1
|
Kim HY, Shim JH, Heo CY. A Rare Skeletal Disorder, Fibrous Dysplasia: A Review of Its Pathogenesis and Therapeutic Prospects. Int J Mol Sci 2023; 24:15591. [PMID: 37958575 PMCID: PMC10650015 DOI: 10.3390/ijms242115591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Fibrous dysplasia (FD) is a rare, non-hereditary skeletal disorder characterized by its chronic course of non-neoplastic fibrous tissue buildup in place of healthy bone. A myriad of factors have been associated with its onset and progression. Perturbation of cell-cell signaling networks and response outputs leading to disrupted building blocks, incoherent multi-level organization, and loss of rigid structural motifs in mineralized tissues are factors that have been identified to participate in FD induction. In more recent years, novel insights into the unique biology of FD are transforming our understandings of its pathology, natural discourse of the disease, and treatment prospects. Herein, we built upon existing knowledge with recent findings to review clinical, etiologic, and histological features of FD and discussed known and potential mechanisms underlying FD manifestations. Subsequently, we ended on a note of optimism by highlighting emerging therapeutic approaches aimed at either halting or ameliorating disease progression.
Collapse
Affiliation(s)
- Ha-Young Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Jung-Hee Shim
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
- Department of Research Administration Team, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Chan-Yeong Heo
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| |
Collapse
|
2
|
Gaydosik AM, Stonesifer CJ, Tabib T, Lafyatis R, Geskin LJ, Fuschiotti P. The mycosis fungoides cutaneous microenvironment shapes dysfunctional cell trafficking, antitumor immunity, matrix interactions, and angiogenesis. JCI Insight 2023; 8:e170015. [PMID: 37669110 PMCID: PMC10619438 DOI: 10.1172/jci.insight.170015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/31/2023] [Indexed: 09/07/2023] Open
Abstract
Malignant T lymphocyte proliferation in mycosis fungoides (MF) is largely restricted to the skin, implying that malignant cells are dependent on their specific cutaneous tumor microenvironment (TME), including interactions with non-malignant immune and stromal cells, cytokines, and other immunomodulatory factors. To explore these interactions, we performed a comprehensive transcriptome analysis of the TME in advanced-stage MF skin tumors by single-cell RNA sequencing. Our analysis identified cell-type compositions, cellular functions, and cell-to-cell interactions in the MF TME that were distinct from those from healthy skin and benign dermatoses. While patterns of gene expression were common among patient samples, high transcriptional diversity was also observed in immune and stromal cells, with dynamic interactions and crosstalk between these cells and malignant T lymphocytes. This heterogeneity mapped to processes such as cell trafficking, matrix interactions, angiogenesis, immune functions, and metabolism that affect cancer cell growth, migration, and invasion, as well as antitumor immunity. By comprehensively characterizing the transcriptomes of immune and stromal cells within the cutaneous microenvironment of individual MF tumors, we have identified patterns of dysfunction common to all tumors that represent a resource for identifying candidates with therapeutic potential as well as patient-specific heterogeneity that has important implications for personalized disease management.
Collapse
Affiliation(s)
- Alyxzandria M. Gaydosik
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Tracy Tabib
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert Lafyatis
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Patrizia Fuschiotti
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
3
|
Uemura M, Sugimoto M, Yoshikawa Y, Inoue R. Electrical Shunting Prevents the Decline of Galvanotaxis After Monophasic Pulsed Microcurrent Stimulation in Human Dermal Fibroblasts. EPLASTY 2022; 22:e27. [PMID: 36000005 PMCID: PMC9361395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Electrical stimulation (ES) therapy is recommended for healing pressure injuries. Monophasic pulsed microcurrent stimulation promotes the migration of human dermal fibroblasts (HDFs) to the cathode, and ES potentially accelerates pressure injury healing. A reverse current is generated after ES in the human body; however, the effects of the electrical shunt in preventing the reverse current from migrating are unclear. Therefore, this study aimed to investigate the effects of an electrical shunt on the migration of HDFs. METHODS In the shunt groups, HDFs were electrically stimulated (0, 200, 400, and 600 µA) for 8 hours, and an electrical shunt was used to remove the electricity after ES. HDFs were observed under time-lapse microscopy for 24 hours. The migration ratio toward the cathode was calculated for each dish. RESULTS The migration ratio was significantly higher in the 200-µA group than in the other groups. HDFs migrated toward the anode after ES in the non-shunt groups with greater than 400 µA ES; however, HDFs did not migrate toward the anode with electrical shunting. CONCLUSIONS A post-ES electrical shunt is important in preventing a decline in the migration effect of ES.
Collapse
Affiliation(s)
- Mikiko Uemura
- Kansai University of Welfare Sciences, Faculty of Health Science, Department of Rehabilitation, Osaka, Japan
| | - Masaharu Sugimoto
- Toyama Rehabilitation Medical Health and Welfare College, Toyama, Toyama
| | - Yoshiyuki Yoshikawa
- Naragakuen University, Faculty of Heath Science, Department of Rehabilitation, Ikoma, Nara
| | - Rieko Inoue
- Department of Rehabilitation, Yoshida Hospital, Kobe, Japan
| |
Collapse
|
4
|
Pande G, Rai M, Sharma S, Agarwal V. Indigenous primary culture protocols for human adult skin fibroblast, pancreatic stellate cells, and peritoneal fibroblasts. INDIAN JOURNAL OF RHEUMATOLOGY 2021. [DOI: 10.4103/injr.injr_160_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
5
|
D'Urso M, Kurniawan NA. Mechanical and Physical Regulation of Fibroblast-Myofibroblast Transition: From Cellular Mechanoresponse to Tissue Pathology. Front Bioeng Biotechnol 2020; 8:609653. [PMID: 33425874 PMCID: PMC7793682 DOI: 10.3389/fbioe.2020.609653] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Fibroblasts are cells present throughout the human body that are primarily responsible for the production and maintenance of the extracellular matrix (ECM) within the tissues. They have the capability to modify the mechanical properties of the ECM within the tissue and transition into myofibroblasts, a cell type that is associated with the development of fibrotic tissue through an acute increase of cell density and protein deposition. This transition from fibroblast to myofibroblast-a well-known cellular hallmark of the pathological state of tissues-and the environmental stimuli that can induce this transition have received a lot of attention, for example in the contexts of asthma and cardiac fibrosis. Recent efforts in understanding how cells sense their physical environment at the micro- and nano-scales have ushered in a new appreciation that the substrates on which the cells adhere provide not only passive influence, but also active stimulus that can affect fibroblast activation. These studies suggest that mechanical interactions at the cell-substrate interface play a key role in regulating this phenotype transition by changing the mechanical and morphological properties of the cells. Here, we briefly summarize the reported chemical and physical cues regulating fibroblast phenotype. We then argue that a better understanding of how cells mechanically interact with the substrate (mechanosensing) and how this influences cell behaviors (mechanotransduction) using well-defined platforms that decouple the physical stimuli from the chemical ones can provide a powerful tool to control the balance between physiological tissue regeneration and pathological fibrotic response.
Collapse
Affiliation(s)
- Mirko D'Urso
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
6
|
Hosio M, Jaks V, Lagus H, Vuola J, Ogawa R, Kankuri E. Primary Ciliary Signaling in the Skin-Contribution to Wound Healing and Scarring. Front Cell Dev Biol 2020; 8:578384. [PMID: 33282860 PMCID: PMC7691485 DOI: 10.3389/fcell.2020.578384] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/08/2020] [Indexed: 12/21/2022] Open
Abstract
Primary cilia (PC) are solitary, post-mitotic, microtubule-based, and membrane-covered protrusions that are found on almost every mammalian cell. PC are specialized cellular sensory organelles that transmit environmental information to the cell. Signaling through PC is involved in the regulation of a variety of cellular processes, including proliferation, differentiation, and migration. Conversely, defective, or abnormal PC signaling can contribute to the development of various pathological conditions. Our knowledge of the role of PC in organ development and function is largely based on ciliopathies, a family of genetic disorders with mutations affecting the structure and function of PC. In this review, we focus on the role of PC in their major signaling pathways active in skin cells, and their contribution to wound healing and scarring. To provide comprehensive insights into the current understanding of PC functions, we have collected data available in the literature, including evidence across cell types, tissues, and animal species. We conclude that PC are underappreciated subcellular organelles that significantly contribute to both physiological and pathological processes of the skin development and wound healing. Thus, PC assembly and disassembly and PC signaling may serve as attractive targets for antifibrotic and antiscarring therapies.
Collapse
Affiliation(s)
- Mayu Hosio
- Faculty of Medicine, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Jyrki Vuola
- Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Esko Kankuri
- Faculty of Medicine, Department of Pharmacology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Nikoloudaki G, Snider P, Simmons O, Conway SJ, Hamilton DW. Periostin and matrix stiffness combine to regulate myofibroblast differentiation and fibronectin synthesis during palatal healing. Matrix Biol 2020; 94:31-56. [PMID: 32777343 DOI: 10.1016/j.matbio.2020.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022]
Abstract
Although the matricellular protein periostin is prominently upregulated in skin and gingival healing, it plays contrasting roles in myofibroblast differentiation and matrix synthesis respectively. Palatal healing is associated with scarring that can alter or restrict maxilla growth, but the expression pattern and contribution of periostin in palatal healing is unknown. Using periostin-knockout (Postn-/-) and wild-type (WT) mice, the contribution of periostin to palatal healing was investigated through 1.5 mm full-thickness excisional wounds in the hard palate. In WT mice, periostin was upregulated 6 days post-wounding, with mRNA levels peaking at day 12. Genetic deletion of periostin significantly reduced wound closure rates compared to WT mice. Absence of periostin reduced mRNA levels of pivotal genes in wound repair, including α-SMA/acta2, fibronectin and βigh3. Recruitment of fibroblasts and inflammatory cells, as visualized by immunofluorescent staining for fibroblast specific factor-1, vimentin, and macrophages markers Arginase-1 and iNOS was also impaired in Postn-/-, but not WT mice. Palatal fibroblasts isolated from the hard palate of mice were cultured on collagen gels and prefabricated silicon substrates with varying stiffness. Postn-/- fibroblasts showed a significantly reduced ability to contract a collagen gel, which was rescued by the exogenous addition of recombinant periostin. As the stiffness increased, Postn-/- fibroblasts increasingly differentiated into myofibroblasts, but not to the same degree as the WT. Pharmacological inhibition of Rac rescued the deficient myofibroblastic phenotype of Postn-/- cells. Low stiffness substrates (0.2 kPa) resulted in upregulation of fibronectin in WT cells, an effect which was significantly reduced in Postn-/- cells. Quantification of immunostaining for vinculin and integrinβ1 adhesions revealed that Periostin is required for the formation of focal and fibrillar adhesions in mPFBs. Our results suggest that periostin modulates myofibroblast differentiation and contraction via integrinβ1/RhoA pathway, and fibronectin synthesis in an ECM stiffness dependent manner in palatal healing.
Collapse
Affiliation(s)
- Georgia Nikoloudaki
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada.
| | - Paige Snider
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, Indianapolis, IN, United States
| | - Olga Simmons
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, Indianapolis, IN, United States
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut, Indianapolis, IN, United States
| | - Douglas W Hamilton
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada; Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada.
| |
Collapse
|
8
|
Nikoloudaki G, Creber K, Hamilton DW. Wound healing and fibrosis: a contrasting role for periostin in skin and the oral mucosa. Am J Physiol Cell Physiol 2020; 318:C1065-C1077. [PMID: 32267719 PMCID: PMC7311745 DOI: 10.1152/ajpcell.00035.2020] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 01/08/2023]
Abstract
Both skin and oral mucosa are characterized by the presence of keratinized epithelium in direct apposition to an underlying collagen-dense connective tissue. Despite significant overlap in structure and physiological function, skin and the oral mucosa exhibit significantly different healing profiles in response to injury. The oral mucosa has a propensity for rapid restoration of barrier function with minimal underlying fibrosis, but in contrast, skin is associated with slower healing and scar formation. Modulators of cell function, matricellular proteins have been shown to play significant roles in cutaneous healing, but their role in restoration of the oral mucosa is poorly defined. As will be discussed in this review, over the last 12 years our research group has been actively investigating the role of the profibrotic matricellular protein periostin in tissue homeostasis and fibrosis, as well as healing, in both skin and gingiva. In the skin, periostin is highly expressed in fibrotic scars and is upregulated during cutaneous wound repair, where it facilitates myofibroblast differentiation. In contrast, in gingival healing, periostin regulates extracellular matrix synthesis but does not appear to be associated with the transition of mesenchymal cells to a contractile phenotype. The significance of these findings will be discussed, with a focus on periostin as a potential therapeutic to augment healing of soft tissues or suppress fibrosis.
Collapse
Affiliation(s)
- Georgia Nikoloudaki
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Kendal Creber
- School of Biomedical Engineering, University of Western Ontario, London, Ontario, Canada
| | - Douglas W Hamilton
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
- School of Biomedical Engineering, University of Western Ontario, London, Ontario, Canada
- Division of Oral Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
Margetts PJ, Bonniaud P. Basic Mechanisms and Clinical Implications of Peritoneal Fibrosis. Perit Dial Int 2020. [DOI: 10.1177/089686080302300604] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Peter J. Margetts
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Philippe Bonniaud
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
10
|
Lee MO, Jung KB, Jo SJ, Hyun SA, Moon KS, Seo JW, Kim SH, Son MY. Modelling cardiac fibrosis using three-dimensional cardiac microtissues derived from human embryonic stem cells. J Biol Eng 2019; 13:15. [PMID: 30809271 PMCID: PMC6375184 DOI: 10.1186/s13036-019-0139-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Background Cardiac fibrosis is the most common pathway of many cardiac diseases. To date, there has been no suitable in vitro cardiac fibrosis model that could sufficiently mimic the complex environment of the human heart. Here, a three-dimensional (3D) cardiac sphere platform of contractile cardiac microtissue, composed of human embryonic stem cell (hESC)-derived cardiomyocytes (CMs) and mesenchymal stem cells (MSCs), is presented to better recapitulate the human heart. Results We hypothesized that MSCs would develop an in vitro fibrotic reaction in response to treatment with transforming growth factor-β1 (TGF-β1), a primary inducer of cardiac fibrosis. The addition of MSCs improved sarcomeric organization, electrophysiological properties, and the expression of cardiac-specific genes, suggesting their physiological relevance in the generation of human cardiac microtissue model in vitro. MSCs could also generate fibroblasts within 3D cardiac microtissues and, subsequently, these fibroblasts were transdifferentiated into myofibroblasts by the exogenous addition of TGF-β1. Cardiac microtissues displayed fibrotic features such as the deposition of collagen, the presence of numerous apoptotic CMs and the dissolution of mitochondrial networks. Furthermore, treatment with pro-fibrotic substances demonstrated that this model could reproduce key molecular and cellular fibrotic events. Conclusions This highlights the potential of our 3D cardiac microtissues as a valuable tool for manifesting and evaluating the pro-fibrotic effects of various agents, thereby representing an important step forward towards an in vitro system for the prediction of drug-induced cardiac fibrosis and the study of the pathological changes in human cardiac fibrosis. Electronic supplementary material The online version of this article (10.1186/s13036-019-0139-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mi-Ok Lee
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 341411 Republic of Korea
| | - Kwang Bo Jung
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 341411 Republic of Korea.,2Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Seong-Jae Jo
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 341411 Republic of Korea.,2Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Sung-Ae Hyun
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, Daejeon, 34114 Republic of Korea
| | - Kyoung-Sik Moon
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, Daejeon, 34114 Republic of Korea
| | - Joung-Wook Seo
- Research Group for Safety Pharmacology, Korea Institute of Toxicology, KRICT, Daejeon, 34114 Republic of Korea
| | - Sang-Heon Kim
- 4Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea.,5Department of Biomedical Engineering, KIST school, UST, Daejeon, 34113 Republic of Korea
| | - Mi-Young Son
- 1Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 341411 Republic of Korea.,2Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| |
Collapse
|
11
|
Hortells L, Sur S, St Hilaire C. Cell Phenotype Transitions in Cardiovascular Calcification. Front Cardiovasc Med 2018; 5:27. [PMID: 29632866 PMCID: PMC5879740 DOI: 10.3389/fcvm.2018.00027] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/14/2018] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular calcification was originally considered a passive, degenerative process, however with the advance of cellular and molecular biology techniques it is now appreciated that ectopic calcification is an active biological process. Vascular calcification is the most common form of ectopic calcification, and aging as well as specific disease states such as atherosclerosis, diabetes, and genetic mutations, exhibit this pathology. In the vessels and valves, endothelial cells, smooth muscle cells, and fibroblast-like cells contribute to the formation of extracellular calcified nodules. Research suggests that these vascular cells undergo a phenotypic switch whereby they acquire osteoblast-like characteristics, however the mechanisms driving the early aspects of these cell transitions are not fully understood. Osteoblasts are true bone-forming cells and differentiate from their pluripotent precursor, the mesenchymal stem cell (MSC); vascular cells that acquire the ability to calcify share aspects of the transcriptional programs exhibited by MSCs differentiating into osteoblasts. What is unknown is whether a fully-differentiated vascular cell directly acquires the ability to calcify by the upregulation of osteogenic genes or, whether these vascular cells first de-differentiate into an MSC-like state before obtaining a “second hit” that induces them to re-differentiate down an osteogenic lineage. Addressing these questions will enable progress in preventative and regenerative medicine strategies to combat vascular calcification pathologies. In this review, we will summarize what is known about the phenotypic switching of vascular endothelial, smooth muscle, and valvular cells.
Collapse
Affiliation(s)
- Luis Hortells
- Division of Cardiology, Department of Medicine, and the Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Swastika Sur
- Division of Cardiology, Department of Medicine, and the Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, and the Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Klesen A, Jakob D, Emig R, Kohl P, Ravens U, Peyronnet R. Cardiac fibroblasts : Active players in (atrial) electrophysiology? Herzschrittmacherther Elektrophysiol 2018; 29:62-69. [PMID: 29392412 DOI: 10.1007/s00399-018-0553-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 06/07/2023]
Abstract
Fibrotic areas in cardiac muscle-be it in ventricular or atrial tissue-are considered as obstacles for conduction of the excitatory wave and can therefore facilitate re-entry, which may contribute to the sustenance of cardiac arrhythmias. Persistence of one of the most frequent arrhythmias, atrial fibrillation (AF), is accompanied by enhanced atrial fibrosis. Any kind of myocardial perturbation, whether via mechanical stress or ischemic damage, inflammation, or irregular and high-frequency electrical activity, activates fibroblasts. This leads to the secretion of paracrine factors and extracellular matrix proteins, especially collagen, and to the differentiation of fibroblasts into myofibroblasts. Excessive collagen production is the hallmark of fibrosis and impairs regular impulse propagation. In addition, direct electrical coupling between cardiomyocytes and nonmyocytes, such as fibroblasts and macrophages, via gap junctions affects conduction. Although fibroblasts are not electrically excitable, they express functional ion channels, in particular K+ channels and mechanosensitive channels, some of which could be involved in tissue remodeling. Here, we briefly review these aspects with special reference to AF.
Collapse
Affiliation(s)
- Alexander Klesen
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Dorothee Jakob
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2q, 79110, Freiburg i. Br., Germany.
- Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany.
| |
Collapse
|
13
|
Mattyasovszky SG, Mausbach S, Ritz U, Langendorf E, Wollstädter J, Baranowski A, Drees P, Rommens PM, Hofmann A. Influence of the anti-inflammatory cytokine interleukin-4 on human joint capsule myofibroblasts. J Orthop Res 2017; 35:1290-1298. [PMID: 27504740 DOI: 10.1002/jor.23386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 08/05/2016] [Indexed: 02/04/2023]
Abstract
Post-traumatic joint contracture was reported to be associated with elevated numbers of contractile myofibroblasts (MFs) in the healing capsule. During the physiological healing process, the number of MFs declines; however, in fibroconnective disorders, MFs persist. The manifold interaction of the cytokines regulating the appearance and persistence of MFs in the pathogenesis of joint contracture remains to be elucidated. The objective of our current study was to analyze the impact of the anti-inflammatory cytokine interleukin (IL)-4 on functional behavior of MFs. Cells were isolated from human joint capsule specimens and challenged with three different concentrations of IL-4 with or without its neutralizing antibody. MF viability, contractile properties, and the gene expression of both alpha-smooth muscle actin (α-SMA) and collagen type I were examined. Immunofluorescence staining revealed the presence of IL-4 receptor (R)-alpha (α) on the membrane of cultured MFs. The cytokine IL-4 promoted MF viability and enhanced MF modulated contraction of collagen gels. Moreover, IL-4 intervened in gene expression by up-regulation of α-SMA and collagen type I mRNA. These effects could be specifically lowered by the neutralizing IL-4 antibody. On the basis of our findings we conclude that the anti-inflammatory cytokine IL-4 specifically regulates viability and the contractile properties of MFs via up-regulating the gene expression of α-SMA and collagen type I. IL-4 may be a helpful target in developing anti-fibrotic therapeutics for post-traumatic joint contracture in human. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1290-1298, 2017.
Collapse
Affiliation(s)
- Stefan G Mattyasovszky
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Stefan Mausbach
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Eva Langendorf
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Jochen Wollstädter
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Andreas Baranowski
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Phillipp Drees
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
14
|
Liu DQ, Li XJ, Weng XJ. Effect of BTXA on Inhibiting Hypertrophic Scar Formation in a Rabbit Ear Model. Aesthetic Plast Surg 2017; 41:721-728. [PMID: 28180922 DOI: 10.1007/s00266-017-0803-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/17/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND Hypertrophic scar (HS) is a refractory skin disease caused by major physical damage or other inflammation. Some reports found that botulinum toxin type A (BTXA) could be an alternative treatment of the HS. Therefore, the authors studied the effects of BTXA on the treatment of HS and the dose response of BTXA. METHODS Hypertrophic scars were harvested from the ears of 18 young adult New Zealand big-eared rabbits and treated with BTXA or triamcinolone acetonide (TAC) in vivo experiment. The hypertrophic index (HI) was measured by histological examination. Collagen fibrils were checked by sirius red straining, and the cell nucleuses of fibroblasts were checked by Ki67. RESULTS The HI of hypertrophic scars with BTXA treatment was lower than that with phosphate-buffered saline treatment (P < 0.05). Compared with the TAC treatment group, the efficacy of treatment with the middle dose of BTXA (1.0, 1.5 IU) had no significant difference, as shown by sirius red staining and immunohistochemistry Ki67. CONCLUSION These results demonstrated that BTXA effectively improved the appearance of hypertrophic scars and inhibited the formation of collagen fibrils and fibroblasts in vivo. Treatment with the middle dose of BTXA achieved similar efficacy as TAC treatment, indicating that BTXA might be useful for inhibiting hypertrophic scars and worth investigating further. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Dong-Qing Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Anhui Medical University, 218# Ji-Xi Road, Hefei, 230022, Anhui, China
| | - Xiao-Jing Li
- Department of Plastic Surgery, The First Affiliated Hospital of Anhui Medical University, 218# Ji-Xi Road, Hefei, 230022, Anhui, China.
| | - Xiao-Juan Weng
- Department of Plastic Surgery, The Fourth Affiliated Hospital of Anhui Medical University, 372# Tun-Xi Road, Hefei, 230023, Anhui, China
| |
Collapse
|
15
|
Lin RJ, Su ZZ, Liang SM, Chen YY, Shu XR, Nie RQ, Wang JF, Xie SL. Role of Circulating Fibrocytes in Cardiac Fibrosis. Chin Med J (Engl) 2017; 129:326-31. [PMID: 26831236 PMCID: PMC4799578 DOI: 10.4103/0366-6999.174503] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE It is revealed that circulating fibrocytes are elevated in patients/animals with cardiac fibrosis, and this review aims to provide an introduction to circulating fibrocytes and their role in cardiac fibrosis. DATA SOURCES This review is based on the data from 1994 to present obtained from PubMed. The search terms were "circulating fibrocytes " and "cardiac fibrosis ". STUDY SELECTION Articles and critical reviews, which are related to circulating fibrocytes and cardiac fibrosis, were selected. RESULTS Circulating fibrocytes, which are derived from hematopoietic stem cells, represent a subset of peripheral blood mononuclear cells exhibiting mixed morphological and molecular characteristics of hematopoietic and mesenchymal cells (CD34+/CD45+/collagen I+). They can produce extracellular matrix and many cytokines. It is shown that circulating fibrocytes participate in many fibrotic diseases, including cardiac fibrosis. Evidence accumulated in recent years shows that aging individuals and patients with hypertension, heart failure, coronary heart disease, and atrial fibrillation have more circulating fibrocytes in peripheral blood and/or heart tissue, and this elevation of circulating fibrocytes is correlated with the degree of fibrosis in the hearts. CONCLUSIONS Circulating fibrocytes are effector cells in cardiac fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuang-Lun Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong 510120, China
| |
Collapse
|
16
|
Wang X, Cao P, Liu J, Du P, Wang Z, Chen W, Liu C, Wu Y. 5-Aminolaevulinic Acid-Based Photodynamic Therapy Restrains Pathological Hyperplasia of Fibroblasts. Med Sci Monit 2017; 23:46-56. [PMID: 28052053 PMCID: PMC5228760 DOI: 10.12659/msm.898221] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background This study aimed to explore whether 5-aminolaevulinic acid-based photodynamic therapy (ALA-PDT) restrains pathological hyperplasia of fibroblasts from hyperplastic scar tissues, and to investigate the potential mechanism. Material/Methods We used MTT assay, flow cytometry, and terminal-deoxynucleotidyl transferase mediated nick-end labeling (TUNEL) to examine the effects of ALA-PDT on proliferation, cell cycle, and apoptosis of fibroblasts isolated from hyperplastic scar tissues. The growth-promoting effect of fibroblasts on vascular endothelial cells was measured by cell co-culture. Real-time PCR and Western blot analysis were performed to detect the expression levels of transforming growth factor-β1 (TGF-β1), α-smooth muscle actin (α-SMA), Collagen I, Collagen III, vascular endothelial growth factor-A (VEGFA), and basic fibroblast growth factor (bFGF). Results ALA-PDT inhibited proliferation delayed cell cycle progress, promoted apoptosis of fibroblasts, and suppressed its growth-promoting effect on vascular endothelial cells, and decreased expression of TGF-β1, α-SMA, Collagen I, Collagen III, VEGFA, and bFGF. Conclusions ALA-PDT effectively restrained pathological hyperplasia of fibroblasts from hyperplastic scar tissues, which may provide a research basis for clinical therapy of hyperplastic scars.
Collapse
Affiliation(s)
- Xiaochuan Wang
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Ping Cao
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Jian Liu
- Department of Hepatobiliary Surgery, The First People's Hospital of Kunming, Kunming, Yunnan, China (mainland)
| | - Peng Du
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Zhiqiong Wang
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| | - Wei Chen
- Department of Genetics, Kunming University of Science and Technology, Kunming, Yunnan, China (mainland)
| | - Chang Liu
- Department of Clinical Medicine, Kunming University of Traditional Chinese Medicine, Kunming, China (mainland)
| | - Yifei Wu
- Department of Dermatology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China (mainland)
| |
Collapse
|
17
|
Abstract
Cardiac stress can induce morphological, structural and functional changes of the heart, referred to as cardiac remodeling. Myocardial infarction or sustained overload as a result of pathological causes such as hypertension or valve insufficiency may result in progressive remodeling and finally lead to heart failure (HF). Whereas pathological and physiological (exercise, pregnancy) overload both stimulate cardiomyocyte growth (hypertrophy), only pathological remodeling is characterized by increased deposition of extracellular matrix proteins, termed fibrosis, and loss of cardiomyocytes by necrosis, apoptosis and/or phagocytosis. HF is strongly associated with age, and cardiomyocyte loss and fibrosis are typical signs of the aging heart. Fibrosis results in stiffening of the heart, conductivity problems and reduced oxygen diffusion, and is associated with diminished ventricular function and arrhythmias. As a consequence, the workload of cardiomyocytes in the fibrotic heart is further augmented, whereas the physiological environment is becoming less favorable. This causes additional cardiomyocyte death and replacement of lost cardiomyocytes by fibrotic material, generating a vicious cycle of further decline of cardiac function. Breaking this fibrosis-cell death axis could halt further pathological and age-related cardiac regression and potentially reverse remodeling. In this review, we will describe the interaction between cardiac fibrosis, cardiomyocyte hypertrophy and cell death, and discuss potential strategies for tackling progressive cardiac remodeling and HF.
Collapse
Affiliation(s)
- A Piek
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - R A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands
| | - H H W Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713GZ, Groningen, The Netherlands.
| |
Collapse
|
18
|
Roche PL, Nagalingam RS, Bagchi RA, Aroutiounova N, Belisle BMJ, Wigle JT, Czubryt MP. Role of scleraxis in mechanical stretch-mediated regulation of cardiac myofibroblast phenotype. Am J Physiol Cell Physiol 2016; 311:C297-307. [PMID: 27357547 DOI: 10.1152/ajpcell.00333.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 06/27/2016] [Indexed: 12/21/2022]
Abstract
The phenotype conversion of fibroblasts to myofibroblasts plays a key role in the pathogenesis of cardiac fibrosis. Numerous triggers of this conversion process have been identified, including plating of cells on solid substrates, cytokines such as transforming growth factor-β, and mechanical stretch; however, the underlying mechanisms remain incompletely defined. Recent studies from our laboratory revealed that the transcription factor scleraxis is a key regulator of cardiac fibroblast phenotype and extracellular matrix expression. Here we report that mechanical stretch induces type I collagen expression and morphological changes indicative of cardiac myofibroblast conversion, as well as scleraxis expression via activation of the scleraxis promoter. Scleraxis causes phenotypic changes similar to stretch, and the effect of stretch is attenuated in scleraxis null cells. Scleraxis was also sufficient to upregulate expression of vinculin and F-actin, to induce stress fiber and focal adhesion formation, and to attenuate both cell migration and proliferation, further evidence of scleraxis-mediated regulation of fibroblast to myofibroblast conversion. Together, these data confirm that scleraxis is sufficient to promote the myofibroblast phenotype and is a required effector of stretch-mediated conversion. Scleraxis may thus represent a potential target for the development of novel antifibrotic therapies aimed at inhibiting myofibroblast formation.
Collapse
Affiliation(s)
- Patricia L Roche
- St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; and
| | - Raghu S Nagalingam
- St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; and
| | - Rushita A Bagchi
- St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; and
| | - Nina Aroutiounova
- St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Breanna M J Belisle
- St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jeffrey T Wigle
- St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael P Czubryt
- St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; and
| |
Collapse
|
19
|
Bonaventura A, Montecucco F, Dallegri F. Cellular recruitment in myocardial ischaemia/reperfusion injury. Eur J Clin Invest 2016; 46:590-601. [PMID: 27090739 DOI: 10.1111/eci.12633] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 04/17/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is strictly linked to atherosclerosis. Beyond the mechanical narrowing of coronary vessels lumen, during MI a great burden of inflammation is carried out. One of the crucial events is represented by the ischaemia/reperfusion injury, a complex event involving inflammatory cells (such as neutrophils, platelets, monocytes/macrophages, lymphocytes and mast cells) and key activating signals (such as cytokines, chemokines and growth factors). Cardiac repair following myocardial infarction is dependent on a finely regulated response involving a sequential recruitment and the clearance of different subsets of inflammatory cells. MATERIALS AND METHODS This narrative review was based on the works detected on PubMed and MEDLINE up to November 2015. RESULTS Infarct healing classically follows three overlapping phases: the inflammatory phase, in which the innate immune pathways are activated and inflammatory leucocytes are recruited in order to clear the wound from dead cells; the proliferative phase, characterized by the suppression of pro-inflammatory signalling and infiltration of 'repairing' cells secreting matrix proteins in the injured area; and the maturation phase, which is associated with the quiescence and the elimination of the reparative cells together with cross-linking of the matrix. All these phases are timely regulated by the production of soluble mediators, such as cytokines, chemokines and growth factors. CONCLUSION Targeting inflammatory cell recruitment early during reperfusion and healing might be promising to selectively inhibit injury and favour repair. This approach might substantially improve adverse postischaemic left ventricle remodelling, characterized by dilation, hypertrophy of viable segments and progressive dysfunction.
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS AOU San Martino - IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| |
Collapse
|
20
|
Mishra PJ, Mishra PJ, Banerjee D. Keratinocyte Induced Differentiation of Mesenchymal Stem Cells into Dermal Myofibroblasts: A Role in Effective Wound Healing. ACTA ACUST UNITED AC 2016; 2016:5-32. [PMID: 27294075 DOI: 10.13052/ijts2246-8765.2016.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We have previously demonstrated that human mesenchymal stem cells (hMSCs) migrate toward human keratinocytes as well as toward conditioned medium from cultured human keratinocytes (KCM) indicating that the hMSCs respond to signals from keratinocytes [1]. Using fluorescently labeled cells we now show that in vitro hMSCs appear to surround keratinocytes, and this organization is recapitulated in vivo. Incubation of hMSCs with KCM induced dermal myofibroblast like differentiation characterized by expression of cytoskeletal markers and increased expression of cytokines including SDF-1, IL-8, IL-6 and CXCL5. Interaction of keratinocytes with hMSCs appears to be important in the wound healing process. Therapeutic efficacy of hMSCs in wound healing was examined in two animal models representing normal and chronic wound healing. Accelerated wound healing was observed when hMSCs and KCM exposed hMSCs (KCMSCs) were injected near wound site in nude and NOD/SCID mice. Long term follow up of wound healing revealed that in the hMSC treated wounds there was little evidence of residual scarring. These dermal myofibroblast like hMSCs add to the wound healing process. Together, the keratinocyte and hMSCs morphed dermal myofibroblast like cells as well as the factors secreted by these cells support wound healing with minimal scarring. The ability of hMSCs to support wound healing process represents another striking example of the importance of keratinocyte and hMSCs interplay in the wound microenvironment resulting in effective wound healing with minimal scarring.
Collapse
Affiliation(s)
- Pravin J Mishra
- Intermountain Precision Genomics, Intermountain Healthcare, Dixie Regional Medical Center 292 South 1470 East, Suite 201 & 301, St. George, UT 84770, USA
| | - Prasun J Mishra
- Department of Biochemical and Cellular Pharmacology, Genentech, 1, DNA Way, South San Francisco, California 94080, USA
| | - Debabrata Banerjee
- Department of Pharmacology, Robert Wood Johnson Medical School, Graduate School of Biomedical Sciences, New Brunswick-Piscataway, Rutgers University, 675 Hoes Lane West, Piscataway, NJ 08854. USA
| |
Collapse
|
21
|
Zhang E, Gao B, Yang L, Wu X, Wang Z. Notoginsenoside Ft1 Promotes Fibroblast Proliferation via PI3K/Akt/mTOR Signaling Pathway and Benefits Wound Healing in Genetically Diabetic Mice. J Pharmacol Exp Ther 2015; 356:324-32. [PMID: 26567319 DOI: 10.1124/jpet.115.229369] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
Wound healing requires the essential participation of fibroblasts, which is impaired in diabetic foot ulcers (DFU). Notoginsenoside Ft1 (Ft1), a saponin from Panax notoginseng, can enhance platelet aggregation by activating signaling network mediated through P2Y12 and induce proliferation, migration, and tube formation in cultured human umbilical vein endothelial cells. However, whether it can accelerate fibroblast proliferation and benefit wound healing, especially DFU, has not been elucidated. In the present study on human dermal fibroblast HDF-a, Ft1 increased cell proliferation and collagen production via PI3K/Akt/mTOR signaling pathway. On the excisional wound splinting model established on db/db diabetic mouse, topical application of Ft1 significantly shortened the wound closure time by 5.1 days in contrast with phosphate-buffered saline (PBS) treatment (15.8 versus 20.9 days). Meanwhile, Ft1 increased the rate of re-epithelialization and the amount of granulation tissue at day 7 and day 14. The molecule also enhanced mRNA expressions of COL1A1, COL3A1, transforming growth factor (TGF)-β1 and TGF-β3 and fibronectin, the genes that contributed to collagen expression, fibroblast proliferation, and consequent scar formation. Moreover, Ft1 facilitated the neovascularization accompanied with elevated vascular endothelial growth factor, platelet-derived growth factor, and fibroblast growth factor at either mRNA or protein levels and alleviated the inflammation of infiltrated monocytes indicated by reduced tumor necrosis factor-α and interleukin-6 mRNA expressions in the diabetic wounds. Altogether, these results indicated that Ft1 might accelerate diabetic wound healing by orchestrating multiple processes, including promoting fibroblast proliferation, enhancing angiogenesis, and attenuating inflammatory response, which provided a great potential application of it in clinics for patients with DFU.
Collapse
Affiliation(s)
- Eryun Zhang
- Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China (E.Z, Z.W.); and Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China (B.G., L.Y., X.W., Z.W.)
| | - Bo Gao
- Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China (E.Z, Z.W.); and Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China (B.G., L.Y., X.W., Z.W.)
| | - Li Yang
- Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China (E.Z, Z.W.); and Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China (B.G., L.Y., X.W., Z.W.)
| | - Xiaojun Wu
- Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China (E.Z, Z.W.); and Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China (B.G., L.Y., X.W., Z.W.)
| | - Zhengtao Wang
- Department of Pharmacognosy, China Pharmaceutical University, Nanjing, China (E.Z, Z.W.); and Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China (B.G., L.Y., X.W., Z.W.)
| |
Collapse
|
22
|
Mesenchymal-epithelial interactions during digestive tract development and epithelial stem cell regeneration. Cell Mol Life Sci 2015; 72:3883-96. [PMID: 26126787 DOI: 10.1007/s00018-015-1975-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 12/16/2022]
Abstract
The gastrointestinal tract develops from a simple and uniform tube into a complex organ with specific differentiation patterns along the anterior-posterior and dorso-ventral axes of asymmetry. It is derived from all three germ layers and their cross-talk is important for the regulated development of fetal and adult gastrointestinal structures and organs. Signals from the adjacent mesoderm are essential for the morphogenesis of the overlying epithelium. These mesenchymal-epithelial interactions govern the development and regionalization of the different gastrointestinal epithelia and involve most of the key morphogens and signaling pathways, such as the Hedgehog, BMPs, Notch, WNT, HOX, SOX and FOXF cascades. Moreover, the mechanisms underlying mesenchyme differentiation into smooth muscle cells influence the regionalization of the gastrointestinal epithelium through interactions with the enteric nervous system. In the neonatal and adult gastrointestinal tract, mesenchymal-epithelial interactions are essential for the maintenance of the epithelial regionalization and digestive epithelial homeostasis. Disruption of these interactions is also associated with bowel dysfunction potentially leading to epithelial tumor development. In this review, we will discuss various aspects of the mesenchymal-epithelial interactions observed during digestive epithelium development and differentiation and also during epithelial stem cell regeneration.
Collapse
|
23
|
Dostal D, Glaser S, Baudino TA. Cardiac Fibroblast Physiology and Pathology. Compr Physiol 2015; 5:887-909. [DOI: 10.1002/cphy.c140053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
24
|
Qiu JY, Liu P, Shi C, Han B. Low-grade myofibroblastic sarcomas of the maxilla. Oncol Lett 2014; 9:619-625. [PMID: 25624890 PMCID: PMC4301534 DOI: 10.3892/ol.2014.2790] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 11/12/2014] [Indexed: 11/22/2022] Open
Abstract
Low-grade myofibroblastic sarcoma (LGMS) is a distinct mesenchymal myofibroblastic malignancy. The tumor may occur at a variety of sites, but is particularly associated with the head and neck. Of the two maxillary sarcomas that were analyzed in the present study, one was misdiagnosed as an inflammatory myofibroblastic tumor during pre-operative excision biopsy, and later presented with a different immunophenotype upon recurrence. Representative paraffin blocks from formalin-fixed tissues were selected from each patient and designated as case 1 and case 2. Immunohistochemical studies were performed on 3-μm thick sections using primary antibodies against α-smooth muscle actin (α-SMA), muscle-specific actin (MSA), desmin, vimentin, calponin, h-caldesmon, fibronectin, cytokeratin, cluster of differentiation 34 (CD34), S-100 protein, anaplastic lymphoma kinase (ALK), epithelial membrane antigen (EMA) and Ki-67. Immunohistochemistry was performed using the streptavidin-biotin-peroxidase complex method. The tumor cells from the two maxillary LGMSs, including the recurrent lesion, were positive for vimentin and fibronectin, and negative for S-100 protein, CD34, EMA, h-caldesmon, ALK, MSA and calponin. The tumor cells from case 1 demonstrated positive staining for α-SMA protein and negative staining for desmin. By contrast, the tumor cells from the primary lesion in case 2 presented with negative staining for α-SMA and positive staining for desmin, while the cells of the recurrent lesion were α-SMA-positive and desmin-negative. The present study concluded that cases of LGMS with immunoprofile alterations are predictive of relatively poor prognoses.
Collapse
Affiliation(s)
- Jin-Yu Qiu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, P.R. China
| | - Peng Liu
- Stomatology Department, School of Medicine, Yanbian University, Yanji, Jilin, P.R. China
| | - Ce Shi
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, P.R. China
| | - Bing Han
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, P.R. China
| |
Collapse
|
25
|
RUNX1 is essential for mesenchymal stem cell proliferation and myofibroblast differentiation. Proc Natl Acad Sci U S A 2014; 111:16389-94. [PMID: 25313057 DOI: 10.1073/pnas.1407097111] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Myofibroblasts are a key cell type in wound repair, cardiovascular disease, and fibrosis and in the tumor-promoting microenvironment. The high accumulation of myofibroblasts in reactive stroma is predictive of the rate of cancer progression in many different tumors, yet the cell types of origin and the mechanisms that regulate proliferation and differentiation are unknown. We report here, for the first time to our knowledge, the characterization of normal human prostate-derived mesenchymal stem cells (MSCs) and the TGF-β1-regulated pathways that modulate MSC proliferation and myofibroblast differentiation. Human prostate MSCs combined with prostate cancer cells expressing TGF-β1 resulted in commitment to myofibroblasts. TGF-β1-regulated runt-related transcription factor 1 (RUNX1) was required for cell cycle progression and proliferation of progenitors. RUNX1 also inhibited, yet did not block, differentiation. Knockdown of RUNX1 in prostate or bone marrow-derived MSCs resulted in cell cycle arrest, attenuated proliferation, and constitutive differentiation to myofibroblasts. These data show that RUNX1 is a key transcription factor for MSC proliferation and cell fate commitment in myofibroblast differentiation. This work also shows that the normal human prostate gland contains tissue-derived MSCs that exhibit multilineage differentiation similar to bone marrow-derived MSCs. Targeting RUNX1 pathways may represent a therapeutic approach to affect myofibroblast proliferation and biology in multiple disease states.
Collapse
|
26
|
Kramann R, DiRocco DP, Humphreys BD. Understanding the origin, activation and regulation of matrix-producing myofibroblasts for treatment of fibrotic disease. J Pathol 2013; 231:273-89. [PMID: 24006178 DOI: 10.1002/path.4253] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 08/26/2013] [Indexed: 12/19/2022]
Abstract
Fibrosis and scar formation results from chronic progressive injury in virtually every tissue and affects a growing number of people around the world. Myofibroblasts drive fibrosis, and recent work has demonstrated that mesenchymal cells, including pericytes and perivascular fibroblasts, are their main progenitors. Understanding the cellular mechanisms of pericyte/fibroblast-to-myofibroblast transition, myofibroblast proliferation and the key signalling pathways that regulate these processes is essential to develop novel targeted therapeutics for the growing patient population suffering from solid organ fibrosis. In this review, we summarize the current knowledge about different progenitor cells of myofibroblasts, discuss major pathways that regulate their transdifferentiation and discuss the current status of novel targeted anti-fibrotic therapeutics in development.
Collapse
Affiliation(s)
- Rafael Kramann
- Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; RWTH Aachen University, Division of Nephrology, Aachen, Germany
| | | | | |
Collapse
|
27
|
Jivraj N, Phinikaridou A, Shah AM, Botnar RM. Molecular imaging of myocardial infarction. Basic Res Cardiol 2013; 109:397. [PMID: 24322905 DOI: 10.1007/s00395-013-0397-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 11/14/2013] [Accepted: 11/27/2013] [Indexed: 11/29/2022]
Abstract
Myocardial infarction (MI), and subsequent heart failure, remains a major healthcare problem in the western and developing world and leads to substantial morbidity and mortality. After MI, the ability of the myocardium to recover is closely associated with a complex immune response that often leads to adverse remodeling of the ventricle, and poor prognosis. Currently used clinical imaging modalities allow the assessment of anatomy, perfusion, function, and viability but do not provide insights into specific biological processes. In contrast, novel non-invasive imaging methods, using targeted imaging agents, allow imaging of the molecular processes underlying the post-MI immune cell response, and subsequent remodeling. Therefore, this may have significant diagnostic, prognostic, and therapeutic value, and may help to improve our understanding of post-infarct remodeling, in vivo. Imaging modalities such as magnetic resonance imaging, single-photon emission computed tomography, and positron emission tomography have been used in concert with radiolabelled and (super) paramagnetic probes to image each phase of the immune response. These probes, which target apoptosis, necrosis, neutrophils, monocytes, enzymes, angiogenesis, extracellular matrix, and scar formation have been assessed and validated pre-clinically. Translating this work to the bedside in a cost-effective, clinically beneficial manner remains a significant challenge. This article reviews these new imaging techniques as well as the corresponding pathophysiology.
Collapse
Affiliation(s)
- Naheed Jivraj
- Division of Imaging Sciences and Biomedical Engineering, King's College London, St. Thomas' Hospital, 4th Floor, Lambeth Wing, London, SE1 7EH, UK,
| | | | | | | |
Collapse
|
28
|
Cunnington RH, Northcott JM, Ghavami S, Filomeno KL, Jahan F, Kavosh MS, Davies JJL, Wigle JT, Dixon IMC. The Ski-Zeb2-Meox2 pathway provides a novel mechanism for regulation of the cardiac myofibroblast phenotype. J Cell Sci 2013; 127:40-9. [PMID: 24155330 DOI: 10.1242/jcs.126722] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cardiac fibrosis is linked to fibroblast-to-myofibroblast phenoconversion and proliferation but the mechanisms underlying this are poorly understood. Ski is a negative regulator of TGF-β-Smad signaling in myofibroblasts, and might redirect the myofibroblast phenotype back to fibroblasts. Meox2 could alter TGF-β-mediated cellular processes and is repressed by Zeb2. Here, we investigated whether Ski diminishes the myofibroblast phenotype by de-repressing Meox2 expression and function through repression of Zeb2 expression. We show that expression of Meox1 and Meox2 mRNA and Meox2 protein is reduced during phenoconversion of fibroblasts to myofibroblasts. Overexpression of Meox2 shifts the myofibroblasts into fibroblasts, whereas the Meox2 DNA-binding mutant has no effect on myofibroblast phenotype. Overexpression of Ski partially restores Meox2 mRNA expression levels to those in cardiac fibroblasts. Expression of Zeb2 increased during phenoconversion and Ski overexpression reduces Zeb2 expression in first-passage myofibroblasts. Furthermore, expression of Meox2 is decreased in scar following myocardial infarction, whereas Zeb2 protein expression increases in the infarct scar. Thus Ski modulates the cardiac myofibroblast phenotype and function through suppression of Zeb2 by upregulating the expression of Meox2. This cascade might regulate cardiac myofibroblast phenotype and presents therapeutic options for treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Ryan H Cunnington
- Department of Physiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Remy C, Jacquemin D, Massage P, Damas P, Rousseau AF. La prise en charge précoce du patient brûlé en kinésithérapie. MEDECINE INTENSIVE REANIMATION 2013. [DOI: 10.1007/s13546-013-0709-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Humphreys BD. Targeting pericyte differentiation as a strategy to modulate kidney fibrosis in diabetic nephropathy. Semin Nephrol 2013; 32:463-70. [PMID: 23062987 DOI: 10.1016/j.semnephrol.2012.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Pericytes are a heterogeneous group of extensively branched cells located in microvessels where they make focal contacts with endothelium. Pericytes stabilize blood vessels, regulate vascular tone, synthesize matrix, participate in repair, and serve as progenitor cells, among other functions. Recent work has highlighted the role of pericytes and pericyte-like cells in fibrosis, in which chronic injury triggers pericyte proliferation and differentiation into collagen-secretory, contractile myofibroblasts with migration away from vessels, causing microvascular rarefaction. In this review the developmental origins of kidney pericytes and perivascular fibroblasts are summarized, pericyte to myofibroblast transition in type I diabetic nephropathy is discussed, and the regulation of pericyte differentiation into myofibroblasts as a therapeutic target for treatment of diabetic nephropathy is described.
Collapse
Affiliation(s)
- Benjamin D Humphreys
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
31
|
Lee SY, Ryan SJ. Pathophysiology of Ocular Trauma. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00098-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
32
|
Karagiannis GS, Poutahidis T, Erdman SE, Kirsch R, Riddell RH, Diamandis EP. Cancer-associated fibroblasts drive the progression of metastasis through both paracrine and mechanical pressure on cancer tissue. Mol Cancer Res 2012; 10:1403-18. [PMID: 23024188 DOI: 10.1158/1541-7786.mcr-12-0307] [Citation(s) in RCA: 405] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neoplastic cells recruit fibroblasts through various growth factors and cytokines. These "cancer-associated fibroblasts" (CAF) actively interact with neoplastic cells and form a myofibroblastic microenvironment that promotes cancer growth and survival and supports malignancy. Several products of their paracrine signaling repertoire have been recognized as tumor growth and metastasis regulators. However, tumor-promoting cell signaling is not the only reason that makes CAFs key components of the "tumor microenvironment," as CAFs affect both the architecture and growth mechanics of the developing tumor. CAFs participate in the remodeling of peritumoral stroma, which is a prerequisite of neoplastic cell invasion, expansion, and metastasis. CAFs are not present peritumorally as individual cells but they act orchestrated to fully deploy a desmoplastic program, characterized by "syncytial" (or collective) configuration and altered cell adhesion properties. Such myofibroblastic cohorts are reminiscent of those encountered in wound-healing processes. The view of "cancer as a wound that does not heal" led to useful comparisons between wound healing and tumorigenesis and expanded our knowledge of the role of CAF cohorts in cancer. In this integrative model of cancer invasion and metastasis, we propose that the CAF-supported microenvironment has a dual tumor-promoting role. Not only does it provide essential signals for cancer cell dedifferentiation, proliferation, and survival but it also facilitates cancer cell local invasion and metastatic phenomena.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Proteomic signatures of the desmoplastic invasion front reveal collagen type XII as a marker of myofibroblastic differentiation during colorectal cancer metastasis. Oncotarget 2012; 3:267-85. [PMID: 22408128 PMCID: PMC3359884 DOI: 10.18632/oncotarget.451] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs), represent a pivotal compartment of solid cancers (desmoplasia), and are causatively implicated in cancer development and progression. CAFs are recruited by growth factors secreted by cancer cells and they present a myofibroblastic phenotype, similar to the one obtained by resident fibroblasts during wound healing. Paracrine signaling between cancer cells and CAFs results in a unique protein expression profile in areas of desmoplastic reaction, which is speculated to drive metastasis. In an attempt to decipher large-scale proteomic profiles of the cancer invasive margins, we developed an in vitro coculture model system, based on tumor-host cell interactions between colon cancer cells and CAFs. Proteomic analysis of conditioned media derived from these cocultures coupled to mass spectrometry and bioinformatic analysis was performed to uncover myofibroblastic signatures of the cancer invasion front. Our analysis resulted in the identification and generation of a desmoplastic protein dataset (DPD), consisting of 152 candidate proteins of desmoplasia. By using monoculture exclusion datasets, a secretome algorithm and gene-expression meta-analysis in DPD, we specified a 22-protein “myofibroblastic signature” with putative importance in the regulation of colorectal cancer metastasis. Of these proteins, we investigated collagen type XII by immunohistochemistry, a fibril-associated collagen with interrupted triple helices (FACIT), whose expression has not been reported in desmoplastic lesions in any type of cancer. Collagen type XII was highly expressed in desmoplastic stroma by and around alpha-smooth muscle actin (α-SMA) positive CAFs, as well as in cancer cells lining the invasion front, in a small cohort of colon cancer patients. Other stromal markers, such as collagen type III, were also expressed in stromal collagen, but not in cancer cells. In a complementary fashion, gene expression meta-analysis revealed that COL12A1 is also an upregulated gene in colorectal cancer. Our proteomic analysis identified previously documented markers of tumor invasion fronts and our DPD could serve as a pool for future investigation of the tumor microenvironment. Collagen type XII is a novel candidate marker of myofibroblasts, and/or cancer cells undergoing dedifferentiation.
Collapse
|
34
|
Mishra PJ, Mishra PJ, Banerjee D. Cell-free derivatives from mesenchymal stem cells are effective in wound therapy. World J Stem Cells 2012; 4:35-43. [PMID: 22993660 PMCID: PMC3443710 DOI: 10.4252/wjsc.v4.i5.35] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/08/2011] [Accepted: 11/15/2011] [Indexed: 02/06/2023] Open
Abstract
AIM To compare the efficacy of cell-free derivatives from Bone marrow derived human mesenchymal stem cells (hMSCs) in wound therapy. METHODS hMSCs have been shown to play an important role in wound therapy. The present study sought to compare efficacy of hMSCs and cell-free derivatives of hMSCs, which may be clinically more relevant as they are easier to prepare, formulate and transport. hMSCs were isolated from human bone marrow and cultured. Multi lineage differentiation of hMSCs was performed to confirm their identity. The ability of hMSCs to migrate was evaluated using in vitro and in vivo migration assays. Cell lysates and conditioned medium concentrate was prepared from hMSCs (see Methods for details). Wounds were induced in mice and wound areas were measure before and after cell and cell-free derivative treatment. RNA and proteins were extracted from the skin and cytokine levels were measured. RESULTS Co-culture of hMSCs with keratinocytes resulted in increased expression of CXCL-12 (SDF1) and ENA78 (CXCL-5) in the conditioned media indicating that the hMSCs can respond to signals from keratinocytes. Accelerated wound closure was observed when hMSCs were injected near the site of excisional wounds in athymic as well as NOD/SCID mice. Interestingly, cell-free lysates prepared from hMSCs were also effective in inducing accelerated wound closure and increased expression of SDF1 and CXCL-5 at the wound bed. Additionally, concentrated media from hMSCs as well as an emulsion containing lysates prepared from hMSCs was also found to be more effective in rapid re-epithelialization than fibroblasts or vehicle-alone control. Use of cell-free derivatives may help replace expensive wound care approaches including use of growth factors, epidermal/dermal substitutes, synthetic membranes, cytokines, and matrix components, and most importantly avoid transmission of pathogens from human and animal products. CONCLUSION These results encourage development of derivatives of hMSCs for wound care and re-epithelialization applications.
Collapse
Affiliation(s)
- Pravin J Mishra
- Pravin J Mishra, Prasun J Mishra, Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, United States
| | | | | |
Collapse
|
35
|
Arenal A, Hernandez J, Perez-David E, Rubio-Guivernau JL, Ledesma-Carbayo MJ, Fernandez-Aviles F. Do the spatial characteristics of myocardial scar tissue determine the risk of ventricular arrhythmias? Cardiovasc Res 2012; 94:324-32. [DOI: 10.1093/cvr/cvs113] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
36
|
Ong HS, Ji T, Zhang CP, Li J, Wang LZ, Li RR, Sun J, Ma CY. Head and neck inflammatory myofibroblastic tumor (IMT): evaluation of clinicopathologic and prognostic features. Oral Oncol 2011; 48:141-8. [PMID: 22014665 DOI: 10.1016/j.oraloncology.2011.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 09/11/2011] [Accepted: 09/13/2011] [Indexed: 12/31/2022]
Abstract
Owing to rarity and awareness deficiency towards inflammatory myofibroblastic tumor (IMT), we sought to review on its clinicopathological features; arising awareness to achieve early diagnosis; exploring prognostic factors and then establishing a treatment protocol. Retrospective study was performed on patients with histological proven IMT between January 2003 and December 2010. Their demographic data, clinical and histological presentations were recorded. Overall survival (OS) and progression-free-survival (PFS) were estimated via Kaplan-Meier method. Cox regression model was applied to determine the significant of prognostic factors. Logistic regression model was established to predict the probability of relapse. A total of 28 patients. Five-year PFS was 65%. Surgical margins primarily and independently determined the survival, followed by size, pseudocapsule of the lesion, intra-lesional necrosis and lastly Ki-67 and ALK overexpression. Logistic model in prediction of relapse was established, with the formula as probability of relapse = 1/(1 + e(-z)) where e = exponential function, z = constant value (3.9) + B*margin + B*size + B*immunohistochemical expression + B*pseudocapsule + B*intra-lesional necrosis. Immunohistochemical overexpression was significant if Ki-67 was strongly expressed with a conditioned ALK overexpression simultaneously. Staining intensity must be at least moderate for those ALK nuclear staining was less than 25%. Weak ALK staining intensity is only significant if nuclear staining was more than 25%. Diagnosis of IMT is achieved via exclusion. Radical resection and obtaining negative margins remains the mainstay of treatment. Both high and moderate-risk groups required post-operative radiotherapy. In low-risk group, post-operative radiotherapy was recommended if the lesion is larger than 5 cm in diameter with a conditioned ALK & Ki-67 overexpression.
Collapse
Affiliation(s)
- Hui Shan Ong
- Department of Oral and Maxillofacial Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Short-term stromal alterations in the rat ventral prostate following alloxan-induced diabetes and the influence of insulin replacement. Micron 2011; 43:326-33. [PMID: 22014851 DOI: 10.1016/j.micron.2011.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 09/21/2011] [Accepted: 09/22/2011] [Indexed: 12/18/2022]
Abstract
The stromal microenvironment is pivotal to prostate physiology and malign transformation. Diabetes leads to testosterone withdrawal and affects the prostate stromal compartment and smooth muscle cells in a similar way to that observed after castration. However the response of these cells and their involvement in extracellular matrix remodeling is not satisfactorily understood. We investigated the changes caused in the short term (one week) by alloxan-induced diabetes in the stromal components of the rat ventral prostate (VP) with an emphasis on morphological alterations of stromal cells, their conversion to a myofibroblast phenotype and the remodeling of extracellular matrix and the influence of insulin therapy. Adult male Wistar rats were assigned into untreated diabetic (n=12), insulin-treated (n=8) diabetic and control (n=10) groups. Diabetes was induced by means of the injection of alloxan (40 mg/kg b.w.), while the control animals received saline solution only. Insulin (5 UI) was administered daily for one week after diabetes diagnosis. Testosterone and estrogen plasma levels were determined. VP was analyzed using transmission electron microscopy. The main stromal cells were identified by means of light microscopy, using immunocytochemistry for specific markers - vimentin for fibroblasts, α-actin for smooth muscle cells (smc) and vimentin/calponin for myofibroblasts, following the estimation of their relative frequency and absolute volume by means of stereology. After one week diabetes led to a marked decrease in testosterone levels and an atrophy of about 35% in the VP. The relative frequency of smc and collagen fibers increased in the VP of diabetic rats but their absolute weight remained unchanged. Experimental diabetes promptly altered smc morphology which assumed at the ultrastructural level a shrunken appearance with the approximation of cytoplasmic dense bodies and also exhibited a decreased immunoreactivity to calponin. The conversion of stromal cells to a myofibroblast phenotype did not occur in alloxan-induced diabetes, as evaluated by double immunoreaction to calponin and vimentin. Insulin treatment maintained testosterone levels and preserved at least partly the cell morphology and collagen fiber organization of the prostate stroma in short-term diabetes. The apparent collagen increase observed by means of microscopic analysis in the stromal prostate compartment in the short term after diabetes is mainly associated with gland atrophy and does not involve the formation of new collagen fibers, the generation of myofibroblast-like cells or the acquisition of a secretory phenotype by stromal cells.
Collapse
|
38
|
Stromal expression of CD34, α-smooth muscle actin and CD26/DPPIV in squamous cell carcinoma of the skin: a comparative immunohistochemical study. Pathol Oncol Res 2011; 18:25-31. [PMID: 21674250 DOI: 10.1007/s12253-011-9412-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 05/03/2011] [Indexed: 12/16/2022]
Abstract
Invasion pathogenesis is one of the most complicated issues in the literature. There are numerous studies concerning the tumor markers implicated in the preinvasive-invasive tumor sequence. Despite ample studies on the invasion pathogenesis of cutaneous melanomas, there is limited and dispersed work presently available on non-melanoma skin cancer. The vast knowledge in the literature concerning this issue in squamous cell carcinoma comes mostly from the studies of the oral cavity, esophagus, larynx, and cervix. In this study, we investigated tumor-free neighboring stroma and tumor stroma in squamous cell carcinomas (SCCs) of the skin as well as keratoacanthomas (KAs) with respect to the presence of stromal CD34-positive (CD34+) fibrocytes and α-smooth muscle actin-positive (α-SMA+) myofibroblasts using seborrheic keratosis (SKs) and non-tumoral skin samples as controls. We also evaluated the stromal expression pattern of CD26/DPPIV (CD26), a tumor suppressor gene product that also has immunoregulatory properties. Immunohistochemistry was performed on samples of 31 SCC, 8 KA, 15 SK and 10 non-tumoral skin samples. Peri-tumoral stroma from resection margins was also evaluated. We found that CD34 and α-SMA demonstrated significantly different staining between benign and malignant squamous skin lesions consisting of a loss of CD34+ fibrocytes paralleled by a gain of α-SMA+ myofibroblasts in malignant tumor stroma. Additionally, it was shown that CD26 expression was lower in tumor stroma when compared to that of tumor neighboring stroma. However, we concluded that this finding may be attributable to the solar elastosis areas in the peritumoral tissue, which shows diffuse strong positivity for this marker.
Collapse
|
39
|
Grimaldi A, Banfi S, Vizioli J, Tettamanti G, Noonan DM, de Eguileor M. Cytokine loaded biopolymers as a novel strategy to study stem cells during wound-healing processes. Macromol Biosci 2011; 11:1008-19. [PMID: 21400659 DOI: 10.1002/mabi.201000452] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 02/10/2011] [Indexed: 12/12/2022]
Abstract
The biopolymer matrigel loaded with cytokine can be used for the recruitment in vivo of specific cell populations and as a vector for the preparation of cell cultures. Data demonstrate that the injection of the matrigel biopolymer supplemented with interleukin-8 (IL-8) in the leech Hirudo medicinalis can be used to purify cell populations showing the same morphofunctional and molecular mechanisms of specific populations of vertebrate hematopoietic precursor cells involved in tissue repair. These cells spontaneously differentiated into myofibroblasts. This approach highlights how the innovative use of a cytokine-loaded biopolymer for an in vivo cell sorting method, applied to a simple invertebrate model, can be a tool for studying myofibroblast cell biology and its regulation, step by step.
Collapse
Affiliation(s)
- Annalisa Grimaldi
- Department of Biotechnology and Molecular Sciences, University of Insubria, Varese, Italy.
| | | | | | | | | | | |
Collapse
|
40
|
Ehrlich HP, Hazard III SW. Thymosin β4 enhances repair by organizing connective tissue and preventing the appearance of myofibroblasts. Ann N Y Acad Sci 2010; 1194:118-24. [DOI: 10.1111/j.1749-6632.2010.05483.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Abstract
The permanent cellular constituents of the heart include cardiac fibroblasts, myocytes, endothelial cells, and vascular smooth muscle cells. Previous studies have demonstrated that there are undulating changes in cardiac cell populations during embryonic development, through neonatal development and into the adult. Transient cell populations include lymphocytes, mast cells, and macrophages, which can interact with these permanent cell types to affect cardiac function. It has also been observed that there are marked differences in the makeup of the cardiac cell populations depending on the species, which may be important when examining myocardial remodeling. Current dogma states that the fibroblast makes up the largest cell population of the heart; however, this appears to vary for different species, especially mice. Cardiac fibroblasts play a critical role in maintaining normal cardiac function, as well as in cardiac remodeling during pathological conditions such as myocardial infarct and hypertension. These cells have numerous functions, including synthesis and deposition of extracellular matrix, cell-cell communication with myocytes, cell-cell signaling with other fibroblasts, as well as with endothelial cells. These contacts affect the electrophysiological properties, secretion of growth factors and cytokines, as well as potentiating blood vessel formation. Although a plethora of information is known about several of these processes, relatively little is understood about fibroblasts and their role in angiogenesis during development or cardiac remodeling. In this review, we provide insight into the various properties of cardiac fibroblasts that helps illustrate their importance in maintaining proper cardiac function, as well as their critical role in the remodeling heart.
Collapse
Affiliation(s)
- Colby A. Souders
- Texas A&M Health Science Center College of Medicine, Division of Molecular Cardiology, Temple, TX 76504
| | - Stephanie L.K. Bowers
- Texas A&M Health Science Center College of Medicine, Division of Molecular Cardiology, Temple, TX 76504
| | - Troy A. Baudino
- Texas A&M Health Science Center College of Medicine, Division of Molecular Cardiology, Temple, TX 76504
| |
Collapse
|
42
|
Cunnington RH, Nazari M, Dixon IM. c-Ski, Smurf2, and Arkadia as regulators of TGF-β signaling: new targets for managing myofibroblast function and cardiac fibrosisThis article is one of a selection of papers published in a special issue celebrating the 125th anniversary of the Faculty of Medicine at the University of Manitoba. Can J Physiol Pharmacol 2009; 87:764-72. [DOI: 10.1139/y09-076] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies demonstrate the critical role of the extracellular matrix in the organization of parenchymal cells in the heart. Thus, an understanding of the modes of regulation of matrix production by cardiac myofibroblasts is essential. Transforming growth factor β (TGF-β) signaling is transduced through the canonical Smad pathway, and the involvement of this pathway in matrix synthesis and other processes requires precise control. Inhibition of Smad signaling may be achieved at the receptor level through the targeting of the TGF-β type I receptors with an inhibitory Smad7 / Smurf2 complex, or at the transcriptional level through c-Ski / receptor-Smad / co-mediator Smad4 interactions. Conversely, Arkadia protein intensifies TGF-β-induced effects by marking c-Ski and inhibitory Smad7 for destruction. The study of these TGF-β mediators is essential for future treatment of fibrotic disease, and this review highlights recent relevant findings that may impact our understanding of cardiac fibrosis.
Collapse
Affiliation(s)
- Ryan H. Cunnington
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Mansoreh Nazari
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| | - Ian M.C. Dixon
- Institute of Cardiovascular Sciences, St. Boniface General Hospital Research Centre, Department of Physiology, University of Manitoba, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
43
|
van den Bogaerdt AJ, van der Veen VC, van Zuijlen PP, Reijnen L, Verkerk M, Bank RA, Middelkoop E, Ulrich MM. Collagen cross-linking by adipose-derived mesenchymal stromal cells and scar-derived mesenchymal cells: Are mesenchymal stromal cells involved in scar formation? Wound Repair Regen 2009; 17:548-58. [DOI: 10.1111/j.1524-475x.2009.00501.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Wernli G, Hasan W, Bhattacherjee A, van Rooijen N, Smith PG. Macrophage depletion suppresses sympathetic hyperinnervation following myocardial infarction. Basic Res Cardiol 2009; 104:681-93. [PMID: 19437062 DOI: 10.1007/s00395-009-0033-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 04/24/2009] [Accepted: 04/29/2009] [Indexed: 01/16/2023]
Abstract
Myocardial infarction induces sympathetic axon sprouting adjacent to the necrotic region, and this has been implicated in the etiology of arrhythmias resulting in sudden cardiac death. Previous studies show that nerve growth factor (NGF) is essential for enhanced post-infarct sympathetic sprouting, but the cell types necessary to supply this neurotrophic protein are unknown. The objective of the present study was to determine whether macrophages, which are known to synthesize NGF, are necessary for post-infarct cardiac sympathetic sprouting. Ovariectomized female rats received left coronary artery ligation or sham operation, followed by intravenous injection of liposomes containing saline vehicle or clodronate, which kills macrophages. Sham-operated myocardium contained some sympathetic axons, few myofibroblasts and T cells and no CD-68-positive macrophages. In rats receiving saline liposomes through 7 days post-ligation, the posterolateral infarct border contained numerous myofibroblasts, macrophages and T cells, and sympathetic innervation was increased twofold. Treatment with clodronate liposomes reduced macrophage numbers by 69%, while myofibroblast area was reduced by 23% and T cell number was unaffected. Clodronate liposome treatment reduced sympathetic axon density to levels comparable to the uninfarcted heart. NGF protein content measured in western blots was reduced to 33% of that present in infarcts where rats received saline-containing liposomes. Tissue morphometry confirmed that NGF immunostaining was dramatically reduced, and this was attributable primarily to reduced macrophage content. These results show that macrophage destruction markedly reduces post-infarction levels of NGF and that the presence of elevated numbers of macrophages is obligatory for development of sympathetic hyperinnervation following myocardial infarction.
Collapse
Affiliation(s)
- Gwenaelle Wernli
- Department of Molecular and Integrative Physiology, Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Mail Stop 3051, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
Myocardial infarction is the most common cause of cardiac injury and results in acute loss of a large number of myocardial cells. Because the heart has negligible regenerative capacity, cardiomyocyte death triggers a reparative response that ultimately results in formation of a scar and is associated with dilative remodeling of the ventricle. Cardiac injury activates innate immune mechanisms initiating an inflammatory reaction. Toll-like receptor-mediated pathways, the complement cascade and reactive oxygen generation induce nuclear factor (NF)-kappaB activation and upregulate chemokine and cytokine synthesis in the infarcted heart. Chemokines stimulate the chemotactic recruitment of inflammatory leukocytes into the infarct, while cytokines promote adhesive interactions between leukocytes and endothelial cells, resulting in transmigration of inflammatory cells into the site of injury. Monocyte subsets play distinct roles in phagocytosis of dead cardiomyocytes and in granulation tissue formation through the release of growth factors. Clearance of dead cells and matrix debris may be essential for resolution of inflammation and transition into the reparative phase. Transforming growth factor (TGF)-beta plays a crucial role in cardiac repair by suppressing inflammation while promoting myofibroblast phenotypic modulation and extracellular matrix deposition. Myofibroblast proliferation and angiogenesis result in formation of highly vascularized granulation tissue. As the healing infarct matures, fibroblasts become apoptotic and a collagen-based matrix is formed, while many infarct neovessels acquire a muscular coat and uncoated vessels regress. Timely resolution of the inflammatory infiltrate and spatial containment of the inflammatory and reparative response into the infarcted area are essential for optimal infarct healing. Targeting inflammatory pathways following infarction may reduce cardiomyocyte injury and attenuate adverse remodeling. In addition, understanding the role of the immune system in cardiac repair is necessary in order to design optimal strategies for cardiac regeneration.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Section of Cardiovascular Sciences, Baylor College of Medicine, One Baylor Plaza BCM620, Houston, TX 77030, United States.
| |
Collapse
|
46
|
Pho M, Lee W, Watt DR, Laschinger C, Simmons CA, McCulloch CA. Cofilin is a marker of myofibroblast differentiation in cells from porcine aortic cardiac valves. Am J Physiol Heart Circ Physiol 2008; 294:H1767-78. [DOI: 10.1152/ajpheart.01305.2007] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The formation of myofibroblasts in valve interstitial cell (VIC) populations contributes to fibrotic valvular disease. We examined myofibroblast differentiation in VICs from porcine aortic valves. In normal valves, cells immunostained for α-smooth muscle actin (α-SMA, a myofibroblast marker) were rare (0.69 ± 0.48%), but in sclerotic valves of animals fed an atherogenic diet, myofibroblasts were spatially clustered and abundant (31.2 ± 6.3%). In cultured VIC populations from normal valves, SMA-positive myofibroblasts were also spatially clustered, abundant (21% positive cells after 1 passage), and stained for collagen type I and vimentin but not desmin. For an analysis of stem cells, two-color flow cytometry of isolated cells stained with Hoechst 33342 demonstrated that 0.5% of VICs were side population cells; none stained for SMA. Upon culture, sorted side population cells generated ∼85% SMA-positive cells, indicating that some myofibroblasts originate from a rare population with stem cell characteristics. Plating cells on rigid collagen substrates enabled the formation of myofibroblasts after 5 days in culture, which was completely blocked by culture of cells on compliant collagen substrates. Exogenous tensile force also significantly increased SMA expression in VICs. Isotope-coded affinity tags and mass spectrometry were used to identify differentially expressed proteins in myofibroblast differentiation of VICs. Of the nine proteins that were identified, cofilin expression and phospho-cofilin were strongly increased by conditions favoring myofibroblast differentiation. Knockdown of cofilin with small-interfering RNA inhibited collagen gel contraction and reduced myofibroblast differentiation as assessed by the SMA incorporation into stress fibers. When compared with normal valves, diseased valves showed strong immunostaining for cofilin that colocalized with SMA in clustered cells. We conclude that in VICs, cofilin is a marker for myofibroblasts in vivo and in vitro that arise from a rare population of stem cells and require a rigid matrix for formation.
Collapse
|
47
|
Enoch S, Moseley R, Stephens P, Thomas D. The oral mucosa: a model of wound healing with reduced scarring. ACTA ACUST UNITED AC 2008. [DOI: 10.1111/j.1752-248x.2007.00005.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
Groger A, Bozkurt A, Piatkowski A, Perez-Bouza A, Fuchs P, Pallua N. Fibromatosis-like myofibroblastic tumour of forearm: case report and interdisciplinary management. ACTA ACUST UNITED AC 2007; 41:329-31. [PMID: 17952816 DOI: 10.1080/02844310600759582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
We present a rare case of a fibromatosis-like myofibroblastic tumour of the forearm with infiltration of muscular, neural, and vascular structures. This is a rare and transitional type of myofibroblastic tumour, and we emphasise important aspects of diagnosis, clinical features, interdisciplinary management, and differential diagnoses.
Collapse
Affiliation(s)
- Andreas Groger
- Department of Plastic, Reconstructive, and Hand Surgery, Burn Center, University Hospital RWTH Aachen, Germany.
| | | | | | | | | | | |
Collapse
|
49
|
Yang T, Vesey DA, Johnson DW, Wei MQ, Gobe GC. Apoptosis of tubulointerstitial chronic inflammatory cells in progressive renal fibrosis after cancer therapies. Transl Res 2007; 150:40-50. [PMID: 17585862 DOI: 10.1016/j.trsl.2007.01.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 01/05/2007] [Accepted: 01/08/2007] [Indexed: 10/23/2022]
Abstract
Progressive renal fibrosis is an unwanted and limiting side effect of cancer treatments, whether they are systemic (for example, chemotherapy), local (for example, radiotherapy), or total body irradiation for allogenic bone marrow transplants. The relative roles of macrophages, myofibroblasts, and lymphocytes and the apoptotic deletion of renal functional or inflammatory cell populations in the pathogenesis of renal fibrosis are yet unclear. In this study, rat models of 2 renal cancer treatments: cis-platinum-(II)-diammine dichloride (cisplatin, 6-mg/kg body weight) and radiation (single dose of 20 Gy) were used. Kidneys were analyzed 4 days to 3 months after treatment. The extent of renal fibrosis was compared with number and localization of chronic inflammatory cell populations, cell death (apoptosis and necrosis), and expression and localization of profibrotic growth factors transforming growth factor-beta1 (TGF-beta1) and tumor necrosis factor-alpha (TNF-alpha). The models provided contrasting rates of fibrogenesis: After cisplatin, development of fibrosis was rapid and extensive (up to 50% fibrosis at 3 months); in comparison, radiation-induced fibrosis was slowly progressive (approximately 10% fibrosis at 3 months). The extent of fibrosis was associated spatially and temporally with increasing numbers of myofibroblasts with TGF-beta1 or macrophages with TNF-alpha. Tubular epithelial apoptosis was highest with high TNF-alpha (P<0.05). A significant inverse correlation existed between extent of tubulointerstitial fibrosis and interstitial cell apoptosis for cisplatin and a similar nonsignificant result for radiation (r(2)=0.8671 for cisplatin, P<0.05; r(2)=0.2935 for radiation, NS). The latter result suggests a role for inflammatory cell apoptosis in minimizing development of renal fibrosis.
Collapse
Affiliation(s)
- Tao Yang
- Discipline of Molecular and Cellular Pathology, School of Medicine, University of Queensland, Herston, Brisbane, Australia
| | | | | | | | | |
Collapse
|
50
|
Huh CH, Oh JK, Kim BJ, Kim MH, Won CH, Eun HC. Photoepilation: a potential threat to wound healing in a mouse. J Cosmet Dermatol 2006; 5:115-20. [PMID: 17173584 DOI: 10.1111/j.1473-2165.2006.00236.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Theoretically, the bulge area which is known to be a reservoir of epidermal stem cells should be destroyed to achieve permanent photoepilation. We wished to determine whether wound healing capability is perturbed after photoepilation. METHODS Twenty C57/BL6 mice were used. After wax epilation to synchronize the hair cycle, one-half of the backs of mice were photoepilated in the early anagen stage. After the two hair cycles of the mice to confirm the hair removal effect, 30% trichloroacetic acid was applied to the both halves of the backs of the mice. A skin biopsy was performed on both sides before and just after the injury, and 2, 6, 9, and 14 days thereafter. The specimens were evaluated histologically after staining with hematoxylin and eosin, Masson trichrome, and Verhoeff-van Gieson. RESULTS No differences in wound healing times were evident upon gross observation by the naked eye. However, the photoepilated hairless skin was observed to have a thicker epidermis and dermis than normal hairy skin by histological evaluation. The cellularity of the healed wound was much denser in the photoepilated. Collagen production of the neodermis in the normal hairy skin was first observed around the lower part of hair follicle, while it started from the upper papillary dermis in photoepilated skin. CONCLUSION Photoepilation may disturb the normal wound healing process, especially dermal wound healing, and increases the risk of producing hypertropic scar or keloid.
Collapse
Affiliation(s)
- Chang Hun Huh
- Seoul National University College of Medicine, Department of Dermatology, Seoul, Korea
| | | | | | | | | | | |
Collapse
|