1
|
Salminen A. The role of immunosuppressive myofibroblasts in the aging process and age-related diseases. J Mol Med (Berl) 2023; 101:1169-1189. [PMID: 37606688 PMCID: PMC10560181 DOI: 10.1007/s00109-023-02360-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
Tissue-resident fibroblasts are mesenchymal cells which control the structural integrity of the extracellular matrix (ECM). Fibroblasts possess a remarkable plasticity to allow them to adapt to the changes in the microenvironment and thus maintain tissue homeostasis. Several stresses, also those associated with the aging process, convert quiescent fibroblasts into myofibroblasts which not only display fibrogenic properties but also act as immune regulators cooperating both with tissue-resident immune cells and those immune cells recruited into affected tissues. TGF-β cytokine and reactive oxygen species (ROS) are major inducers of myofibroblast differentiation in pathological conditions either from quiescent fibroblasts or via transdifferentiation from certain other cell types, e.g., macrophages, adipocytes, pericytes, and endothelial cells. Intriguingly, TGF-β and ROS are also important signaling mediators between immunosuppressive cells, such as MDSCs, Tregs, and M2 macrophages. It seems that in pathological states, myofibroblasts are able to interact with the immunosuppressive network. There is clear evidence that a low-grade chronic inflammatory state in aging tissues is counteracted by activation of compensatory immunosuppression. Interestingly, common enhancers of the aging process, such as oxidative stress, loss of DNA integrity, and inflammatory insults, are inducers of myofibroblasts, whereas anti-aging treatments with metformin and rapamycin suppress the differentiation of myofibroblasts and thus prevent age-related tissue fibrosis. I will examine the reciprocal interactions between myofibroblasts and immunosuppressive cells within aging tissues. It seems that the differentiation of myofibroblasts with age-related harmful stresses enhances the activity of the immunosuppressive network which promotes tissue fibrosis and degeneration in elderly individuals.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
2
|
Tiendrébéogo AJF, Soumagne T, Pellegrin F, Dagouassat M, Tran Van Nhieu J, Caramelle P, Paul EN, Even B, Zysman M, Julé Y, Samb A, Boczkowski J, Lanone S, Schlemmer F. The telomerase activator TA-65 protects from cigarette smoke-induced small airway remodeling in mice through extra-telomeric effects. Sci Rep 2023; 13:25. [PMID: 36646720 PMCID: PMC9842758 DOI: 10.1038/s41598-022-25993-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
Small airway remodeling (SAR) is a key phenomenon of airflow obstruction in smokers, leading to chronic obstructive pulmonary disease (COPD). SAR results in an increased thickness of small airway walls, with a combination of peribronchiolar fibrosis with increased fibrous tissue and accumulation of mesenchymal and epithelial cells. SAR pathogenesis is still unclear but recent data suggest that alterations in telomerase activity could represent a possible underlying mechanism of SAR. Our study was dedicated to identify a potential protective role of TA-65, a pharmacological telomerase activator, in a cigarette smoke (CS) model of SAR in mice, and to further precise if extra-telomeric effects of telomerase, involving oxidative stress modulation, could explain it. C57BL/6J mice were daily exposed to air or CS during 4 weeks with or without a concomitant administration of TA-65 starting 7 days before CS exposure. Morphological analyses were performed, and mucus production, myofibroblast differentiation, collagen deposition, as well as transforming growth factor-β1 (TGF-β1) expression in the small airway walls were examined. In addition, the effects of TA-65 treatment on TGF-β expression, fibroblast-to-myofibroblast differentiation, reactive oxygen species (ROS) production and catalase expression and activity were evaluated in primary cultures of pulmonary fibroblasts and/or mouse embryonic fibroblasts in vitro. Exposure to CS during 4 weeks induced SAR in mice, characterized by small airway walls thickening and peribronchiolar fibrosis (increased deposition of collagen, expression of α-SMA in small airway walls), without mucus overproduction. Treatment of mice with TA-65 protected them from CS-induced SAR. This effect was associated with the prevention of CS-induced TGF-β expression in vivo, the blockade of TGF-β-induced myofibroblast differentiation, and the reduction of TGF-β-induced ROS production that correlates with an increase of catalase expression and activity. Our findings demonstrate that telomerase is a critical player of SAR, probably through extra-telomeric anti-oxidant effects, and therefore provide new insights in the understanding and treatment of COPD pathogenesis.
Collapse
Affiliation(s)
- Arnaud Jean Florent Tiendrébéogo
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,Laboratoire de physiologie et d’explorations fonctionnelles physiologiques, Université Cheik Anta Diop, Dakar, Senegal
| | - Thibaud Soumagne
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - François Pellegrin
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Maylis Dagouassat
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Jeanne Tran Van Nhieu
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,grid.412116.10000 0004 1799 3934Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Pathologie, 94000 Créteil, France
| | - Philippe Caramelle
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Emmanuel N. Paul
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Benjamin Even
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Maeva Zysman
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | | | - Abdoulaye Samb
- Laboratoire de physiologie et d’explorations fonctionnelles physiologiques, Université Cheik Anta Diop, Dakar, Senegal
| | - Jorge Boczkowski
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,grid.412116.10000 0004 1799 3934Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Service d’explorations fonctionnelles respiratoires, DHU A-TVB, FHU Senec, 94000 Créteil, France
| | - Sophie Lanone
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Frédéric Schlemmer
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,grid.412116.10000 0004 1799 3934Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Unité de Pneumologie, DHU A-TVB, FHU Senec, 94000 Créteil, France
| |
Collapse
|
3
|
Chan M, Yuan H, Soifer I, Maile TM, Wang RY, Ireland A, O'Brien JJ, Goudeau J, Chan LJ, Vijay T, Freund A, Kenyon C, Bennett BD, McAllister FE, Kelley DR, Roy M, Cohen RL, Levinson AD, Botstein D, Hendrickson DG. Novel insights from a multiomics dissection of the hayflick limit. eLife 2022; 11:70283. [PMID: 35119359 PMCID: PMC8933007 DOI: 10.7554/elife.70283] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 01/31/2022] [Indexed: 01/10/2023] Open
Abstract
The process wherein dividing cells exhaust proliferative capacity and enter into replicative senescence has become a prominent model for cellular aging in vitro. Despite decades of study, this cellular state is not fully understood in culture and even much less so during aging. Here, we revisit Leonard Hayflick’s original observation of replicative senescence in WI-38 human lung fibroblasts equipped with a battery of modern techniques including RNA-seq, single-cell RNA-seq, proteomics, metabolomics, and ATAC-seq. We find evidence that the transition to a senescent state manifests early, increases gradually, and corresponds to a concomitant global increase in DNA accessibility in nucleolar and lamin associated domains. Furthermore, we demonstrate that senescent WI-38 cells acquire a striking resemblance to myofibroblasts in a process similar to the epithelial to mesenchymal transition (EMT) that is regulated by t YAP1/TEAD1 and TGF-β2. Lastly, we show that verteporfin inhibition of YAP1/TEAD1 activity in aged WI-38 cells robustly attenuates this gene expression program.
Collapse
Affiliation(s)
- Michelle Chan
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Han Yuan
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Ilya Soifer
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Tobias M Maile
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Rebecca Y Wang
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Andrea Ireland
- Calico Life Sciences, LLC, South San Francisco, United States
| | | | - Jérôme Goudeau
- Calico Life Sciences LLC, South San Francisco, United States
| | - Leanne Jg Chan
- Calico Life Sciences LLC, South San Francisco, United States
| | - Twaritha Vijay
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Adam Freund
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Cynthia Kenyon
- Calico Life Sciences LLC, South San Francisco, United States
| | | | | | - David R Kelley
- Calico Life Sciences, LLC, South San Francisco, United States
| | - Margaret Roy
- Calico Life Sciences LLC, South San Francisco, United States
| | - Robert L Cohen
- Calico Life Sciences, LLC, South San Francisco, United States
| | | | - David Botstein
- Calico Life Sciences, LLC, South San Francisco, United States
| | | |
Collapse
|
4
|
Harada M, Hu B, Lu J, Wang J, Rinke AE, Wu Z, Liu T, Phan SH. The dual distinct role of telomerase in repression of senescence and myofibroblast differentiation. Aging (Albany NY) 2021; 13:16957-16973. [PMID: 34253690 PMCID: PMC8312426 DOI: 10.18632/aging.203246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
Many aging related diseases such as cancer implicate the myofibroblast in disease progression. Furthermore genesis of the myofibroblast is associated with manifestation of cellular senescence of unclear significance. In this study we investigated the role of a common regulator, namely telomerase reverse transcriptase (TERT), in order to evaluate the potential significance of this association between both processes. We analyzed the effects of TERT overexpression or deficiency on expression of CDKN2A and ACTA2 as indicators of senescence and differentiation, respectively. We assess binding of TERT or YB-1, a repressor of both genes, to their promoters. TERT repressed both CDKN2A and ACTA2 expression, and abolished stress-induced expression of both genes. Conversely, TERT deficiency enhanced their expression. Altering CDKN2A expression had no effect on ACTA2 expression. Both TERT and YB-1 were shown to bind the CDKN2A promoter but only YB-1 was shown to bind the ACTA2 promoter. TERT overexpression inhibited CDKN2A promoter activity while stimulating YB-1 expression and activation to repress ACTA2 gene. TERT repressed myofibroblast differentiation and senescence via distinct mechanisms. The latter was associated with TERT binding to the CDKN2A promoter, but not to the ACTA2 promoter, which may require interaction with co-factors such as YB-1.
Collapse
Affiliation(s)
- Masanori Harada
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Department of Respiratory Medicine, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jeffrey Lu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jing Wang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Xinjiang Key Laboratory of Respiratory Disease Research, Traditional Chinese Medicine Affiliated Hospital of Xinjiang Medical University, Urumqi 830000, China
| | - Andrew E Rinke
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sem H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Luppi F, Kalluri M, Faverio P, Kreuter M, Ferrara G. Idiopathic pulmonary fibrosis beyond the lung: understanding disease mechanisms to improve diagnosis and management. Respir Res 2021; 22:109. [PMID: 33865386 PMCID: PMC8052779 DOI: 10.1186/s12931-021-01711-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/11/2021] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive disorder with an estimated median survival time of 3–5 years after diagnosis. This condition occurs primarily in elderly subjects, and epidemiological studies suggest that the main risk factors, ageing and exposure to cigarette smoke, are associated with both pulmonary and extrapulmonary comorbidities (defined as the occurrence of two or more disorders in a single individual). Ageing and senescence, through interactions with environmental factors, may contribute to the pathogenesis of IPF by various mechanisms, causing lung epithelium damage and increasing the resistance of myofibroblasts to apoptosis, eventually resulting in extracellular matrix accumulation and pulmonary fibrosis. As a paradigm, syndromes featuring short telomeres represent archetypal premature ageing syndromes and are often associated with pulmonary fibrosis. The pathophysiological features induced by ageing and senescence in patients with IPF may translate to pulmonary and extrapulmonary features, including emphysema, pulmonary hypertension, lung cancer, coronary artery disease, gastro-oesophageal reflux, diabetes mellitus and many other chronic diseases, which may lead to substantial negative consequences in terms of various outcome parameters in IPF. Therefore, the careful diagnosis and treatment of comorbidities may represent an outstanding chance to improve quality of life and survival, and it is necessary to contemplate all possible management options for IPF, including early identification and treatment of comorbidities.
Collapse
Affiliation(s)
- Fabrizio Luppi
- Respiratory Unit, University of Milano Bicocca, S. Gerardo Hospital, ASST Monza, Monza, Italy
| | - Meena Kalluri
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, 3-134 Clinical Sciences Building, 11304 83 Ave., Edmonton, AB, T6G 2G3, Canada
| | - Paola Faverio
- Respiratory Unit, University of Milano Bicocca, S. Gerardo Hospital, ASST Monza, Monza, Italy
| | - Michael Kreuter
- Centre for Interstitial and Rare Lung Diseases, Pneumology and Respiratory Critical Care Medicine, University of Heidelberg, German Center for Lung Research, ThoraxklinikHeidelberg, Germany
| | - Giovanni Ferrara
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada. .,Division of Pulmonary Medicine, Department of Medicine, University of Alberta, 3-134 Clinical Sciences Building, 11304 83 Ave., Edmonton, AB, T6G 2G3, Canada.
| |
Collapse
|
6
|
Hinz B, Lagares D. Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases. Nat Rev Rheumatol 2020; 16:11-31. [PMID: 31792399 PMCID: PMC7913072 DOI: 10.1038/s41584-019-0324-5] [Citation(s) in RCA: 338] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
Organ fibrosis is a lethal outcome of autoimmune rheumatic diseases such as systemic sclerosis. Myofibroblasts are scar-forming cells that are ultimately responsible for the excessive synthesis, deposition and remodelling of extracellular matrix proteins in fibrosis. Advances have been made in our understanding of the mechanisms that keep myofibroblasts in an activated state and control myofibroblast functions. However, the mechanisms that help myofibroblasts to persist in fibrotic tissues remain poorly understood. Myofibroblasts evade apoptosis by activating molecular mechanisms in response to pro-survival biomechanical and growth factor signals from the fibrotic microenvironment, which can ultimately lead to the acquisition of a senescent phenotype. Growing evidence suggests that myofibroblasts and senescent myofibroblasts, rather than being resistant to apoptosis, are actually primed for apoptosis owing to concomitant activation of cell death signalling pathways; these cells are poised to apoptose when survival pathways are inhibited. This knowledge of apoptotic priming has paved the way for new therapies that trigger apoptosis in myofibroblasts by blocking pro-survival mechanisms, target senescent myofibroblast for apoptosis or promote the reprogramming of myofibroblasts into scar-resolving cells. These novel strategies are not only poised to prevent progressive tissue scarring, but also have the potential to reverse established fibrosis and to regenerate chronically injured tissues.
Collapse
Affiliation(s)
- Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Liu T, Gonzalez De Los Santos F, Zhao Y, Wu Z, Rinke AE, Kim KK, Phan SH. Telomerase reverse transcriptase ameliorates lung fibrosis by protecting alveolar epithelial cells against senescence. J Biol Chem 2019; 294:8861-8871. [PMID: 31000627 DOI: 10.1074/jbc.ra118.006615] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/11/2019] [Indexed: 11/06/2022] Open
Abstract
Mutations in the genes encoding telomerase reverse transcriptase (TERT) and telomerase's RNA components as well as shortened telomeres are risk factors for idiopathic pulmonary fibrosis, where repetitive injury to the alveolar epithelium is considered a key factor in pathogenesis. Given the importance of TERT in stem cells, we hypothesized that TERT plays an important role in epithelial repair and that its deficiency results in exacerbation of fibrosis by impairing this repair/regenerative process. To evaluate the role of TERT in epithelial cells, we generated type II alveolar epithelial cell (AECII)-specific TERT conditional knockout (SPC-Tert cKO) mice by crossing floxed Tert mice with inducible SPC-driven Cre mice. SPC-Tert cKO mice did not develop pulmonary fibrosis spontaneously up to 9 months of TERT deficiency. However, upon bleomycin treatment, they exhibited enhanced lung injury, inflammation, and fibrosis compared with control mice, accompanied by increased pro-fibrogenic cytokine expression but without a significant effect on AECII telomere length. Moreover, selective TERT deficiency in AECII diminished their proliferation and induced cellular senescence. These findings suggest that AECII-specific TERT deficiency enhances pulmonary fibrosis by heightening susceptibility to bleomycin-induced epithelial injury and diminishing epithelial regenerative capacity because of increased cellular senescence. We confirmed evidence for increased AECII senescence in idiopathic pulmonary fibrosis lungs, suggesting potential clinical relevance of the findings from our animal model. Our results suggest that TERT has a protective role in AECII, unlike its pro-fibrotic activity, observed previously in fibroblasts, indicating that TERT's role in pulmonary fibrosis is cell type-specific.
Collapse
Affiliation(s)
| | | | | | - Zhe Wu
- From the Departments of Pathology and
| | | | - Kevin K Kim
- Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | | |
Collapse
|
8
|
Razdan N, Vasilopoulos T, Herbig U. Telomere dysfunction promotes transdifferentiation of human fibroblasts into myofibroblasts. Aging Cell 2018; 17:e12838. [PMID: 30244523 PMCID: PMC6260909 DOI: 10.1111/acel.12838] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/25/2018] [Accepted: 08/05/2018] [Indexed: 12/13/2022] Open
Abstract
Cells that had undergone telomere dysfunction-induced senescence secrete numerous cytokines and other molecules, collectively called the senescence-associated secretory phenotype (SASP). Although certain SASP factors have been demonstrated to promote cellular senescence in neighboring cells in a paracrine manner, the mechanisms leading to bystander senescence and the functional significance of these effects are currently unclear. Here, we demonstrate that TGF-β1, a component of the SASP, causes telomere dysfunction in normal somatic human fibroblasts in a Smad3/NOX4/ROS-dependent manner. Surprisingly, instead of activating cellular senescence, TGF-β1-induced telomere dysfunction caused fibroblasts to transdifferentiate into α-SMA-expressing myofibroblasts, a mesenchymal and contractile cell type that is critical for wound healing and tissue repair. Despite the presence of dysfunctional telomeres, transdifferentiated cells acquired the ability to contract collagen lattices and displayed a gene expression signature characteristic of functional myofibroblasts. Significantly, the formation of dysfunctional telomeres and downstream p53 signaling was necessary for myofibroblast transdifferentiation, as suppressing telomere dysfunction by expression of hTERT, inhibiting the signaling pathways that lead to stochastic telomere dysfunction, and suppressing p53 function prevented the generation of myofibroblasts in response to TGF-β1 signaling. Furthermore, inducing telomere dysfunction using shRNA against TRF2 also caused cells to develop features that are characteristic of myofibroblasts, even in the absence of exogenous TGF-β1. Overall, our data demonstrate that telomere dysfunction is not only compatible with cell functionality, but they also demonstrate that the generation of dysfunctional telomeres is an essential step for transdifferentiation of human fibroblasts into myofibroblasts.
Collapse
Affiliation(s)
- Neetu Razdan
- New Jersey Medical School, Cancer Institute of New Jersey-Newark; Rutgers Biomedical and Health Sciences; Newark New Jersey
- Department of Microbiology, Biochemistry and Molecular Genetics; Rutgers Biomedical and Health Sciences; Newark New Jersey
| | - Themistoklis Vasilopoulos
- New Jersey Medical School, Cancer Institute of New Jersey-Newark; Rutgers Biomedical and Health Sciences; Newark New Jersey
| | - Utz Herbig
- New Jersey Medical School, Cancer Institute of New Jersey-Newark; Rutgers Biomedical and Health Sciences; Newark New Jersey
- Department of Microbiology, Biochemistry and Molecular Genetics; Rutgers Biomedical and Health Sciences; Newark New Jersey
| |
Collapse
|
9
|
Liu T, Yu H, Ding L, Wu Z, Gonzalez De Los Santos F, Liu J, Ullenbruch M, Hu B, Martins V, Phan SH. Conditional Knockout of Telomerase Reverse Transcriptase in Mesenchymal Cells Impairs Mouse Pulmonary Fibrosis. PLoS One 2015; 10:e0142547. [PMID: 26555817 PMCID: PMC4640706 DOI: 10.1371/journal.pone.0142547] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/25/2015] [Indexed: 12/31/2022] Open
Abstract
Telomerase is typically expressed in cellular populations capable of extended replication, such as germ cells, tumor cells, and stem cells, but is also induced in tissue injury, repair and fibrosis. Its catalytic component, telomerase reverse transcriptase (TERT) is induced in lung fibroblasts from patients with fibrotic interstitial lung disease and in rodents with bleomycin-induced pulmonary fibrosis. To evaluate the fibroblast specific role of TERT in pulmonary fibrosis, transgenic mice bearing a floxed TERT allele were generated, and then crossed with an inducible collagen α2(I)-Cre mouse line to generate fibroblast specific TERT conditional knockout mice. TERT-specific deficiency in mesenchymal cells caused attenuation of pulmonary fibrosis as manifested by reduced lung hydroxyproline content, type I collagen and α-smooth muscle actin mRNA levels. The TERT-deficient mouse lung fibroblasts displayed decreased cell proliferative capacity and higher susceptibility to induced apoptosis compared with control cells. Additionally TERT deficiency was associated with heightened α-smooth muscle actin expression indicative of myofibroblast differentiation. However the impairment of cell proliferation and increased susceptibility to apoptosis would cause a reduction in the myofibroblast progenitor population necessary to mount a successful myofibroblast-dependent fibrotic response. These findings identified a key role for TERT in fibroblast proliferation and survival essential for pulmonary fibrosis.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Hongfeng Yu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Lin Ding
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Zhe Wu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | | | - Jianhua Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Biao Hu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Vanessa Martins
- Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Sem H. Phan
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
10
|
Arish N, Cohen PY, Golan-Gerstl R, Fridlender Z, Dayan MR, Zisman P, Breuer R, Wallach-Dayan SB. Overexpression of Telomerase Protects Human and Murine Lung Epithelial Cells from Fas- and Bleomycin-Induced Apoptosis via FLIP Upregulation. PLoS One 2015; 10:e0126730. [PMID: 25951185 PMCID: PMC4423936 DOI: 10.1371/journal.pone.0126730] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 04/07/2015] [Indexed: 11/18/2022] Open
Abstract
High doses of bleomycin administered to patients with lymphomas and other tumors lead to significant lung toxicity in general, and to apoptosis of epithelial cells, in particular. Apoptosis of alveolar epithelium is an important step in the pathogenesis of bleomycin-induced pulmonary fibrosis. The Fas-FasL pathway is one of the main apoptotic pathways involved. Telomerase is a ribonucleoprotein RNA-dependent DNA polymerase complex consisting of an RNA template and a catalytic protein, telomerase reverse transcriptase (TERT). Telomerase also possess extra-telomeric roles, including modulation of transcription of anti-apoptotic genes, differentiation signals, and more. We hypothesized that telomerase overexpression affects Fas-induced epithelial cell apoptosis by an extra-telomeric role such as regulation of anti-apoptotic genes, specifically FLICE-like inhibitory protein (FLIP). Telomerase in mouse (MLE) and human (A549) lung epithelial cell lines was upregulated by transient transfection using cDNA hTERT expression vector. Telomerase activity was detected using a real-time PCR-based system. Bleomycin, and bleomycin-induced Fas-mediated apoptosis following treatment with anti-Fas activating mAb or control IgG, were assessed by Annexin V staining, FACS analysis, and confocal microscopy; caspase cleavage by Western blot; FLIP or Fas molecule detection by Western blot and flow cytometry. hTERT transfection of lung epithelial cells resulted in a 100% increase in their telomerase activity. Fas-induced lung epithelial cell apoptosis was significantly reduced in hTERT-transfected cells compared to controls in all experiments. Lung epithelial cells with increased telomerase activity had higher levels of FLIP expression but membrane Fas expression was unchanged. Upregulation of hTERT+ in human lung epithelial cells and subsequent downregulation of FLIP by shFLIP-RNA annulled hTERT-mediated resistance to apoptosis. Telomerase-mediated FLIP overexpression may be a novel mechanism to confer protection from apoptosis in bleomycin-exposed human lung epithelial cells.
Collapse
Affiliation(s)
- Nissim Arish
- Laboratory for Lung Cellular & Molecular Biology, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Pazit Y. Cohen
- Laboratory for Lung Cellular & Molecular Biology, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Regina Golan-Gerstl
- Laboratory for Lung Cellular & Molecular Biology, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Zvi Fridlender
- Laboratory for Lung Cellular & Molecular Biology, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
- Department of Pulmonary and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, United States of America
| | - Mark Richter Dayan
- Department of Emergency Medicine, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Philip Zisman
- Laboratory for Lung Cellular & Molecular Biology, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Raphael Breuer
- Laboratory for Lung Cellular & Molecular Biology, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
- Department of Pathology, Boston University School of Medicine, Boston, MA, United States of America
| | - Shulamit B. Wallach-Dayan
- Laboratory for Lung Cellular & Molecular Biology, Institute of Pulmonary Medicine, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
11
|
Liu T, Ullenbruch M, Young Choi Y, Yu H, Ding L, Xaubet A, Pereda J, Feghali-Bostwick CA, Bitterman PB, Henke CA, Pardo A, Selman M, Phan SH. Telomerase and telomere length in pulmonary fibrosis. Am J Respir Cell Mol Biol 2013; 49:260-8. [PMID: 23526226 DOI: 10.1165/rcmb.2012-0514oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In addition to its expression in stem cells and many cancers, telomerase activity is transiently induced in murine bleomycin (BLM)-induced pulmonary fibrosis with increased levels of telomerase transcriptase (TERT) expression, which is essential for fibrosis. To extend these observations to human chronic fibrotic lung disease, we investigated the expression of telomerase activity in lung fibroblasts from patients with interstitial lung diseases (ILDs), including idiopathic pulmonary fibrosis (IPF). The results showed that telomerase activity was induced in more than 66% of IPF lung fibroblast samples, in comparison with less than 29% from control samples, some of which were obtained from lung cancer resections. Less than 4% of the human IPF lung fibroblast samples exhibited shortened telomeres, whereas less than 6% of peripheral blood leukocyte samples from patients with IPF or hypersensitivity pneumonitis demonstrated shortened telomeres. Moreover, shortened telomeres in late-generation telomerase RNA component knockout mice did not exert a significant effect on BLM-induced pulmonary fibrosis. In contrast, TERT knockout mice exhibited deficient fibrosis that was independent of telomere length. Finally, TERT expression was up-regulated by a histone deacetylase inhibitor, while the induction of TERT in lung fibroblasts was associated with the binding of acetylated histone H3K9 to the TERT promoter region. These findings indicate that significant telomerase induction was evident in fibroblasts from fibrotic murine lungs and a majority of IPF lung samples, whereas telomere shortening was not a common finding in the human blood and lung fibroblast samples. Notably, the animal studies indicated that the pathogenesis of pulmonary fibrosis was independent of telomere length.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Bradykinin-induced asthmatic fibroblast/myofibroblast activities via bradykinin B2 receptor and different MAPK pathways. Eur J Pharmacol 2013; 710:100-9. [PMID: 23588115 DOI: 10.1016/j.ejphar.2013.03.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 03/21/2013] [Accepted: 03/28/2013] [Indexed: 02/05/2023]
Abstract
Bradykinin drives normal lung fibroblasts into myofibroblasts, induces fibroblast proliferation and activates mitogen activated protein kinase pathways (MAPK) but its effects on bronchial fibroblasts from asthmatics (HBAFb) have not been yet studied. We studied bradykinin-induced fibroblast proliferation and differentiation and the related intracellular mechanisms in HBAFb compared to normal bronchial fibroblasts (HNBFb). Bradykinin-stimulated HBAFb and HNBFb were used to assess: bradykinin B2 receptor expression by Western blot analysis; cell proliferation by [(3)H] thymidine incorporation; α-smooth muscle actin (SMA) expression/polymerization by Western blot and immunofluorescence; epidermal growth factor (EGF) receptor, extracellular-regulated kinase (ERK) 1/2 and p38 MAPK activation by immunoprecipitation and Western blot, respectively. Constitutive bradykinin B2 receptor and α-SMA expression was higher in HBAFb as compared to HNBFb. Bradykinin increased bradykinin B2 receptor expression in HBAFb. Bradykinin, via bradykinin B2 receptor, significantly increased fibroblast proliferation at lower concentration (10(-11)M) and α-SMA expression/polymerization at higher concentration (10(-6)M) in both cells. Bradykinin increased ERK1/2 and p38 phosphorylation via bradykinin B2 receptor; EGF receptor inhibitor AG1478 and panmetalloproteinase inhibitor GM6001 blocked bradykinin-induced ERK1/2 activation but not p38 phosphorylation. Bradykinin, via bradykinin B2 receptor, induced EGF receptor phosphorylation that was suppressed by AG1478. In HBAFb AG1478, GM6001, the ERK1/2-inhibitor U0126 and the p38 inhibitor SB203580 suppressed bradykinin-induced cell proliferation, but only SB203580 reduced myofibroblast differentiation. These data indicate that bradykinin is actively involved in asthmatic bronchial fibroblast proliferation and differentiation, through MAPK pathways and EGF receptor transactivation, by which bradykinin may contribute to airway remodeling in asthma, opening new horizons for potential therapeutic implications in asthmatic patients.
Collapse
|
13
|
In vitro transdifferentiation of umbilical cord stem cells into cardiac myocytes: Role of growth factors. EGYPTIAN JOURNAL OF CRITICAL CARE MEDICINE 2013. [DOI: 10.1016/j.ejccm.2013.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
14
|
Le Saux CJ, Davy P, Brampton C, Ahuja SS, Fauce S, Shivshankar P, Nguyen H, Ramaseshan M, Tressler R, Pirot Z, Harley CB, Allsopp R. A novel telomerase activator suppresses lung damage in a murine model of idiopathic pulmonary fibrosis. PLoS One 2013; 8:e58423. [PMID: 23516479 PMCID: PMC3597721 DOI: 10.1371/journal.pone.0058423] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 02/06/2013] [Indexed: 11/24/2022] Open
Abstract
The emergence of diseases associated with telomere dysfunction, including AIDS, aplastic anemia and pulmonary fibrosis, has bolstered interest in telomerase activators. We report identification of a new small molecule activator, GRN510, with activity ex vivo and in vivo. Using a novel mouse model, we tested the potential of GRN510 to limit fibrosis induced by bleomycin in mTERT heterozygous mice. Treatment with GRN510 at 10 mg/kg/day activated telomerase 2–4 fold both in hematopoietic progenitors ex vivo and in bone marrow and lung tissue in vivo, respectively. Telomerase activation was countered by co-treatment with Imetelstat (GRN163L), a potent telomerase inhibitor. In this model of bleomycin-induced fibrosis, treatment with GRN510 suppressed the development of fibrosis and accumulation of senescent cells in the lung via a mechanism dependent upon telomerase activation. Treatment of small airway epithelial cells (SAEC) or lung fibroblasts ex vivo with GRN510 revealed telomerase activating and replicative lifespan promoting effects only in the SAEC, suggesting that the mechanism accounting for the protective effects of GRN510 against induced lung fibrosis involves specific types of lung cells. Together, these results support the use of small molecule activators of telomerase in therapies to treat idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Claude Jourdan Le Saux
- University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Philip Davy
- John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Christopher Brampton
- John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Seema S. Ahuja
- University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Steven Fauce
- Geron Corporation, Menlo Park, California, United States of America
- Beckman Coulter, Inc., Brea, California, United States of America
| | - Pooja Shivshankar
- University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Hieu Nguyen
- John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
| | | | - Robert Tressler
- Geron Corporation, Menlo Park, California, United States of America
- Cellerant, Redwood City, California, United States of America
| | - Zhu Pirot
- Geron Corporation, Menlo Park, California, United States of America
| | - Calvin B. Harley
- Geron Corporation, Menlo Park, California, United States of America
| | - Richard Allsopp
- John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This review seeks to inform readers of evolving concepts of ageing-associated risks for developing interstitial lung disease (ILD) and current approaches to the diagnosis and management of ILD in elderly patients. RECENT FINDINGS Various aspects of cellular and immune senescence have been identified that may explain the increased susceptibility of the elderly to developing fibrotic lung disease. New guidelines have been recently published concerning the diagnosis and management of idiopathic pulmonary fibrosis (IPF), which is highly prevalent in elderly patients. Nontransplant therapies that can have a significant impact on disease progression for patients with IPF have yet to be identified. Additionally, evidence is accumulating that abnormal gastroesophageal reflux and microaspiration may play a role in IPF pathogenesis. SUMMARY High-resolution computed tomographic scanning of the thorax can play a key role in making a specific ILD diagnosis and be used to make a confident diagnosis of various forms of ILD, especially IPF, when combined with a consistent clinical presentation. Management of ILD in the elderly should be not only disease specific but potentially therapeutic, and supportive interventions should be tailored to each individual patient and not entail significant risk of adverse complications, especially for the frail elderly patient.
Collapse
Affiliation(s)
- Keith C Meyer
- Section of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792-9988, USA.
| |
Collapse
|
16
|
Waisberg DR, Parra ER, Barbas-Filho JV, Fernezlian S, Capelozzi VL. Increased fibroblast telomerase expression precedes myofibroblast α-smooth muscle actin expression in idiopathic pulmonary fibrosis. Clinics (Sao Paulo) 2012; 67:1039-46. [PMID: 23018301 PMCID: PMC3438244 DOI: 10.6061/clinics/2012(09)10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 05/10/2012] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE This study sought to identify the relationship between fibroblast telomerase expression, myofibroblasts, and telomerase-mediated regulatory signals in idiopathic pulmonary fibrosis. METHODS Thirty-four surgical lung biopsies, which had been obtained from patients with idiopathic pulmonary fibrosis and histologically classified as usual interstitial pneumonia, were examined. Immunohistochemistry was used to evaluate fibroblast telomerase expression, myofibroblast α-smooth muscle actin expression and the tissue expression of inter leu kin-4, transforming growth factor-β, and basic fibroblast growth factor. The point-counting technique was used to quantify the expression of these markers in unaffected, collapsed, mural fibrosis, and honeycombing areas. The results were correlated to patient survival. RESULTS Fibroblast telomerase expression and basic fibroblast growth factor tissue expression were higher in collapsed areas, whereas myofibroblast expression and interleukine-4 tissue expression were higher in areas of mural fibrosis. Transforming growth factor-β expression was higher in collapsed, mural fibrosis and honeycombing areas in comparison to unaffected areas. Positive correlations were found between basic fibroblast growth factor tissue expression and fibroblast telomerase expression and between interleukin-4 tissue expression and myofibroblast α-smooth muscle actin expression. Negative correlations were observed between interleukin-4 expression and basic fibroblast growth factor tissue expression in areas of mural fibrosis. Myofibroblast α-smooth muscle actin expression and interleukin-4 tissue expression in areas of mural fibrosis were negatively associated with patient survival. CONCLUSION Fibroblast telomerase expression is higher in areas of early remodeling in lung tissues demonstrating typical interstitial pneumonia, whereas myofibroblast α-smooth muscle actin expression predominates in areas of late remodeling. These events seem to be regulated by basic fibroblast growth factor and interleukin-4 tissue expression, respectively.
Collapse
Affiliation(s)
- Daniel Reis Waisberg
- Department of Pathology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | | | | |
Collapse
|
17
|
Richardson GD, Breault D, Horrocks G, Cormack S, Hole N, Owens WA. Telomerase expression in the mammalian heart. FASEB J 2012; 26:4832-40. [PMID: 22919071 PMCID: PMC3509052 DOI: 10.1096/fj.12-208843] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
While the mammalian heart has low, but functionally significant, levels of telomerase expression, the cellular population responsible remains incompletely characterized. This study aimed to identify the cell types responsible for cardiac telomerase activity in neonatal, adult, and cryoinjured adult hearts using transgenic mice expressing green fluorescent protein (GFP), driven by the promoter for murine telomerase reverse transcriptase (mTert), which is a necessary and rate-limiting component of telomerase. A rare population of mTert-GFP-expressing cells was identified that possessed all detectable cardiac telomerase RNA and telomerase activity. It was heterogeneous and included cells coexpressing markers of cardiomyocytic, endothelial, and mesenchymal lineages, putative cardiac stem cell markers, and, interestingly, cardiomyocytes with a differentiated phenotype. Quantification using both flow cytometry and immunofluorescence identified a significant decline in mTert-GFP cells in adult animals compared to neonates (∼9- and ∼20-fold, respectively). Cardiac injury resulted in a ∼6.45-fold expansion of this population (P<0.005) compared with sham-operated controls. This study identifies the cells responsible for cardiac telomerase activity, demonstrates a significant diminution with age but a marked response to injury, and, given the relationship between telomerase activity and stem cell populations, suggests that they represent a potential target for further investigation of cardiac regenerative potential.—Richardson, G. D., Breault, D., Horrocks, G., Cormack, S., Hole, N., Owens, W. A. Telomerase expression in the mammalian heart.
Collapse
Affiliation(s)
- Gavin D Richardson
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK.
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
|
19
|
Abstract
Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
Collapse
Affiliation(s)
- Thomas A Wynn
- Immunopathogenesis Section, Program in Barrier Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
20
|
Chilosi M, Poletti V, Rossi A. The pathogenesis of COPD and IPF: distinct horns of the same devil? Respir Res 2012; 13:3. [PMID: 22235752 PMCID: PMC3282644 DOI: 10.1186/1465-9921-13-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/11/2012] [Indexed: 01/08/2023] Open
Abstract
New paradigms have been recently proposed in the pathogenesis of both chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), evidencing surprising similarities between these deadly diseases, despite their obvious clinical, radiological and pathologic differences. There is growing evidence supporting a "double hit" pathogenic model where in both COPD and IPF the cumulative action of an accelerated senescence of pulmonary parenchyma (determined by either telomere dysfunction and/or a variety of genetic predisposing factors), and the noxious activity of cigarette smoke-induced oxidative damage are able to severely compromise the regenerative potential of two pulmonary precursor cell compartments (alveolar epithelial precursors in IPF, mesenchymal precursor cells in COPD/emphysema). The consequent divergent derangement of signalling pathways involved in lung tissue renewal (mainly Wnt and Notch), can eventually lead to the distinct abnormal tissue remodelling and functional impairment that characterise the alveolar parenchyma in these diseases (irreversible fibrosis and bronchiolar honeycombing in IPF, emphysema and airway chronic inflammation in COPD).
Collapse
Affiliation(s)
- Marco Chilosi
- Department of Pathology, University of Verona, Italy.
| | | | | |
Collapse
|
21
|
Maeda T, Kurita R, Yokoo T, Tani K, Makino N. Telomerase inhibition promotes an initial step of cell differentiation of primate embryonic stem cell. Biochem Biophys Res Commun 2011; 407:491-4. [DOI: 10.1016/j.bbrc.2011.03.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 03/10/2011] [Indexed: 12/22/2022]
|
22
|
Granick M, Kimura M, Kim S, Daniali L, Cao X, Herbig U, Aviv A. Telomere dynamics in keloids. EPLASTY 2011; 11:e15. [PMID: 21436892 PMCID: PMC3060056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
OBJECTIVE Little is known about telomere dynamics in keloids. As keloid formation is dependent on cell replication, in theory telomeres should be shorter in keloids than in normal skin. We examined this concept in the present study. METHODS We measured by Southern blot analysis telomere length in keloids and in adjacent normal skin of 16 individuals. When available, we also measured telomere length in blood (leukocytes) and subcutaneous fat. RESULTS Telomere length was highly variable among individuals but highly correlated among tissues (cells) within the individual. The mean telomere length in the keloids was longer than that in the adjacent normal skin and displayed a length gradient, with the mean length of telomeres shorter just below the epidermis and longer at the base of the keloids. No apparent telomerase activity was detected in the keloids. CONCLUSIONS Our findings suggest a transient activation of telomerase, the reverse transcriptase that prevents telomere shortening, probably during the early phase of keloid formation. The activation of telomerase serves to maintain (or even elongate) telomere length in the keloid. However, telomerase activity is repressed in the fully developed keloid.
Collapse
Affiliation(s)
- Mark Granick
- aDivision of Plastic Surgery, Department of Surgery
| | | | - Soyeon Kim
- cDepartment of Preventive Medicine and Community Health
| | - Lily Daniali
- aDivision of Plastic Surgery, Department of Surgery
| | | | - Utz Herbig
- dDepartment of Microbiology and Molecular Genetics, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark
| | - Abraham Aviv
- bCenter of Human Development and Aging,Correspondence:
| |
Collapse
|
23
|
Cassar L, Li H, Jiang FX, Liu JP. TGF-beta induces telomerase-dependent pancreatic tumor cell cycle arrest. Mol Cell Endocrinol 2010; 320:97-105. [PMID: 20138964 DOI: 10.1016/j.mce.2010.02.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Revised: 01/27/2010] [Accepted: 02/02/2010] [Indexed: 12/21/2022]
Abstract
Recent studies suggest that transforming growth factor beta (TGF-beta) inhibits telomerase activity by repression of the telomerase reverse transcriptase (TERT) gene. In this report, we show that TGF-beta induces TERT repression-dependent apoptosis in pancreatic tumor, vascular smooth muscle, and cervical cancer cell cultures. TGF-beta activates Smad3 signaling, induces TERT gene repression and results in G1/S phase cell cycle arrest and apoptosis. TERT over-expression stimulates the G1/S phase transition and alienates TGF-beta-induced cell cycle arrest and apoptosis. Our data suggest that telomere maintenance is a limiting factor of the transition of the cell cycle. TGF-beta triggers cell cycle arrest and death by a mechanism involving telomerase deregulation of telomere maintenance.
Collapse
Affiliation(s)
- Lucy Cassar
- Department of Immunology, Monash University, Central Clinical School, AMREP, Commercial Road, Melbourne, Victoria 3004, Australia.
| | | | | | | |
Collapse
|
24
|
Hu B, Wu YM, Wu Z, Phan SH. Nkx2.5/Csx represses myofibroblast differentiation. Am J Respir Cell Mol Biol 2009; 42:218-26. [PMID: 19395679 DOI: 10.1165/rcmb.2008-0404oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Myofibroblasts are known to play key roles in wound healing and fibrosis. They are thought to arise de novo from fibroblasts, and are characterized by induction of alpha-smooth muscle actin (alpha-SMA) expression. The homeobox transcription factor Csx/Nkx2.5 is critical in heart development and cardiogenesis by inducing expression of genes associated with cardiomyocyte differentiation. Here, we report a novel repressor activity of Nkx2.5 on myofibroblast differentiation. Because a key marker of myofibroblast differentiation is expression of the alpha-SMA gene, we first scanned its promoter region for possible cis-acting elements. The results show three potential binding sites (designated Nkx2.5 element [NKE]-1, -2, and -3) containing the Nkx2.5/Csx consensus binding motif (5'-TNNAGTG-3'). To determine their functional importance, site-directed mutagenesis directed at these elements individually and in combination indicated that mutation of NKE1 and -3 significantly enhanced alpha-SMA gene promoter activity, whereas mutation of NKE2 did not have a significant effect. The results of gel shift assays confirmed that Nkx2.5 could bind to both NKE1 and -3, but not to NKE2. Consistent with the mutagenesis studies, ectopically induced expression of Nkx2.5 inhibited alpha-SMA gene expression. Analysis of nkx2.5 gene expression indicates that it was significantly induced by basic fibroblast growth factor treatment of isolated lung fibroblasts in vitro. In vivo, lung Nkx2.5 expression was significantly diminished in bleomycin-induced pulmonary fibrosis. These findings are consistent with a novel function for Nkx2.5 as a repressor of alpha-SMA gene transcription, which may be of homeostatic significance as a means of suppressing myofibroblast differentiation in the absence of tissue injury.
Collapse
Affiliation(s)
- Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | | | | | |
Collapse
|
25
|
Dolgachev VA, Ullenbruch MR, Lukacs NW, Phan SH. Role of stem cell factor and bone marrow-derived fibroblasts in airway remodeling. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:390-400. [PMID: 19147822 DOI: 10.2353/ajpath.2009.080513] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent evidence suggests that bone marrow-derived fibroblasts are involved in airway remodeling in asthma, but the role and mechanism of recruitment of these fibroblasts remains unclear. Stem cell factor (SCF), a key factor in the propagation of hematopoietic stem cells, is important in the process of airway remodeling as well. To test the hypothesis that SCF is involved in the recruitment and differentiation of bone marrow-derived progenitor cells, GFP-bone marrow chimeric mice were created. These mice were then sensitized and chronically challenged with cockroach antigen to induce chronic airway disease. Fluorescence microscopy revealed an influx of significant numbers of GFP-expressing fibroblasts in the airways of these mice, which was confirmed by flow cytometric analysis of cells co-expressing both GFP and collagen I. These cells preferentially expressed c-kit, interleukin-31 receptor, and telomerase reverse transcriptase when compared with control lung-derived fibroblasts. Interestingly, SCF stimulated interleukin-31 receptor expression in bone marrow cells, whereas interleukin-31 strongly induced telomerase reverse transcriptase expression in fibroblasts. Treatment with neutralizing antibodies to SCF significantly reduced airway remodeling and suppressed the recruitment of these bone marrow-derived cells to the lung. Thus SCF in conjunction with interleukin-31 may play a significant role in airway remodeling by promoting the recruitment of bone marrow-derived fibroblast precursors into the lung with the capacity to promote lung myofibroblast differentiation.
Collapse
Affiliation(s)
- Vladislav A Dolgachev
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA.
| | | | | | | |
Collapse
|
26
|
Abstract
The pulmonary circulation represents a unique vascular bed, receiving 100% of the cardiac output while maintaining low blood pressure. Multiple different cell types, including endothelium, smooth muscle, and fibroblasts, contribute to normal vascular function, and to the vascular response to injury. Our understanding of the basic cell biology of these various cell types, and the roles they play in vascular homeostasis and disease, remains quite limited despite several decades of study. Recent advances in approaches that enable the mapping of cell origin and the study of the molecular basis of structure and function have resulted in a rapid accumulation of new information that is essential to vascular biology. A recent National Institutes of Health workshop was held to discuss emerging concepts in lung vascular biology. The findings of this workshop are summarized in this article.
Collapse
|
27
|
Abstract
Despite their importance in fibrosis, the origin of fibroblasts and the genesis of the various subpopulations characterized by distinct phenotypes remain unclear. Various studies have described distinct and relatively stable phenotypes in fibroblasts isolated from lung tissue undergoing remodeling, which were not present in the normal intact tissue. This indicates a process by which these distinct fibroblast subpopulations could arise de novo from resident lung progenitors or precursor cells and/or be recruited from distal organs, such as the bone marrow. Evidence for these possibilities is reviewed, but there is as yet incomplete understanding of the precise precursor cells and the potential interrelationships between the various phenotypes, especially as to how they relate to the distinct myofibroblast phenotype. Moreover, the complexity of the mechanism for the genesis of these phenotypes, such as the myofibroblast, is highlighted by the multilevel regulation of the differentiation process, with evidence for the importance of multiple signaling pathways, transcription factors, and epigenetic mechanisms. Future studies into these various unsettled areas are essential to provide further insights that may help provide the pathway for novel translational approaches.
Collapse
|
28
|
Liu T, Chung MJ, Ullenbruch M, Yu H, Jin H, Hu B, Choi YY, Ishikawa F, Phan SH. Telomerase activity is required for bleomycin-induced pulmonary fibrosis in mice. J Clin Invest 2008; 117:3800-9. [PMID: 18008008 DOI: 10.1172/jci32369] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Accepted: 09/12/2007] [Indexed: 01/01/2023] Open
Abstract
In addition to its well-known expression in the germline and in cells of certain cancers, telomerase activity is induced in lung fibrosis, although its role in this process is unknown. To identify the pathogenetic importance of telomerase in lung fibrosis, we examined the effects of telomerase reverse transcriptase (TERT) deficiency in a murine model of pulmonary injury. TERT-deficient mice showed significantly reduced lung fibrosis following bleomycin (BLM) insult. This was accompanied by a significant reduction in expression of lung alpha-SMA, a marker of myofibroblast differentiation. Furthermore, lung fibroblasts isolated from BLM-treated TERT-deficient mice showed significantly decreased proliferation and increased apoptosis rates compared with cells isolated from control mice. Transplantation of WT BM into TERT-deficient mice restored BLM-induced lung telomerase activity and fibrosis to WT levels. Conversely, transplantation of BM from TERT-deficient mice into WT recipients resulted in reduced telomerase activity and fibrosis. These findings suggest that induction of telomerase in injured lungs may be caused by BM-derived cells, which appear to play an important role in pulmonary fibrosis. Moreover, TERT induction is associated with increased survival of lung fibroblasts, which favors the development of fibrosis instead of injury resolution.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Petrov VV, van Pelt JF, Vermeesch JR, Van Duppen VJ, Vekemans K, Fagard RH, Lijnen PJ. TGF-beta1-induced cardiac myofibroblasts are nonproliferating functional cells carrying DNA damages. Exp Cell Res 2008; 314:1480-94. [PMID: 18295203 DOI: 10.1016/j.yexcr.2008.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 12/03/2007] [Accepted: 01/09/2008] [Indexed: 01/07/2023]
Abstract
TGF-beta1 induces differentiation and total inhibition of cardiac MyoFb cell division and DNA synthesis. These effects of TGF-beta1 are irreversible. Inhibition of MyoFb proliferation is accompanied with the expression of Smad1, Mad1, p15Ink4B and total inhibition of telomerase activity. Surprisingly, TGF-beta1-activated MyoFbs are growth-arrested not only at G1-phase but also at S-phase of the cell cycle. Staining with TUNEL indicates that these cells carry DNA damages. However, the absolute majority of MyoFbs are non-apoptotic cells as established with two apoptosis-specific methods, flow cytometry and caspase-dependent cleavage of cytokeratin 18. Expression in MyoFbs of proliferative cell nuclear antigen even in the absence of serum confirms that these MyoFbs perform repair of DNA damages. These results suggest that TGF-beta1-activated MyoFbs can be growth-arrested by two checkpoints, the G1/S checkpoint, which prevents cells from entering S-phase and the intra-S checkpoint, which is activated by encountering DNA damage during the S phase or by unrepaired damage that escapes the G1/S checkpoint. Despite carrying of the DNA damages TGF-beta1-activated MyoFbs are highly functional cells producing lysyl oxidase and contracting the collagen matrix.
Collapse
Affiliation(s)
- Victor V Petrov
- Department of Heart Diseases, University of Leuven (KULeuven), Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
30
|
Hagood JS, Olman MA. Muscle fatigue: MK2 signaling and myofibroblast differentiation. Am J Respir Cell Mol Biol 2007; 37:503-6. [PMID: 17940320 DOI: 10.1165/rcmb.2007-0005ed] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
31
|
Gharaee-Kermani M, Gyetko MR, Hu B, Phan SH. New Insights into the Pathogenesis and Treatment of Idiopathic Pulmonary Fibrosis: A Potential Role for Stem Cells in the Lung Parenchyma and Implications for Therapy. Pharm Res 2007; 24:819-41. [PMID: 17333393 DOI: 10.1007/s11095-006-9216-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Accepted: 12/13/2006] [Indexed: 02/06/2023]
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronic, progressive, and often fatal form of interstitial lung disease. It is characterized by injury with loss of lung epithelial cells and abnormal tissue repair, resulting in replacement of normal functional tissue, abnormal accumulation of fibroblasts and myofibroblasts, deposition of extracellular matrix, and distortion of lung architecture which results in respiratory failure. Despite improvements in the diagnostic approach to IPF and active research in recent years, the molecular mechanisms of the disease remain poorly understood. This highly lethal lung disorder continues to pose major clinical challenges since an effective therapeutic regimen has yet to be identified and developed. For example, a treatment modality has been based on the assumption that IPF is a chronic inflammatory disease, yet most available anti-inflammatory drugs are not effective in treating it. Hence researchers are now focusing on understanding alternative underlying mechanisms involved in the pathogenesis of IPF in the hope of discovering potentially new pharmaceutical targets. This paper will focus on lung tissue repair, regeneration, remodeling, and cell types that may be important to consider in therapeutic interventions and includes a more detailed discussion of the potential targets of current therapeutic attack in pulmonary fibrosis. The discovery that adult bone marrow stem cells can contribute to the formation of differentiated cell types in other tissues, especially after injury, implies that they have the potential to participate in tissue remodeling, and perhaps regeneration. The current promise of the use of adult stem cells for tissue regeneration, and the belief that once irreversibly damaged tissue could be restored to a normal functional capacity using stem cell-based therapy, suggests a novel approach for treatment of diverse chronic diseases. However this optimism is tempered by current evidence that the pathogenesis of pulmonary fibrosis may involve the recruitment of bone marrow-derived fibroblasts, which are the key contributors to the pathogenesis of this chronic progressive disorder. Nevertheless, stem cell-related therapies are widely viewed as promising treatment options for patients suffering from various types of pulmonary diseases. Gender mismatched bone marrow or lung transplant recipients serve as natural populations in which to study the role of bone marrow-derived stem cells in recovery from pulmonary diseases. Understanding the mechanism of recruitment of stem cells to sites of injury, and their involvement in tissue repair, regeneration, and remodeling may offer a novel therapeutic target for developing more effective treatments against this fatal disorder. This article reviews the new concepts in the pathogenesis, current and future treatment options of pulmonary fibrosis, and the recent advances regarding the roles of stem cells in lung tissue repair, regeneration, and remodeling.
Collapse
Affiliation(s)
- Mehrnaz Gharaee-Kermani
- Division of Pulmonary Medicine & Critical Care, Department of Internal Medicine, University of Michigan Medical School, 2215 Fuller Rd. VAMC 11R, Ann Arbor, Michigan 48105, USA.
| | | | | | | |
Collapse
|
32
|
Ramos C, Montaño M, Becerril C, Cisneros-Lira J, Barrera L, Ruíz V, Pardo A, Selman M. Acidic fibroblast growth factor decreases α-smooth muscle actin expression and induces apoptosis in human normal lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2006; 291:L871-9. [PMID: 16766579 DOI: 10.1152/ajplung.00019.2006] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Fibroblast/myofibroblast expansion is critical in the pathogenesis of pulmonary fibrosis. To date, most research has focused on profibrotic mediators, whereas studies on antifibrotic factors are scanty. In this study, we explored the effects of acidic fibroblast growth factor (FGF-1) and FGF-1 plus heparin (FGF-1+H) on fibroblast growth rate, apoptosis, and myofibroblast differentiation. Heparin was used because it participates in FGF-1 signaling. Growth rate was evaluated by WST-1 colorimetric assay, DNA synthesis by [3H]thymidine incorporation, and apoptosis by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and cleaved caspase 3. Expression of α-smooth muscle actin (α-SMA) was examined by immunocytochemistry, flow cytometry, real-time PCR, and immunoblotting. Despite the induction of DNA synthesis, FGF-1+H significantly reduced fibroblast growth rate. This correlated with a significant increase in apoptosis, evaluated by TUNEL (41.6 ± 1.4% vs. 12.5 ± 0.6% from controls; P < 0.01) and cleaved caspase 3 (295 ± 32 vs. 200 ± 19 ng/106cells from controls; P < 0.05). Double immunostaining (α-SMA-TUNEL) revealed that the levels of induced apoptosis were similar in fibroblasts and myofibroblasts. FGF-1+H inhibited the effect of TGF-β1 on myofibroblast differentiation. α-SMA-positive cells were reduced by immunocytochemistry from 44.5 ± 6.5% to 10.9 ± 1.9% and by flow cytometry from 30.6 ± 2.5% to 7.7 ± 0.6% ( P < 0.01). Also, FGF-1+H significantly inhibited the TGF-β1 induction of α-SMA quantified by real-time PCR and Western blot. This decrease was associated with a 35% reduction in TGF-β1-induced collagen gel contraction. The effect of FGF-1+H was mediated by a significant decrease of TGF-β1-induced Smad2 phosphorylation. FGF-1 alone exhibited similar but lower effects. These findings suggest that FGF-1 can have an antifibrogenic role, inducing apoptosis of fibroblasts and inhibiting myofibroblast differentiation.
Collapse
Affiliation(s)
- Carlos Ramos
- Instituto Nacional de Enfermedades Respiratorias, Tlalpan 4502, CP 14080, México DF, México
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Hu B, Wu Z, Liu T, Ullenbruch MR, Jin H, Phan SH. Gut-enriched Krüppel-like factor interaction with Smad3 inhibits myofibroblast differentiation. Am J Respir Cell Mol Biol 2006; 36:78-84. [PMID: 16858008 PMCID: PMC1899300 DOI: 10.1165/rcmb.2006-0043oc] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Gut-enriched Krüppel-like factor (GKLF) has been reported to partially inhibit alpha-smooth muscle actin (alpha-SMA) gene transcription by competing for binding to the TGF-beta control element (TCE) with known activators such as Sp1 and other Krüppel-like factors. This incomplete inhibition via the TCE suggests an additional mechanism, which was evaluated in this study. The results showed that an alpha-SMA promoter mutated in the TCE remained susceptible to inhibition by GKLF in rat lung fibroblasts consistent with the existence of an additional TCE-independent mechanism. Since TGF-beta- induced alpha-SMA expression is Smad3-dependent, potential interaction between GKLF and Smad3 was examined as a basis for this additional inhibitory mechanism. Co-immunoprecipitation and yeast two-hybrid assays revealed that GKLF could bind Smad3 through the Smad3 MH2 domain. Electrophoretic mobility shift assays and ChIP assay indicated that this GKLF-Smad3 interaction inhibited Smad3 binding to the Smad3-binding element (SBE) in the alpha-SMA promoter, and the activity of an SBE containing artificial promoter. Further analysis using smad3(-/-) fibroblasts confirmed that the TCE-independent inhibition by GKLF was dependent on Smad3. These data taken together suggest that in addition to inhibition via the TCE, GKLF represses alpha-SMA gene expression by interacting with Smad3 to prevent Smad3 binding to the SBE. It represents the first evidence to directly link GKLF with Smad3, a key intracellular mediator of TGF-beta signaling, which should lead to a clearer understanding of the mechanism of how GKLF regulates TGF-beta-induced gene expression.
Collapse
Affiliation(s)
- Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0602, USA
| | | | | | | | | | | |
Collapse
|
34
|
Schissel SL, Layne MD. Telomerase, Myofibroblasts, and Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2006; 34:520-2. [PMID: 16618786 DOI: 10.1165/rcmb.f312] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
35
|
Kim SJ, Song JS, Song CH, Yoo JH, Kim BS. Adenovirus-Mediated Antisense Vector-Induced Inhibition of Human Telomerase RNA May Induce Differentiation of CD34+ Cells. THE KOREAN JOURNAL OF HEMATOLOGY 2006. [DOI: 10.5045/kjh.2006.41.3.172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Seok Jin Kim
- Department of Internal Medicine, Korea University Medical Center, Instititue of Stem Cell Research, Korea University, Seoul, Korea
| | - Joon-Seok Song
- Department of Internal Medicine, Korea University Medical Center, Instititue of Stem Cell Research, Korea University, Seoul, Korea
| | - Chang Hee Song
- Department of Internal Medicine, Korea University Medical Center, Instititue of Stem Cell Research, Korea University, Seoul, Korea
| | - Ji Hyun Yoo
- Department of Internal Medicine, Korea University Medical Center, Instititue of Stem Cell Research, Korea University, Seoul, Korea
| | - Byung Soo Kim
- Department of Internal Medicine, Korea University Medical Center, Instititue of Stem Cell Research, Korea University, Seoul, Korea
| |
Collapse
|