnm.13.38] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aim: The aim of this work was to study the feasibility of fabricating human hair keratin matrices through electrospinning and to evaluate the potential of these matrices for tissue regeneration. Materials & methods: Keratin was extracted from human hair using Na2S and blended with poly(ethylene oxide) in the weight ratio of 60:1 for electrospinning. Physical morphology and chemical properties of the matrices were characterized using scanning electron microscopy and Fourier transform infrared spectroscopy, respectively. Cell viability and morphology of murine and human fibroblasts cultured on the matrices were evaluated through the Live/Dead® assay and scanning electron microscopy. Results: Electrospun keratin matrices were successfully produced without affecting the chemical conformation of keratin. Fibroblasts cultured on keratin matrices showed healthy morphology and penetration into matrices at day 7. Conclusion: Electrospun human hair keratin matrices provide a bioinductive and structural environment for cell growth and are thus attractive as alternative templates for tissue regeneration.
Collapse
Affiliation(s)
- Wan Ting Sow
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Yuan Siang Lui
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
- Institute for Sports Research, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Kee Woei Ng
- School of Materials Science & Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore.
| |
Collapse
|
13
|
Kaposi's sarcoma-associated herpesvirus forms a multimolecular complex of integrins (alphaVbeta5, alphaVbeta3, and alpha3beta1) and CD98-xCT during infection of human dermal microvascular endothelial cells, and CD98-xCT is essential for the postentry stage of infection. J Virol 2008; 82:12126-44. [PMID: 18829766 DOI: 10.1128/jvi.01146-08] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) interacts with cell surface heparan sulfate (HS) and alpha3beta1 integrin during the early stages of infection of human dermal microvascular endothelial cells (HMVEC-d) and human foreskin fibroblasts (HFF), and these interactions are followed by virus entry overlapping with the induction of preexisting host cell signal pathways. KSHV also utilizes the amino acid transporter protein xCT for infection of adherent cells, and the xCT molecule is part of the cell surface heterodimeric membrane glycoprotein CD98 (4F2 antigen) complex known to interact with alpha3beta1 and alphaVbeta3 integrins. KSHV gB mediates adhesion of HMVEC-d, CV-1, and HT-1080 cells and HFF via its RGD sequence. Anti-alphaV and -beta1 integrin antibodies inhibited the cell adhesion mediated by KSHV-gB. Variable levels of neutralization of HMVEC-d and HFF infection were observed with antibodies against alphaVbeta3 and alphaVbeta5 integrins. Similarly, variable levels of inhibition of virus entry into adherent HMVEC-d, 293 and Vero cells, and HFF was observed by preincubating virus with soluble alpha3beta1, alphaVbeta3, and alphaVbeta5 integrins, and cumulative inhibition was observed with a combination of integrins. We were unable to infect HT1080 cells. Virus binding and DNA internalization studies suggest that alphaVbeta3 and alphaVbeta5 integrins also play roles in KSHV entry. We observed time-dependent temporal KSHV interactions with HMVEC-d integrins and CD98/xCT with three different patterns of association and dissociation. Integrin alphaVbeta5 interaction with CD98/xCT predominantly occurred by 1 min postinfection (p.i.) and dissociated at 10 min p.i., whereas alpha3beta1-CD98/xCT interaction was maximal at 10 min p.i. and dissociated at 30 min p.i., and alphaVbeta3-CD98/xCT interaction was maximal at 10 min p.i. and remained at the observed 30 min p.i. Fluorescence microscopy also showed a similar time-dependent interaction of alphaVbeta5-CD98. Confocal-microscopy studies confirmed the association of CD98/xCT with alpha3beta1 and KSHV. Preincubation of KSHV with soluble heparin and alpha3beta1 significantly inhibited this association, suggesting that the first contact with HS and integrin is an essential element in subsequent CD98-xCT interactions. Anti-CD98 and xCT antibodies did not block virus binding and entry and nuclear delivery of viral DNA; however, viral-gene expression was significantly inhibited, suggesting that CD98-xCT play roles in the post-entry stage of infection, possibly in mediating signal cascades essential for viral-gene expression. Together, these studies suggest that KSHV interacts with functionally related integrins (alphaVbeta3, alpha3beta1, and alphaVbeta5) and CD98/xCT molecules in a temporal fashion to form a multimolecular complex during the early stages of endothelial cell infection, probably mediating multiple roles in entry, signal transduction, and viral-gene expression.
Collapse
|
14
|
Rodenberg EJ, Pavalko FM. Peptides Derived from Fibronectin Type III Connecting Segments Promote Endothelial Cell Adhesion but Not Platelet Adhesion: Implications in Tissue-Engineered Vascular Grafts. ACTA ACUST UNITED AC 2007; 13:2653-66. [PMID: 17883325 DOI: 10.1089/ten.2007.0037] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The development of a completely tissue-engineered small-caliber prosthesis suitable for incorporation into an in vivo vascular network is fraught with many challenges, including overcoming resistance to endothelialization and susceptibility to thrombogenesis. In this work, recombinant human fibronectin-derived low-molecular-weight peptide fragments were studied for their ability to promote cell type-specific alpha(4) integrin-mediated adhesion. Two populations of primary human endothelial cells were examined and found to express alpha(4) integrin receptors on their surfaces; on the contrary, human platelets were not found to be expressers of alpha(4) integrins. A peptide fragment isolated from the variably spliced human fibronectin type III connecting segment-1 (CS-1) domain was determined to mediate statistically significant endothelial cell alpha(4) integrin-mediated adhesion. In contrast, the fibronectin type III CS-1 fragment did not support human platelet adhesion under physiological fluid shear conditions, although fully intact human fibronectin molecules supported shear-induced platelet adhesion. This suggests that platelets bind to fibronectin in regions not encompassing the CS-1 domain. In conclusion, this work has demonstrated that the low-molecular-weight peptide CS-1 could serve as a cell-selective adhesion mediator in the engineering of a more-compatible small-caliber vascular graft lumen interface.
Collapse
Affiliation(s)
- Eric J Rodenberg
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | |
Collapse
|
15
|
Singh P, Reimer CL, Peters JH, Stepp MA, Hynes RO, Van De Water L. The Spatial and Temporal Expression Patterns of Integrin α9β1 and One of Its Ligands, the EIIIA Segment of Fibronectin, in Cutaneous Wound Healing. J Invest Dermatol 2004; 123:1176-81. [PMID: 15610531 DOI: 10.1111/j.0022-202x.2004.23485.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The fibronectins (FN) comprise a family of adhesive extracellular matrix proteins thought to mediate important functions in cutaneous wounds. Plasma fibronectin (pFN) extravasates for days from intact hyperpermeable vessels following injury whereas mRNAs encoding the cellular fibronectins (cFN) that include two segments, termed EIIIA (EDA) and EIIIB (EDB), are expressed by wound cells. Wounds in mice null for pFN appear to heal normally whereas those in EIIIA null mice exhibit defects, suggesting that cFN may play a role when pFN is missing. Integrin alpha9beta1, a receptor for several extracellular matrix proteins as well as the EIIIA segment, is expressed normally in the basal layer of squamous epithelia. We report results from immunohistochemistry on healing wounds demonstrating that EIIIA-containing cFN are deposited abundantly but transiently from day 4 to 7 whereas EIIIB-containing cFN persist at least through day 14. Elevated expression of alpha9beta1 is seen in basal and suprabasal epidermal keratinocytes in wounds. The spatial expression patterns of cFN and alpha9beta1 are distinct, but overlap in the dermal-epidermal junction, and both are expressed contemporaneously. These observations suggest a role for alpha9beta1-EIIIA interactions in wound keratinocyte function.
Collapse
Affiliation(s)
- Purva Singh
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, New York, USA
| | | | | | | | | | | |
Collapse
|
16
|
Anusaksathien O, Webb SA, Jin QM, Giannobile WV. Platelet-derived growth factor gene delivery stimulates ex vivo gingival repair. ACTA ACUST UNITED AC 2004; 9:745-56. [PMID: 13678451 PMCID: PMC2586961 DOI: 10.1089/107632703768247421] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Destruction of tooth support due to the chronic inflammatory disease periodontitis is a major cause of tooth loss. There are limitations with available treatment options to tissue engineer soft tissue periodontal defects. The exogenous application of growth factors (GFs) such as platelet-derived growth factor (PDGF) has shown promise to enhance oral and periodontal tissue regeneration. However, the topical administration of GFs has not led to clinically significant improvements in tissue regeneration because of problems in maintaining therapeutic protein levels at the defect site. The utilization of PDGF gene transfer may circumvent many of the limitations with protein delivery to soft tissue wounds. The objective of this study was to test the effect of PDGF-A and PDGF-B gene transfer to human gingival fibroblasts (HGFs) on ex vivo repair in three-dimensional collagen lattices. HGFs were transduced with adenovirus encoding PDGF-A and PDGF-B genes. Defect fill of bilayer collagen gels was measured by image analysis of cell repopulation into the gingival defects. The modulation of gene expression at the defect site and periphery was measured by RT-PCR during a 10-day time course after gene delivery. The results demonstrated that PDGF-B gene transfer stimulated potent (>4-fold) increases in cell repopulation and defect fill above that of PDGF-A and corresponding controls. PDGF-A and PDGF-B gene expression was maintained for at least 10 days. PDGF gene transfer upregulated the expression of phosphatidylinosital 3-kinase and integrin alpha5 subunit at 5 days after adenovirus transduction. These results suggest that PDGF gene transfer has potential for periodontal soft tissue-engineering applications.
Collapse
Affiliation(s)
- Orasa Anusaksathien
- Center for Craniofacial Regeneration and Department of Periodontics, Prevention, and Geriatrics, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109-1078, USA
| | | | | | | |
Collapse
|
17
|
Clark RAF, Lin F, Greiling D, An J, Couchman JR. Fibroblast invasive migration into fibronectin/fibrin gels requires a previously uncharacterized dermatan sulfate-CD44 proteoglycan. J Invest Dermatol 2004; 122:266-77. [PMID: 15009704 DOI: 10.1046/j.0022-202x.2004.22205.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
After tissue injury, fibroblast migration from the peri-wound collagenous stroma into the fibrin-laden wound is critical for granulation tissue formation and subsequent healing. Recently we found that fibroblast transmigration from a collagen matrix into a fibrin matrix required the presence of fibronectin. Several integrins-alpha 4 beta 1, alpha 5 beta 1, and alpha v beta 3-with known fibronectin binding affinity were necessary for this invasive migration. Here we examined another family of cell surface receptors: the proteoglycans. We found that dermatan sulfate was required for fibroblast migration into a fibronectin/fibrin gel. This conclusion was based on beta-xyloside inhibition of glycanation and specific glycosaminoglycan degradation. CD44, a cell surface receptor known to bind hyaluronan, not infrequently exists as a proteoglycan, decorated with various glycosaminoglycan chains including heparan sulfate and chondroitin sulfate, and as such can bind fibronectin. We found that CD44H, the non-spliced isoform of CD44, was necessary for fibroblast invasion into fibronectin/fibrin gels. Resting fibroblasts expressed mostly nonglycanated CD44H core protein, which became glycanated with chondroitin sulfate and dermatan sulfate, but not heparan sulfate, after a 24 h incubation with platelet-derived growth factor, the stimulus used in the migration assay. These results demonstrate that dermatan sulfate-CD44H proteoglycan is essential for fibroblast migration into fibrin clots and that platelet-derived growth factor, the stimulus for migration, induces the production of chondroitin-sulfate- and dermatan-sulfate-glycanated CD44H.
Collapse
Affiliation(s)
- Richard A F Clark
- Department of Dermatology, School of Medicine, SUNY at Stony Brook, New York, 11794-8165, USA.
| | | | | | | | | |
Collapse
|
18
|
Clark RAF, An JQ, Greiling D, Khan A, Schwarzbauer JE. Fibroblast migration on fibronectin requires three distinct functional domains. J Invest Dermatol 2003; 121:695-705. [PMID: 14632184 DOI: 10.1046/j.1523-1747.2003.12484.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mesenchymal cell movement is normally constrained; however, fibronectin can provide a pathway for stromal cell migration during embryogenesis, morphogenesis, and wound healing. Cells can adhere to fibronectin via integrin and nonintegrin receptors, which bind multiple unique peptide sequences. Synthetic peptides and recombinant proteins were used to delineate the functional domains needed for human fibroblast migration over fibronectin. The 9th and 10th fibronectin type III repeats, which contain RGD and PHSRN synergy cell attachment sequences, support almost maximal fibroblast attachment, but not migration of primary dermal fibroblasts. Specific sequences within the heparin domain and the IIICS region are also required for migration. These findings predict and additional data confirm the necessity for the cooperation of multiple integrin and nonintegrin receptors for fibroblast migration on fibronectin. Such stringency of migration most likely imposes an immense constraint on normal mesenchymal cell mobility in unperturbed tissue. Loss of such restraint may be critical for the migration cancer cells through the extracellular matrix.
Collapse
Affiliation(s)
- Richard A F Clark
- Department of Dermatology, School of Medicine, SUNY at Stony Brook, Stony Brook, New York 11794-8165, USA.
| | | | | | | | | |
Collapse
|
19
|
Hersel U, Dahmen C, Kessler H. RGD modified polymers: biomaterials for stimulated cell adhesion and beyond. Biomaterials 2003; 24:4385-415. [PMID: 12922151 DOI: 10.1016/s0142-9612(03)00343-0] [Citation(s) in RCA: 1764] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since RGD peptides (R: arginine; G: glycine; D: aspartic acid) have been found to promote cell adhesion in 1984 (Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule, Nature 309 (1984) 30), numerous materials have been RGD functionalized for academic studies or medical applications. This review gives an overview of RGD modified polymers, that have been used for cell adhesion, and provides information about technical aspects of RGD immobilization on polymers. The impacts of RGD peptide surface density, spatial arrangement as well as integrin affinity and selectivity on cell responses like adhesion and migration are discussed.
Collapse
Affiliation(s)
- Ulrich Hersel
- Institut für Organische Chemie und Biochemie, Technische Universität München, Lichtenbergstr. 4, D-85747, Garching, Germany
| | | | | |
Collapse
|
20
|
Sethi KK, Yannas IV, Mudera V, Eastwood M, McFarland C, Brown RA. Evidence for sequential utilization of fibronectin, vitronectin, and collagen during fibroblast-mediated collagen contraction. Wound Repair Regen 2002; 10:397-408. [PMID: 12453144 DOI: 10.1046/j.1524-475x.2002.10609.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Contraction plays a major role in wound healing and is inevitably mediated through the mechanical interaction of fibroblast cytoskeleton and integrins with their extracellular matrix ligands. Cell-matrix attachment is critical for such events. In human dermal fibroblasts most such interactions are mediated by the beta1-type integrins. This study investigated the role played by key components in this system, notably fibronectin, vitronectin, and integrin subcomponents alpha2 and alpha5, which recognize collagen and fibronectin. Inhibition of adhesion through these ligands was studied either by antibody blocking or with fibronectin and/or vitronectin depletion. Functional effects of inhibition were monitored as force generation in collagen-glycosaminoglycan (IntegraTM) sponges, over 20 hours using a culture force monitor. Dose and time-course inhibition studies indicated that initial attachment and force generation (approx. 0-5 hours postseeding) was through fibronectin receptors and this was followed by vitronectin ligand and receptor utilization (4 hours onward). Utilization of the collagen integrin subcomponent alpha2 appeared to be increasingly important between 6 and 16 hours and dominant thereafter. Additionally, there was evidence for functional interdependence between the three ligand systems fibronectin, vitronectin, and collagen. We propose that there is a short cascade of sequential integrin-ligand interactions as cells attach to, extend through, and eventually contract their matrix. (WOUND REP REG 2002;10:-408)
Collapse
Affiliation(s)
- Kamaljit K Sethi
- University College London, RFUCMS, Tissue Repair & Engineering Center, Institute of Orthopaedics, RNOH campus, Stanmore, United Kingdom
| | | | | | | | | | | |
Collapse
|
21
|
Sethi KK, Mudera V, Sutterlin R, Baschong W, Brown RA. Contraction-mediated pinocytosis of RGD-peptide by dermal fibroblasts: inhibition of matrix attachment blocks contraction and disrupts microfilament organisation. CELL MOTILITY AND THE CYTOSKELETON 2002; 52:231-41. [PMID: 12112137 DOI: 10.1002/cm.10047] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Force generation in collagen and matrix contraction are basic functions of fibroblasts and important elements of tissue repair. Cell-matrix attachment is critical to this contraction, involving RGD-binding integrins. We have investigated how this process operates, in terms of force generation (in the Culture Force Monitor) and cytoskeletal structure, using a synthetic RGD-decapeptide. The RGD-peptide blocked force generation over the first 6 h, followed by near complete recovery by 20 h. However, dose response was complex indicating multiple processes were operating. Analysis of cytoskeletal structure after treatment with RGD-peptide indicated major disruption with condensed aggregates of actin and microtubular fragmentation. Fluorescent labeling and tracking of the RGD-peptide demonstrated intracellular uptake into discrete cytoplasmic aggregates. Critically, these RGD-peptide pools co-localised with the condensed actin microfilament aggregates. It is concluded that RGD-peptide uptake was by a form of contraction-mediated pinocytosis, resulting from mechanical tension applied to the untethered RGD-peptide-integrin, as contractile microfilament were assembled. These findings emphasize the importance of sound mechanical attachment of ligand-occupied integrins (e.g., to extracellular matrix) for normal cytoskeletal function. Conversely, this aspect of unrestrained cytoskeletal contraction may have important pathogenic and therapeutic applications.
Collapse
Affiliation(s)
- K K Sethi
- University College London, Tissue Repair and Engineering Centre (TREC), Institute of Orthopaedics, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | | | | | | | | |
Collapse
|
22
|
Steffensen B, Häkkinen L, Larjava H. Proteolytic events of wound-healing--coordinated interactions among matrix metalloproteinases (MMPs), integrins, and extracellular matrix molecules. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 2002; 12:373-98. [PMID: 12002821 DOI: 10.1177/10454411010120050201] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During wound-healing, cells are required to migrate rapidly into the wound site via a proteolytically generated pathway in the provisional matrix, to produce new extracellular matrix, and, subsequently, to remodel the newly formed tissue matrix during the maturation phase. Two classes of molecules cooperate closely to achieve this goal, namely, the matrix adhesion and signaling receptors, the integrins, and matrix-degrading and -processing enzymes, the matrix metalloproteinases (MMPs). There is now substantial experimental evidence that blocking key molecules of either group will prevent or seriously delay wound-healing. It has been known for some time now that cell adhesion by means of the integrins regulates the expression of MMPs. In addition, certain MMPs can bind to integrins or other receptors on the cell surface involved in enzyme activation, thereby providing a mechanism for localized matrix degradation. By proteolytically modifying the existing matrix molecules, the MMPs can then induce changes in cell behavior and function from a state of rest to migration. During wound repair, the expression of integrins and MMPs is simultaneously up-regulated. This review will focus on those aspects of the extensive knowledge of fibroblast and keratinocyte MMPs and integrins in biological processes that relate to wound-healing.
Collapse
Affiliation(s)
- B Steffensen
- Department of Periodontics, University of Texas Health Science Center at San Antonio, 78229-3900, USA.
| | | | | |
Collapse
|
23
|
Liao YF, Gotwals PJ, Koteliansky VE, Sheppard D, Van De Water L. The EIIIA segment of fibronectin is a ligand for integrins alpha 9beta 1 and alpha 4beta 1 providing a novel mechanism for regulating cell adhesion by alternative splicing. J Biol Chem 2002; 277:14467-74. [PMID: 11839764 DOI: 10.1074/jbc.m201100200] [Citation(s) in RCA: 167] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Alternative splicing of the fibronectin gene transcript gives rise to forms that include the EIIIA (or ED-A) segment. EIIIA-containing fibronectins are prominently expressed during embryogenesis and wound healing and appear to mediate changes in cell adhesion and gene expression. Nonetheless, integrins that bind the EIIIA segment have not been identified. We previously mapped the epitope for two function-blocking monoclonal antibodies to the C-C' loop region of the EIIIA segment (Liao, Y.-F., Wieder, K. G., Classen, J. M., and Van De Water, L. (1999) J. Biol. Chem. 274, 17876-17884). The sequence of this epitope ((39)PEDGIHELFP(48)) resembles the sequence within tenascin-C to which the integrin alpha(9)beta(1) binds. We now report that either integrin alpha(9)beta(1) or alpha(4)beta(1) can mediate cell adhesion to the EIIIA segment. Moreover, this interaction is blocked both by epitope-mapped EIIIA antibodies as well as by the respective anti-integrins. Deletion mutants of the EIIIA segment that include the C-C' loop and flanking sequence bind cells expressing either alpha(9)beta(1) or alpha(4)beta(1). Adhesion of alpha(4)beta(1)-containing MOLT-3 cells to the EIIIA segment stimulates phosphorylation of p44/42 MAP kinase. Our observation that two integrins bind the EIIIA segment establishes a novel mechanism by which cell adhesion to fibronectin is regulated by alternative splicing.
Collapse
Affiliation(s)
- Yung-Feng Liao
- Center for Engineering in Medicine and Surgical Service, Massachusetts General Hospital and Harvard Medical School, the Shriners Burns Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | |
Collapse
|
24
|
Kubo M, Van de Water L, Plantefaber LC, Mosesson MW, Simon M, Tonnesen MG, Taichman L, Clark RA. Fibrinogen and fibrin are anti-adhesive for keratinocytes: a mechanism for fibrin eschar slough during wound repair. J Invest Dermatol 2001; 117:1369-81. [PMID: 11886497 DOI: 10.1046/j.0022-202x.2001.01551.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During cutaneous wound repair the epidermis avoids the fibrin-rich clot; rather it migrates down the collagen-rich dermal wound margin and over fibronectin-rich granulation tissue. The mechanism(s) underlying keratinocyte movement in this precise pathway has not been previously addressed. Here we demonstrate that cultured human keratinocytes do not express functional fibrinogen/fibrin receptors, specifically alpha v beta 3. Biologic modifiers known to induce integrin expression or activation did not induce adhesion to fibrin, fibrinogen, or its fragments. Epidermal explant outgrowth and single epidermal cell migration failed to occur on either fibrin or fibrinogen. Surprisingly, fibrin and fibrinogen mixed at physiologic molar ratios with fibronectin abrogated keratinocyte attachment to fibronectin. Keratinocytes transduced with the beta 3 integrin subunit cDNA, expressed alpha v beta 3 on their surface and attached to and spread on fibrinogen and fibrin. beta-gal cDNA-transduced keratinocytes did not demonstrate this activity. Furthermore, beta 3 cDNA-transduced keratinocyte adhesion to fibrin was inhibited by LM609 monoclonal antibody to alpha v beta 3 in a concentration-dependent fashion. From these data, we conclude that normal human keratinocytes cannot interact with fibrinogen and its derivatives due to the lack of alpha v beta 3. Thus, fibrinogen and fibrin are authentic anti-adhesive for keratinocytes. This may be a fundamental reason why the migrating epidermis dissects the fibrin eschar from wounds.
Collapse
Affiliation(s)
- M Kubo
- Department of Dermatology, School of Medicine, SUNY at Stony Brook, Stony Brook, New York, New York 11794-8165, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Regan MC, Flavin BM, Fitzpatrick JM, O'Connell PR. Stricture formation in Crohn's disease: the role of intestinal fibroblasts. Ann Surg 2000; 231:46-50. [PMID: 10636101 PMCID: PMC1420964 DOI: 10.1097/00000658-200001000-00007] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To determine whether intestinal fibroblasts in patients with Crohn's disease (CD) have an enhanced capacity to reorganize collagen and thus cause stricture formation. SUMMARY BACKGROUND DATA Stricture formation is a characteristic feature of CD that may distinguish it from other forms of inflammatory bowel disease. Methods Fibroblasts were obtained at surgery from the colon and ileum of patients with CD and ulcerative colitis (UC) and control patients. Primary fibroblast cultures were obtained by explant technique. Fibroblast contractile activity was measured using fibroblast-populated collagen lattices (FPCLs), in which the cultured fibroblasts were seeded in free-floating collagen gel matrices that they reorganize and contract. Fibroblast contractile activity was measured as the reduction of surface area (mm2) of collagen gel matrix at 24-hour intervals for 1 week. RESULTS Fibroblasts from patients with CD displayed enhanced capacity to contract FPCL when compared to UC and control fibroblasts. This activity was maximal in fibroblasts recovered from strictured regions in CD. Fibroblasts from patients with UC had a contractile capacity similar to that of controls. Hydrocortisone inhibited this in vitro contractile activity in a dose-dependent manner. CONCLUSIONS Intestinal fibroblasts in CD possess enhanced capacity for collagen reorganization and contractile activity in vitro. This activity may be responsible for stricture formation in CD.
Collapse
Affiliation(s)
- M C Regan
- Department of Surgery, Mater Misericordiae Hospital, Dublin, Ireland
| | | | | | | |
Collapse
|
26
|
Gailit J, Clarke C, Newman D, Tonnesen MG, Mosesson MW, Clark RA. Human fibroblasts bind directly to fibrinogen at RGD sites through integrin alpha(v)beta3. Exp Cell Res 1997; 232:118-26. [PMID: 9141628 DOI: 10.1006/excr.1997.3512] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Fibroblast migration into the blood clot initially filling a wound requires close interaction between fibroblasts and the matrix of the fibrin clot. However, very little is known about the specific receptor-ligand interactions that mediate fibroblast attachment to fibrin. Using an attachment assay developed to measure even relatively weak interactions, we demonstrate here that normal human dermal fibroblasts can attach to substrates coated with fibrinogen, fibrin, or the fibrinogen breakdown product I-9D. Fibroblast attachment to these ligands did not require the presence of fibronectin on the cell surface or as a component of the substrate. Cells treated with cycloheximide and monensin, to limit the synthesis and secretion of endogenous fibronectin, attached as well as untreated cells. The synthetic peptide GRGDS inhibited adhesion to fibrinogen, fibrin, and fibrinogen I-9D by about 60%, while the control peptide GRGES had no substantial effect. We conclude that attachment to these ligands is mediated at least partially by direct interactions between the substrates and one specific receptor, the integrin alpha(v)beta3. Affinity chromatography demonstrated that alpha(v)beta3 from detergent lysates of fibroblasts bound to a fibrinogen matrix and was eluted with EDTA. Furthermore, antibodies against the alpha(v)beta3 complex or against the alpha(v) subunit inhibited fibroblast attachment to fibrinogen and fibrin by 50-70%. An inhibitory antibody against the integrin beta1 subunit had no effect. The observation that integrin antagonists could not produce complete inhibition suggests that there may be other fibroblast cell surface proteins that can bind directly to fibrinogen.
Collapse
Affiliation(s)
- J Gailit
- Department of Dermatology, State University of New York at Stony Brook, 11794, USA
| | | | | | | | | | | |
Collapse
|
27
|
Greiling D, Clark RA. Fibronectin provides a conduit for fibroblast transmigration from collagenous stroma into fibrin clot provisional matrix. J Cell Sci 1997; 110 ( Pt 7):861-70. [PMID: 9133673 DOI: 10.1242/jcs.110.7.861] [Citation(s) in RCA: 197] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
After injury, the wound space is filled with a fibrin/fibronectin clot containing growth factors released by platelets and monocytes. In response to these factors, fibroblasts migrate into the fibrin clot and contribute to the formation of granulation tissue. The functional mechanisms allowing fibroblasts to leave the collagenous matrix of normal connective tissue and invade the provisional matrix of the fibrin clot have not been fully defined. To investigate these mechanisms we established a new in vitro model which simulates specific aspects of early wound healing, that is, the migration of fibroblasts from a three-dimensional collagen matrix into a fibrin clot. This transmigration could be induced by physiological concentrations of platelet releasate or platelet-derived growth factor BB (PDGF-BB) in a concentration-dependent manner. At 24 hours irradiated fibroblasts invaded the fibrin gel almost as well as non-irradiated cells, indicating that transmigration was independent of proliferation. Plasminogen and its activators appear to be necessary for invasion of the fibrin clot since protease inhibitors decreased the amount of migration. These serine proteases, however, were not necessary for exit from the collagen gel as fibroblasts migrated out of the collagen gel onto a surface coated with fibrin fibrils even in the presence of inhibitors. Removal of fibronectin (FN) from either the collagen gel or the fibrin gel markedly decreased the number of migrating cells, suggesting that FN provides a conduit for transmigration. Cell movement in the in vitro model was inhibited by RGD peptide, and by monoclonal antibodies against the subunits of the alpha5 beta1 and alpha v beta3 integrin receptor. Thus, the functional requirements for fibroblast transmigration from collagen-rich to fibrin-rich matrices, such as occurs in early wound healing, have been partially defined using an in vitro paradigm of this important biologic process.
Collapse
Affiliation(s)
- D Greiling
- Department of Dermatology, School of Medicine, SUNY at Stony Brook, New York 11794-8165, USA
| | | |
Collapse
|
28
|
Gailit J, Clark RA. Studies in vitro on the role of alpha v and beta 1 integrins in the adhesion of human dermal fibroblasts to provisional matrix proteins fibronectin, vitronectin, and fibrinogen. J Invest Dermatol 1996; 106:102-8. [PMID: 8592057 DOI: 10.1111/1523-1747.ep12328177] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibroblasts that migrate into a wound during the early stages of repair use cell surface integrins to interact with extracellular molecules as they move away from the interstitial matrix of normal tissue and into the provisional matrix of the wound. Therefore, to understand a critical phase of wound healing, it is necessary to understand the details of integrin involvement. Normal adult human dermal fibroblasts in culture express many receptors for the provisional matrix proteins fibronectin, vitronectin, and fibrinogen, including the integrins alpha 3 beta 1, alpha 4 beta 1, alpha 5 beta 1, alpha v beta 1, alpha v beta 3, and alpha v beta 5. We used quantitative flow cytometry to estimate the relative numbers of these receptors and immunoprecipitation to confirm the expression of alpha v beta 1. Adult human dermal fibroblasts primarily use beta 1 integrins, alpha 4 beta 1, alpha 5 beta 1, and possibly alpha v beta 1, for attachment to fibronectin. alpha v beta 3 and perhaps other integrins containing the alpha v subunit serve fibroblasts as secondary or auxiliary receptors for fibronectin. In contrast, these cells use alpha v integrins but probably not beta 1 integrins for attachment to vitronectin. alpha v beta 3 and alpha v beta 5 apparently act in concert to mediate attachment to vitronectin, and these two integrins may perform different functions during wound repair. Fibroblast adhesion to certain preparations of fibrinogen occurs, at least partially, through the small amount of fibronectin present in the preparations. Fibroblast attachment to fibrinogen purified free of fibronectin also occurs, and that was demonstrated with a sensitive new assay called electrical cell-substrate impedance sensing. Fibroblast attachment to pure fibrinogen can be inhibited by RGD peptide, suggesting that integrins are involved.
Collapse
Affiliation(s)
- J Gailit
- Department of Dermatology, State University of New York at Stony Brook 11794-8165, USA
| | | |
Collapse
|
29
|
Bronson RA, Gailit J, Bronson S, Oula L. Echistatin, a disintegrin, inhibits sperm-oolemmal adhesion but not oocyte penetration**Funded in part by Research Foundation Grant No. 9384, State University of New York at Stony Brook, Stony Brook, New York. Fertil Steril 1995. [DOI: 10.1016/s0015-0282(16)57744-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
30
|
Huhtala P, Humphries MJ, McCarthy JB, Tremble PM, Werb Z, Damsky CH. Cooperative signaling by alpha 5 beta 1 and alpha 4 beta 1 integrins regulates metalloproteinase gene expression in fibroblasts adhering to fibronectin. J Cell Biol 1995; 129:867-79. [PMID: 7537277 PMCID: PMC2120442 DOI: 10.1083/jcb.129.3.867] [Citation(s) in RCA: 295] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Rabbit synovial fibroblasts (RSF) express basal levels of the metalloproteinases (MMP) collagenase, stromelysin-1 and 92-kD gelatinase when plated on intact fibronectin (FN), but elevated levels when plated on either the central RGD-containing cell-binding region of FN (120FN) or antibody against the alpha 5 beta 1 integrin, suggesting that domains outside 120FN may suppress the induction of MMP (Werb, Z., P. M. Tremble, O. Behrendtsen, E. Crowley, and C.H. Damsky. 1989. J. Cell Biol. 109:877-889). We therefore attempted to reconstitute the basal signaling of intact FN by plating RSF on 120FN together with domains of FN outside this region. Large COOH-terminal fragments containing both the heparin-binding and HICS domains suppressed MMP when combined with 120FN. To map the active sequences, peptides from this region and larger fragments that did, or did not, include the CS-1 portion of IIICS were tested. Only CS-1 peptide, or larger fragments containing CS-1, suppressed MMP expression induced by 120FN. In contrast, peptide V from the heparin-binding region, shown previously to stimulate focal contact formation, further enhanced MMP expression by RSF when present on the substrate with 120FN. RSF expressed alpha 4 beta 1 integrin, the receptor for CS-1, and the anti-alpha 4 mAb blocked the ability of CS-1 to suppress MMP induction by 120FN. These results show that signals modulating MMP expression and focal contact assembly are regulated independently, and that cooperative signaling by alpha 5 beta 1 and alpha 4 beta 1 integrins plays a dominant role in regulating expression of these extracellular matrix-remodeling genes in response to FN. This work demonstrates directly the modular way in which information in the extracellular matrix is detected and processed by cell surface receptors.
Collapse
Affiliation(s)
- P Huhtala
- Department of Stomatology, University of California, San Francisco 94143, USA
| | | | | | | | | | | |
Collapse
|
31
|
Dalton BA, McFarland CD, Underwood PA, Steele JG. Role of the heparin binding domain of fibronectin in attachment and spreading of human bone-derived cells. J Cell Sci 1995; 108 ( Pt 5):2083-92. [PMID: 7657726 DOI: 10.1242/jcs.108.5.2083] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human bone-derived cells are known to attach and spread on surfaces which have been precoated with fibronectin, but the contributions made by specific domains of the molecule have not yet been defined. Here we refer to the osteoblast-like cells as human bone cells. We have determined the relevance of separate regions of fibronectin, particularly the heparin-binding region, for the initial attachment and spreading of these cells. Human bone cells attached to fragments from each of the cell- and heparin-binding regions of fibronectin, but failed to attach to a fragment from the gelatin-binding region. Bovine corneal epithelial cells, which were included as an example of an alternative primary cell strain, attached to the cell-binding fragment but showed no specific short-term attachment to the heparin or gelatin-binding fragments. Monoclonal antibody MAb17, which binds to the cell binding region of fibronectin, partially inhibited the attachment of both human bone cells and corneal epithelial cells to intact fibronectin when present at 50 micrograms/ml and reduced human bone cell attachment to the cell-binding region fragment of fibronectin. Monoclonal antibody, MAb 32, which binds to the heparin-binding region of fibronectin, failed to inhibit attachment of the human bone cells to fibronectin but reduced the attachment of these cells to the heparin-binding region fragment. Heparin and chondroitin sulphate were able to inhibit human bone cell attachment to the heparin-binding fragment of fibronectin but had no effect on their attachment to intact fibronectin or the cell-binding region of fibronectin. Immunofluorescent staining and confocal microscopy showed extensive spreading and actin filament formation when human bone cells were cultured on intact fibronectin. Cells cultured on the heparin-binding fragment showed only minimal spreading coinciding with less extensive actin filament organisation. On the cell-binding fragment of fibronectin more spreading was seen than on the heparin-binding fragment but it was not as extensive as on intact fibronectin. Taken together, these data suggest that human bone cells, unlike bovine corneal epithelial cells, have an attachment mechanism for the heparin-binding region of fibronectin. Attachment to this region is probably mediated by cell surface proteoglycans. However, interaction with the cell-binding domain is required for effective cell spreading of human bone cells on fibronectin during the first 90 minutes after seeding into culture.
Collapse
Affiliation(s)
- B A Dalton
- CSIRO Division of Biomolecular Engineering, Sydney Laboratory, North Ryde, NSW, Australia
| | | | | | | |
Collapse
|
32
|
Audet JF, Masson JY, Rosen GD, Salesse C, Guérin SL. Multiple regulatory elements control the basal promoter activity of the human alpha 4 integrin gene. DNA Cell Biol 1994; 13:1071-85. [PMID: 7702751 DOI: 10.1089/dna.1994.13.1071] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
It has been suggested that expression of the genes encoding the alpha 4/beta 1 integrin increases during wound healing of the cornea. As a first step in understanding the mechanisms required to stimulate alpha 4 gene expression during this process, we defined the minimal upstream sequence required to direct basal promoter activity for this gene. Using deletion analyses of the alpha 4 gene upstream sequence, we identified two functionally important negative regulatory elements. Dimethylsulfate (DMS) methylation interference assays provided evidence for the binding of a single nuclear protein to tandemly repeated homologous cis-acting elements (designated alpha 4.1 and alpha 4.2) from the alpha 4 basal promoter that share the core sequence 5'-GTGGGT-3'. The formation of a protection only at alpha 4.1 in DNase I footprinting suggested that it is the primary target element for the binding of nuclear proteins. Three distinct nuclear proteins bound a double-stranded oligonucleotide bearing the DNA sequence of alpha 4.1 to produce specific DNA-protein complexes (R1 to R3) in gel-shift assays, from which that producing R3 was identified as the protein yielding DNase I protection at alpha 4.1. Detailed mutational analysis of alpha 4.1 and alpha 4.2 indicated that both elements positively regulate gene expression in primary cultures of corneal epithelial cells and Jurkat tissue culture cells, which is consistent with the deletion analysis. However, when transiently transfected into pituitary GH4C1, the alpha 4.2 mutants yielded increased chloramphenicol acetyl transferase activity therefore demonstrating that these elements have the ability to function either as positive or negative regulators of gene transcription in a manner that is dependent on the type of cell transfected.
Collapse
Affiliation(s)
- J F Audet
- Unit of Ophthalmology, CHUL Research Center, Ste-Foy, Québec, Canada
| | | | | | | | | |
Collapse
|
33
|
Noiri E, Gailit J, Sheth D, Magazine H, Gurrath M, Muller G, Kessler H, Goligorsky MS. Cyclic RGD peptides ameliorate ischemic acute renal failure in rats. Kidney Int 1994; 46:1050-8. [PMID: 7861698 DOI: 10.1038/ki.1994.366] [Citation(s) in RCA: 91] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Renal tubular obstruction is an important contributor to the pathophysiology of acute renal failure. Based on the previous findings of the role played by arginine-glycine-aspartic acid (RGD) recognizing integrins in tubular obstruction, this study examined the effect of RGD peptides on the course of ischemic acute renal failure in rats. For in vivo studies, animals were subjected to 45 minutes of unilateral renal ischemia with contralateral nephrectomy, and cyclic RGD peptides or a linear biotinylated RGD peptide were injected systemically after the release of renal artery clamp. In vitro studies compared the potency of the peptides in inhibiting BS-C-1 cell-matrix and cell-cell adhesion. Two novel cyclic RGD peptides utilized in these studies showed different inhibitory potency in preventing cell-matrix adhesion: cyclic RGDDFV was a highly potent in vitro inhibitor of BS-C-1 cell-matrix adhesion, whereas cyclic RGDDFLG was less potent. In cell-cell adhesion assays, however, both peptides were equipotent. Despite the differences in inhibiting cell-matrix adhesion, a single systemic administration of either peptide improved creatinine clearance postoperatively and accelerated recovery of renal function with a rank order: cyclic RGDDFV > or = RGDDFLG >> RDADFV (inactive control). These findings represent the first in vivo demonstration of the effectiveness of cyclic RGD peptides in ameliorating ischemic acute renal failure, and suggest that in this setting RGD peptides predominantly inhibit cell-cell adhesion, whereas inhibition of cell-matrix adhesion is of lesser significance.
Collapse
Affiliation(s)
- E Noiri
- Department of Medicine, State University of New York, Stony Brook
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Gailit J, Welch MP, Clark RA. TGF-beta 1 stimulates expression of keratinocyte integrins during re-epithelialization of cutaneous wounds. J Invest Dermatol 1994; 103:221-7. [PMID: 8040614 DOI: 10.1111/1523-1747.ep12393176] [Citation(s) in RCA: 170] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Epidermal keratinocytes migrate over a provisional matrix during the re-epithelialization of cutaneous wounds. We have investigated the expression of integrins and of transforming growth factor-beta 1 (TGF-beta 1) during re-epithelialization in a porcine model. Tissue specimens were collected at different times after injury and stained with antibodies against subunits of the fibronectin receptor, integrin alpha 5 beta 1, and the vitronectin receptor, integrin alpha v beta 5. Intense staining was observed in the migrating keratinocytes of 5-d wounds; basal and suprabasal cells were stained around the entire cell periphery. Staining returned toward normal levels in 14-d wounds. The appearance of the extracellular form of TGF-beta 1 seemed to be coordinated with the increased expression of integrin subunits: it was detected in migrating keratinocytes and in the adjacent epidermis of early wounds at 5 and 7 d. We also investigated the effect of TGF-beta 1 on cultured epidermal cells. Treating human keratinocytes with TGF-beta 1 increased the levels of mRNA for the integrin subunits alpha 5, alpha v, and beta 5, but had little effect on beta 1. The corresponding cell-surface expression of alpha 5 and alpha v was also increased after treatment. Thus, during wound repair, TGF-beta 1 may induce epidermal keratinocytes to express integrins that facilitate the migratory component of re-epithelialization.
Collapse
Affiliation(s)
- J Gailit
- Department of Dermatology, School of Medicine, SUNY at Stony Brook 11794-8165
| | | | | |
Collapse
|
35
|
Sunardhi-Widyaputra S, Van Damme B. Distribution of the VLA family of integrins in normal salivary gland and in pleomorphic adenoma. Pathol Res Pract 1994; 190:600-8. [PMID: 7984519 DOI: 10.1016/s0344-0338(11)80398-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The extracellular matrix (ECM) and the alterations in tumour stroma affect the biologic behaviour of tumours. These interactions are, at least in part, mediated by the integrin superfamily. In this study we examined the immunohistochemical expression of the "very late activation" (VLA)-integrins in pleomorphic adenomas and in the corresponding normal tissues. Normal salivary gland expresses integrins in the myoepithelial cells (MEC) and in the basal and dark cells of ducts. This pattern is retained in pleomorphic adenoma. Regardless of the straining intensity, these tumour cells have a VLA expression similar to normal basal and dark cells, with only a few cells bearing MEC characteristics. We suggest that the tumour cells of pleomorphic adenoma have a common origin but show different stages of differentiation. Only cells bearing basement membrane (BM)-associated material form typical tubulo-ductal structures.
Collapse
Affiliation(s)
- S Sunardhi-Widyaputra
- Department of Pathology, Sint Raphael University Hospital, Catholic University of Leuven, Belgium
| | | |
Collapse
|
36
|
High AS, Robinson PA. Differences in adhesion and collagen gel contraction between fibroblasts from various types of odontogenic cyst. Arch Oral Biol 1994; 39:387-93. [PMID: 8060261 DOI: 10.1016/0003-9969(94)90168-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Fibroblasts were grown from explants of normal gingiva and foreskin, and from walls of dentigerous, radicular and residual cysts, as well as keratocysts of basal-cell naevus syndrome and non-syndrome origin. Dentigerous-cyst fibroblast adhesion to poly-L-lysine-coated glass was unaffected by all adhesion-related glycoproteins. Chondroitin sulphate, fibronectin and heparan sulphate enhanced attachment of all other fibroblasts. Chondroitin-sulphate and fibronectin-enhanced adhesion was blocked by an arg-gly-asp peptide. Fibronectin, chondroitin sulphate and laminin all promoted collagen lattice contraction using normal gingival fibroblasts and low-serum media. Fibronectin had a greater effect than chondroitin sulphate and laminin. In media with standard serum, all cyst fibroblast lines examined demonstrated similar gel contraction curves with the exception of dentigerous cyst-derived fibroblasts, which contracted at decreased rates. Suppression of gel contraction was seen with dentigerous-cyst fibroblasts with all extracellular matrix glycoproteins and low serum. Dentigerous-cyst fibroblast attachment to glass and behaviour in gel lattices suggest that these cells express different functional attachment factors from other cyst fibroblast types.
Collapse
Affiliation(s)
- A S High
- Division of Dental Surgery, Leeds Dental Institute, U.K
| | | |
Collapse
|
37
|
LeRoy EC. The control of fibrosis in systemic sclerosis: a strategy involving extracellular matrix, cytokines, and growth factors. J Dermatol 1994; 21:1-4. [PMID: 8157815 DOI: 10.1111/j.1346-8138.1994.tb01401.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- E C LeRoy
- Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston 29425-2229
| |
Collapse
|
38
|
|