1
|
Usategui A, Municio C, Arias-Salgado EG, Martín M, Fernández-Varas B, Del Rey MJ, Carreira P, González A, Criado G, Perona R, Pablos JL. Evidence of telomere attrition and a potential role for DNA damage in systemic sclerosis. IMMUNITY & AGEING 2022; 19:7. [PMID: 35086525 PMCID: PMC8793167 DOI: 10.1186/s12979-022-00263-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/19/2022] [Indexed: 01/10/2023]
Abstract
Abstract
Background
To investigate the role of cell senescence in systemic sclerosis (SSc), we analyzed telomere shortening (TS) in SSc patients and the effect of targeting DNA damage in the bleomycin model of skin fibrosis.
Results
Telomere length (TL) in blood leukocytes of 174 SSc patients and 68 healthy controls was measured by Southern blot, and we found shorter age-standardized TL in SSc patients compared to healthy controls. TL was shorter in SSc patients with ILD compared to those without ILD and in anti-topoisomerase I positive compared to anti-centromere positive patients. To analyze the potential role of DNA damage in skin fibrosis, we evaluated the effects of the DNA protective GSE4 peptide in the bleomycin mouse model of scleroderma and the fibrotic response of cultured human dermal fibroblasts. Administration of GSE4-nanoparticles attenuated bleomycin-induced skin fibrosis as measured by Masson’s staining of collagen and reduced Acta2 and Ctgf mRNA expression, whereas transduction of dermal fibroblasts with a lentiviral GSE4 expression vector reduced COL1A1, ACTA2 and CTGF gene expression after stimulation with bleomycin or TGF-β, in parallel to a reduction of the phospho-histone H2A.X marker of DNA damage.
Conclusions
SSc is associated with TS, particularly in patients with lung disease or anti-topoisomerase I antibodies. Administration of GSE4 peptide attenuated experimental skin fibrosis and reduced fibroblast expression of profibrotic factors, supporting a role for oxidative DNA damage in scleroderma.
Collapse
|
2
|
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
|
3
|
Piñeiro-Hermida S, Autilio C, Martínez P, Bosch F, Pérez-Gil J, Blasco MA. Telomerase treatment prevents lung profibrotic pathologies associated with physiological aging. J Cell Biol 2021; 219:152010. [PMID: 32777016 PMCID: PMC7659728 DOI: 10.1083/jcb.202002120] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/23/2020] [Accepted: 07/17/2020] [Indexed: 02/08/2023] Open
Abstract
Short/dysfunctional telomeres are at the origin of idiopathic pulmonary fibrosis (IPF) in patients mutant for telomere maintenance genes. However, it remains unknown whether physiological aging leads to short telomeres in the lung, thus leading to IPF with aging. Here, we find that physiological aging in wild-type mice leads to telomere shortening and a reduced proliferative potential of alveolar type II cells and club cells, increased cellular senescence and DNA damage, increased fibroblast activation and collagen deposits, and impaired lung biophysics, suggestive of a fibrosis-like pathology. Treatment of both wild-type and telomerase-deficient mice with telomerase gene therapy prevented the onset of lung profibrotic pathologies. These findings suggest that short telomeres associated with physiological aging are at the origin of IPF and that a potential treatment for IPF based on telomerase activation would be of interest not only for patients with telomerase mutations but also for sporadic cases of IPF associated with physiological aging.
Collapse
Affiliation(s)
- Sergio Piñeiro-Hermida
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, Madrid, Spain
| | - Chiara Autilio
- Department of Biochemistry and Molecular Biology, Research Institute "Hospital 12 de Octubre (imas12)," Complutense University, Madrid, Spain
| | - Paula Martínez
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, Madrid, Spain
| | - Fátima Bosch
- Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jesús Pérez-Gil
- Department of Biochemistry and Molecular Biology, Research Institute "Hospital 12 de Octubre (imas12)," Complutense University, Madrid, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, Madrid, Spain
| |
Collapse
|
4
|
Katzen J, Beers MF. Contributions of alveolar epithelial cell quality control to pulmonary fibrosis. J Clin Invest 2021; 130:5088-5099. [PMID: 32870817 DOI: 10.1172/jci139519] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Epithelial cell dysfunction has emerged as a central component of the pathophysiology of diffuse parenchymal diseases including idiopathic pulmonary fibrosis (IPF). Alveolar type 2 (AT2) cells represent a metabolically active lung cell population important for surfactant biosynthesis and alveolar homeostasis. AT2 cells and other distal lung epithelia, like all eukaryotic cells, contain an elegant quality control network to respond to intrinsic metabolic and biosynthetic challenges imparted by mutant protein conformers, dysfunctional subcellular organelles, and dysregulated telomeres. Failed AT2 quality control components (the ubiquitin-proteasome system, unfolded protein response, macroautophagy, mitophagy, and telomere maintenance) result in diverse cellular endophenotypes and molecular signatures including ER stress, defective autophagy, mitochondrial dysfunction, apoptosis, inflammatory cell recruitment, profibrotic signaling, and altered progenitor function that ultimately converge to drive downstream fibrotic remodeling in the IPF lung. As this complex network becomes increasingly better understood, opportunities will emerge to identify targets and therapeutic strategies for IPF.
Collapse
Affiliation(s)
- Jeremy Katzen
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, and
| | - Michael F Beers
- Pulmonary, Allergy, and Critical Care Division, Department of Medicine, and.,Penn-CHOP Lung Biology Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Lin Y, Xu Z. Fibroblast Senescence in Idiopathic Pulmonary Fibrosis. Front Cell Dev Biol 2020; 8:593283. [PMID: 33324646 PMCID: PMC7723977 DOI: 10.3389/fcell.2020.593283] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable and complex natural phenomenon due to the increase in age. Cellular senescence means a non-proliferative but viable cellular physiological state. It is the basis of aging, and it exists in the body at any time point. Idiopathic pulmonary fibrosis (IPF) is an interstitial fibrous lung disease with unknown etiology, characterized by irreversible destruction of lung structure and function. Aging is one of the most critical risk factors for IPF, and extensive epidemiological data confirms IPF as an aging-related disease. Senescent fibroblasts in IPF show abnormal activation, telomere shortening, metabolic reprogramming, mitochondrial dysfunction, apoptosis resistance, autophagy deficiency, and senescence-associated secretory phenotypes (SASP). These characteristics of senescent fibroblasts establish a close link between cellular senescence and IPF. The treatment of senescence-related molecules and pathways is continually emerging, and using senolytics eliminating senescent fibroblasts is also actively tried as a new therapy for IPF. In this review, we discuss the roles of aging and cellular senescence in IPF. In particular, we summarize the signaling pathways through which senescent fibroblasts influence the occurrence and development of IPF. On this basis, we further talk about the current treatment ideas, hoping this paper can be used as a helpful reference for future researches.
Collapse
Affiliation(s)
- Yifan Lin
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Zhihao Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
6
|
Fu X, Zhang P, Song H, Wu C, Li S, Li S, Yan C. LTBP1 plays a potential bridge between depressive disorder and glioblastoma. J Transl Med 2020; 18:391. [PMID: 33059753 PMCID: PMC7566028 DOI: 10.1186/s12967-020-02509-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most malignant tumor in human brain. Diagnosis and treatment of GBM may lead to psychological disorders such as depressive and anxiety disorders. There was no research focusing on the correlation between depressive/anxiety disorder and the outcome of GBM. Thus, the aim of this study was to investigate the possibility of depressive/anxiety disorder correlated with the outcome of GBM patients, as well as the overlapped mechanism bridge which could link depressive/anxiety disorders and GBM. Methods Patient Health Questionnaire (PHQ-9) and Generalized Anxiety Disorder (GAD-7) were used to investigate the psychological condition of GBM patients in our department. To further explore the potential mechanism, bioinformatic methods were used to screen out genes that could be indicators of outcome in GBM, followed by gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and protein–protein interaction (PPI) analysis. Further, cellular experiments were conducted to evaluate the proliferation, migration capacity of primary GBM cells from the patients. Results It was revealed that patients with higher PHQ-9 and GAD-7 scores had significantly worse prognosis than their lower-scored counterparts. Bioinformatic mining revealed that LTBP1 could be a potential genetic mechanism in both depressive/anxiety disorder and GBM. Primary GBM cells with different expression level of LTBP1 should significantly different proliferation and migration capacity. GO, KEGG analysis confirmed that extracellular matrix (ECM) was the most enriched function of LTBP1. PPI network showed the interaction of proteins altered by LTBP1. Hub genes COL1A2, COL5A1 and COL10A1, as well as mesenchymal marker CD44 and Vimentin were statistically higher expressed in LTBP1 high group; while proneural marker E-cadherin was significantly higher expressed in low LTBP1 group. Conclusion There is closely correlation between depressive/anxiety disorders and GBM. LTBP1 could be a potential bridge linking the two diseases through the regulation of ECM.
Collapse
Affiliation(s)
- Xiaojun Fu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.,Capital Medical University, Beijing, People's Republic of China
| | - Pei Zhang
- Beijing Institute of Technology, Beijing, China
| | - Hongwang Song
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Chenxing Wu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China
| | | | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.
| | - Changxiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.
| |
Collapse
|
7
|
Abstract
The interstitial lung diseases (ILDs) are a group of progressive disorders characterized by chronic inflammation and/or fibrosis in the lung. While some ILDs can be linked to specific environmental causes (i.e., asbestosis, silicosis), in many individuals, no culprit exposure can be identified; these patients are deemed to have "idiopathic interstitial pneumonia" (IIP). Family history is now recognized as the strongest risk factor for IIP, and IIP cases that run in families comprise a syndrome termed "familial interstitial pneumonia" (FIP). Mutations in more than 10 different genes have been implicated as responsible for disease in FIP families. Diverse ILD clinical phenotypes can be seen within a family, and available evidence suggests underlying genetic risk is the primary determinant of disease outcomes. Together, these FIP studies have provided unique insights into the pathobiology of ILDs, and brought focus on the unique issues that arise in the care of patients with FIP.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- U.S. Department of Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
8
|
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: 346] [Impact Index Per Article: 69.2] [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
|
9
|
Nakashima T, Liu T, Hu B, Wu Z, Ullenbruch M, Omori K, Ding L, Hattori N, Phan SH. Role of B7H3/IL-33 Signaling in Pulmonary Fibrosis-induced Profibrogenic Alterations in Bone Marrow. Am J Respir Crit Care Med 2019; 200:1032-1044. [PMID: 31106564 PMCID: PMC6794107 DOI: 10.1164/rccm.201808-1560oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 05/14/2019] [Indexed: 01/12/2023] Open
Abstract
Rationale: The impact of lung insult on the bone marrow (BM) and subsequent disease is unknown.Objectives: To study alterations in the BM in response to lung injury/fibrosis and examine their impact on subsequent lung insult.Methods: BM cells from control or bleomycin-treated donor mice were transplanted into naive mice, which were subsequently evaluated for bleomycin-induced pulmonary fibrosis. In addition, the effect of prior bleomycin treatment on subsequent fibrosis was examined in wild-type and B7H3-knockout mice. Samples from patients with idiopathic pulmonary fibrosis were analyzed for potential clinical relevance of the findings.Measurements and Main Results: Recipient mice transplanted with BM from bleomycin-pretreated donors showed significant exacerbation of subsequent fibrosis with increased B7H3+ cell numbers and a T-helper cell type 2-skewed phenotype. Pretreatment with a minimally fibrogenic/nonfibrogenic dose of bleomycin also caused exacerbation, but not in B7H3-deficient mice. Exacerbation was not observed if the mice received naive BM cell transplant after the initial bleomycin pretreatment. Soluble B7H3 stimulated BM Ly6Chi monocytic cell expansion in vitro and caused similar expansion in the lung in vivo. Notably, soluble B7H3 was elevated in plasma of patients with idiopathic pulmonary fibrosis and in BAL fluid in those with acute exacerbation. Finally, ST2 deficiency diminished the bleomycin-induced B7H3 and IL-13 upregulation, suggesting a role for type 2 innate lymphoid cells.Conclusions: Pulmonary fibrosis caused significant alterations in BM with expansion and activation of monocytic cells, which enhanced fibrosis when transplanted to naive recipients with potential mediation by a novel role for B7H3 in the pathophysiology of pulmonary fibrosis in both mice and humans.
Collapse
Affiliation(s)
- Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Keitaro Omori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Lin Ding
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Sem H. Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
10
|
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
|
11
|
Kinoshita T, Goto T. Molecular Mechanisms of Pulmonary Fibrogenesis and Its Progression to Lung Cancer: A Review. Int J Mol Sci 2019; 20:ijms20061461. [PMID: 30909462 PMCID: PMC6471841 DOI: 10.3390/ijms20061461] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is defined as a specific form of chronic, progressive fibrosing interstitial pneumonia of unknown cause, occurring primarily in older adults, and limited to the lungs. Despite the increasing research interest in the pathogenesis of IPF, unfavorable survival rates remain associated with this condition. Recently, novel therapeutic agents have been shown to control the progression of IPF. However, these drugs do not improve lung function and have not been tested prospectively in patients with IPF and coexisting lung cancer, which is a common comorbidity of IPF. Optimal management of patients with IPF and lung cancer requires understanding of pathogenic mechanisms and molecular pathways that are common to both diseases. This review article reflects the current state of knowledge regarding the pathogenesis of pulmonary fibrosis and summarizes the pathways that are common to IPF and lung cancer by focusing on the molecular mechanisms.
Collapse
Affiliation(s)
- Tomonari Kinoshita
- Division of General Thoracic Surgery, Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 1608582, Japan.
| | - Taichiro Goto
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, Kofu, Yamanashi 4008506, Japan.
| |
Collapse
|
12
|
Winters NI, Burman A, Kropski JA, Blackwell TS. Epithelial Injury and Dysfunction in the Pathogenesis of Idiopathic PulmonaryFibrosis. Am J Med Sci 2019; 357:374-378. [PMID: 31010463 DOI: 10.1016/j.amjms.2019.01.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 12/16/2022]
Abstract
Idiopathic pulmonary fibrosis is a disease of older adults leading to progressive dyspnea and reduced exercise capacity, typically resulting in death within 3-5years of diagnosis. Underlying genetic susceptibility combined with environmental insults is proposed to trigger a chronic wound repair response, leading to activation of the fibrotic cascade. Perturbations in several molecular pathways mediate vulnerability of the alveolar epithelium to injurious agents, including the unfolded protein response, autophagy, mitophagy, and cellular senescence. These cellular responses are intricately intertwined and link genetic susceptibility to the progressive fibrotic phenotype. Ongoing studies investigating these pathways in type II alveolar epithelial cells show promise for identifying new targeted interventions that could prevent or halt the progression of IPF.
Collapse
Affiliation(s)
- Nichelle I Winters
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicineand
| | - Ankita Burman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Jonathan A Kropski
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicineand; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Timothy S Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicineand; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Department of Veterans Affairs Medical Center, Nashville, Tennessee.
| |
Collapse
|
13
|
Jost RT, Dias HB, Krause GC, de Souza RG, de Souza TR, Nuñez NK, Dos Santos FG, Haute GV, da Silva Melo DA, Pitrez PM, da Silva VD, Donadio MVF, de Oliveira JR. Fructose-1,6-Bisphosphate Prevents Bleomycin-Induced Pulmonary Fibrosis in Mice and Inhibits the Proliferation of Lung Fibroblasts. Inflammation 2019; 41:1987-2001. [PMID: 29995294 DOI: 10.1007/s10753-018-0842-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis is a specific form of interstitial pneumonia. In addition to the idiopathic cause, it may be caused by drugs such as bleomycin (BLM)-used in the treatment of tumors. Fructose-1,6-bisphosphate (FBP) is a high-energy endogenous glycolytic compound that has antifibrotic, anti-inflammatory, and immunomodulatory effects. The aim of this study was to investigate the effects of FBP on both BLM-induced pulmonary fibrosis in mice and in a human embryonic lung fibroblast (MRC-5) culture system. C57BL/6 mice were divided into four groups: control, FBP, BLM, and BLM plus FBP. A single dose of bleomycin (7.5 U/kg) was administered intratracheally, and survival, body weight, Ashcroft score, and histological analysis were evaluated. Pulmonary function and bronchoalveolar lavage fluid (BALF) were also evaluated after a single dose of bleomycin (1.2 U/kg-intratracheally). Treatment with FBP (500 mg/kg) was given on day 0 intraperitoneally. Fibroblasts (MRC-5 cells) were used to access the effect of FBP in vitro. In vivo, FBP increased the survival rate and reduced body weight loss (BLM vs. BLM plus FBP-p < 0.05). FBP also prevented BLM-induced loss of pulmonary function and decreased BALF inflammatory cells, level of fibrosis, and superficial collagen density (p < 0.05). In vitro, FBP (0.62 and 1.25 mM) had inhibitory activity on MRC-5 cells and was able to induce senescence in fibroblasts. These results showed that FBP has the potential of reducing the toxic effects of BLM and may provide supportive therapy for conventional methods used for the treatment of cancer.
Collapse
Affiliation(s)
- Renan Trevisan Jost
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil.,Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Henrique Bregolin Dias
- Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Gabriele Catyana Krause
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Rodrigo Godinho de Souza
- Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Tássia Rezende de Souza
- Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Nailê Karine Nuñez
- Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | | | - Gabriela Viegas Haute
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Denizar Alberto da Silva Melo
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Paulo Márcio Pitrez
- Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | | | - Márcio Vinícius Fagundes Donadio
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil.,Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul-PUCRS, Porto Alegre, Brazil.
| |
Collapse
|
14
|
Zheng CM, Zhan X, Yang YH, Jiang T, Ye Q, Lu Y. A Rare Missense Variant in Telomerase Reverse Transcriptase is Associated with Idiopathic Pulmonary Fibrosis in a Chinese Han Family. Chin Med J (Engl) 2018; 131:2205-2209. [PMID: 30203795 PMCID: PMC6144832 DOI: 10.4103/0366-6999.240802] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is an age-related and progressive interstitial lung disease. Up to 20% of cases of IPF cluster in families, genetic factors contribute significantly to the pathogenesis of the disease. This study aimed to explore the association between rare genetic variants and IPF in Chinese Han families. Methods: A Han family, comprising three IPF patients and five unaffected their first-degree relatives, and 100 ethnically matched control individuals from North China were enrolled in this study. Peripheral blood was collected, and genomic DNA was extracted. To elucidate if rare genetic variants are associated with the familial IPF, we performed whole-exome sequencing of affected members from a Chinese Han IPF family. Candidate rare variants were then confirmed by Sanger sequencing. Results: We identified a potentially damaging rare variant-a heterozygous mutation c.2146G>A in exon 6 of the gene encoding for telomerase reverse transcriptase (TERT), which results in an amino acid substitution (p.Ala716Thr). We confirmed the missense mutation by Sanger sequencing in all the affected family members but did not detect this mutation in 100 ethnically matched healthy controls. Patients carried this mutation were characterized by the frequently acute exacerbation of IPF phenotype, with poor prognosis. The mean time to death was 2.8 years after diagnosis. Conclusion: Using next-generation sequencing technology in familial IPF patients, we identified the heterozygous rare variant in TERT gene, and strengthened the importance of genetic variants in telomere-related pathogenesis in Chinese IPF patients.
Collapse
Affiliation(s)
- Chun-Ming Zheng
- Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Xi Zhan
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital; Clinical Center for Interstitial Lung Diseases, Capital Medical University, Beijing 100020, China
| | - Yuan-Hua Yang
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Tao Jiang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Qiao Ye
- Clinical Center for Interstitial Lung Diseases; Department of Occupational Medicine and Toxicology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yong Lu
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
15
|
Zhou Y, Li P, Duan JX, Liu T, Guan XX, Mei WX, Liu YP, Sun GY, Wan L, Zhong WJ, Ouyang DS, Guan CX. Aucubin Alleviates Bleomycin-Induced Pulmonary Fibrosis in a Mouse Model. Inflammation 2018; 40:2062-2073. [PMID: 28785877 DOI: 10.1007/s10753-017-0646-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pulmonary fibrosis is a life-threatening disease characterized by progressive dyspnea and worsening of pulmonary function. No effective therapeutic strategy for pulmonary fibrosis has been established. Aucubin is a natural constituent with a monoterpene cyclic ring system. The potency of aucubin in protecting cellular components against inflammation, oxidative stress, and proliferation effects is well documented. In this study, we investigated the protective effect of aucubin against pulmonary fibrosis in mice. A mouse model of pulmonary fibrosis was established by intratracheal injection of bleomycin (BLM), and aucubin was administered for 21 days after BLM injection. We found that aucubin decreased the breathing frequency and increased the lung dynamic compliance of BLM-stimulated mice detected by Buxco pulmonary function testing system. Histological examination showed that aucubin alleviated BLM-induced lung parenchymal fibrotic changes. Aucubin also reduced the intrapulmonary collagen disposition and inflammatory injury induced by BLM. In addition, aucubin reduced the expression of pro-fibrotic protein transforming growth factor (TGF)-β1 and α-smooth muscle actin (α-SMA) of pulmonary fibrosis mice induced by BLM. Furthermore, the effect of aucubin on the proliferation and differentiation of fibroblast was investigated in vitro. Aucubin inhibited the mRNA and protein expression of Ki67 and proliferating cell nuclear antigen (PCNA) induced by TGF-β1 and reduced the cell proliferation in a murine fibroblast cell NIH3T3. Aucubin also reduced the collagen syntheses and α-SMA expression induced by TGF-β1 in fibroblast. Our results indicate that aucubin inhibits inflammation, fibroblast proliferation, and differentiation, exerting protective effects against BLM-induced pulmonary fibrosis in a mouse model. This study provides an evidence that aucubin may be a novel drug for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Ping Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Jia-Xi Duan
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Tian Liu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Xiu Mei
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Yong-Ping Liu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Guo-Ying Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Li Wan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Dong-Sheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
16
|
Abstract
Genetic investigations of fibrotic diseases, including those of late onset, often yield unanticipated insights into disease pathogenesis. This Review focuses on pathways underlying lung fibrosis that are generalizable to other organs. Herein, we discuss genetic variants subdivided into those that shorten telomeres, activate the DNA damage response, change resident protein expression or function, or affect organelle activity. Genetic studies provide a window into the downstream cascade of maladaptive responses and pathways that lead to tissue fibrosis. In addition, these studies reveal interactions between genetic variants, environmental factors, and age that influence the phenotypic spectrum of disease. The discovery of forces counterbalancing inherited risk alleles identifies potential therapeutic targets, thus providing hope for future prevention or reversal of fibrosis.
Collapse
|
17
|
Rodríguez-Hernández CJ, Mateo-Lozano S, García M, Casalà C, Briansó F, Castrejón N, Rodríguez E, Suñol M, Carcaboso AM, Lavarino C, Mora J, de Torres C. Cinacalcet inhibits neuroblastoma tumor growth and upregulates cancer-testis antigens. Oncotarget 2017; 7:16112-29. [PMID: 26893368 PMCID: PMC4941301 DOI: 10.18632/oncotarget.7448] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/05/2016] [Indexed: 12/14/2022] Open
Abstract
The calcium–sensing receptor is a G protein-coupled receptor that exerts cell-type specific functions in numerous tissues and some cancers. We have previously reported that this receptor exhibits tumor suppressor properties in neuroblastoma. We have now assessed cinacalcet, an allosteric activator of the CaSR approved for clinical use, as targeted therapy for this developmental tumor using neuroblastoma cell lines and patient-derived xenografts (PDX) with different MYCN and TP53 status. In vitro, acute exposure to cinacalcet induced endoplasmic reticulum stress coupled to apoptosis via ATF4-CHOP-TRB3 in CaSR-positive, MYCN-amplified cells. Both phenotypes were partially abrogated by phospholipase C inhibitor U73122. Prolonged in vitro treatment also promoted dose- and time-dependent apoptosis in CaSR-positive, MYCN-amplified cells and, irrespective of MYCN status, differentiation in surviving cells. Cinacalcet significantly inhibited tumor growth in MYCN-amplified xenografts and reduced that of MYCN-non amplified PDX. Morphology assessment showed fibrosis in MYCN-amplified xenografts exposed to the drug. Microarrays analyses revealed up-regulation of cancer-testis antigens (CTAs) in cinacalcet-treated MYCN-amplified tumors. These were predominantly CTAs encoded by genes mapping on chromosome X, which are the most immunogenic. Other modulated genes upon prolonged exposure to cinacalcet were involved in differentiation, cell cycle exit, microenvironment remodeling and calcium signaling pathways. CTAs were up-regulated in PDX and in vitro models as well. Moreover, progressive increase of CaSR expression upon cinacalcet treatment was seen both in vitro and in vivo. In summary, cinacalcet reduces neuroblastoma tumor growth and up-regulates CTAs. This effect represents a therapeutic opportunity and provides surrogate circulating markers of neuroblastoma response to this treatment.
Collapse
Affiliation(s)
- Carlos J Rodríguez-Hernández
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Silvia Mateo-Lozano
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Marta García
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Carla Casalà
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Ferran Briansó
- Statistics and Bioinformatics Unit, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Nerea Castrejón
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Eva Rodríguez
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Mariona Suñol
- Department of Pathology, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Angel M Carcaboso
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Cinzia Lavarino
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.,Department of Oncology, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Jaume Mora
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.,Department of Oncology, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Carmen de Torres
- Developmental Tumor Biology Laboratory, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain.,Department of Oncology, Institut de Recerca Pediàtrica - Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| |
Collapse
|
18
|
Tashiro J, Rubio GA, Limper AH, Williams K, Elliot SJ, Ninou I, Aidinis V, Tzouvelekis A, Glassberg MK. Exploring Animal Models That Resemble Idiopathic Pulmonary Fibrosis. Front Med (Lausanne) 2017; 4:118. [PMID: 28804709 PMCID: PMC5532376 DOI: 10.3389/fmed.2017.00118] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/11/2017] [Indexed: 02/03/2023] Open
Abstract
Large multicenter clinical trials have led to two recently approved drugs for patients with idiopathic pulmonary fibrosis (IPF); yet, both of these therapies only slow disease progression and do not provide a definitive cure. Traditionally, preclinical trials have utilized mouse models of bleomycin (BLM)-induced pulmonary fibrosis—though several limitations prevent direct translation to human IPF. Spontaneous pulmonary fibrosis occurs in other animal species, including dogs, horses, donkeys, and cats. While the fibrotic lungs of these animals share many characteristics with lungs of patients with IPF, current veterinary classifications of fibrotic lung disease are not entirely equivalent. Additional studies that profile these examples of spontaneous fibroses in animals for similarities to human IPF should prove useful for both human and animal investigators. In the meantime, studies of BLM-induced fibrosis in aged male mice remain the most clinically relevant model for preclinical study for human IPF. Addressing issues such as time course of treatment, animal size and characteristics, clinically irrelevant treatment endpoints, and reproducibility of therapeutic outcomes will improve the current status of preclinical studies. Elucidating the mechanisms responsible for the development of fibrosis and disrepair associated with aging through a collaborative approach between researchers will promote the development of models that more accurately represent the realm of interstitial lung diseases in humans.
Collapse
Affiliation(s)
- Jun Tashiro
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gustavo A Rubio
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Andrew H Limper
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Kurt Williams
- Department Pathobiology and Diagnostic Investigations, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Sharon J Elliot
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ioanna Ninou
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Argyrios Tzouvelekis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Marilyn K Glassberg
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
19
|
Telomere Damage Response and Low-Grade Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1024:213-224. [PMID: 28921472 DOI: 10.1007/978-981-10-5987-2_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Telomeres at the ends of chromosomes safeguard genome integrity and stability in human nucleated cells. However, telomere repeats shed off during cell proliferation and other stress responses. Our recent studies show that telomere attrition induces not only epithelial stem cell senescence but also low-grade inflammation in the lungs. The senescence-associated low-grade inflammation (SALI) is characteristic of alveolar stem cell replicative senescence, increased proinflammatory and anti-inflammatory cytokines, infiltrated immune cells, and spillover effects. To date, the mechanisms underlying SALI remain unclear. Investigations demonstrate that senescent epithelial stem cells with telomere erosion are not the source of secreted cytokines, containing no significant increase in expression of the genes coding for increased cytokines, suggesting an alternative senescence-associated secretory phenotype (A-SASP). Given that telomere loss results in significant alterations in the genomes and accumulations of the cleaved telomeric DNA in the cells and milieu externe, we conclude that telomere position effects (TPEs) on gene expression and damage-associated molecular patterns (DAMPs) in antigen presentation are involved in A-SASP and SALI in response to telomere damage in mammals.
Collapse
|
20
|
Abstract
Interstitial lung disease (ILD) comprises a large number of chronic lung disease characterized by varying degrees of inflammation and fibrosis. Mostly they are idiopathic including idiopathic pulmonary fibrosis (IPF), which is a specific disorder characterized by progressive fibrosis leading commonly to end-stage lung disease, respiratory failure, and fatal outcome. IPF and many of these fibrotic ILDs lack effective therapy despite recent approval of two drugs to slow progression in certain IPF patients. Because there are no natural models for IPF, the use of animal models that reproduce key known features of the disease is warranted. Thus, different animal models have been developed to investigate key mechanisms underlying pathogenesis of pulmonary fibrosis and identify potential therapeutic targets for IPF. While no animal model can recapitulate all features of human disease, several are available to address select features of IPF and other fibrotic ILDs. Historically, among the first to be developed and used widely is the bleomycin model, which is the best-characterized and currently most extensively used animal model due to its ability to reproduce many aspects of IPF and other fibrotic ILDs, good reproducibility, and ease of induction. Studies using the bleomycin model have identified many of the cellular and molecular mechanisms now recognized as being important in pathogenesis of IPF and other fibrotic ILDs, as well as novel therapies for these diseases, including two recent drugs approved for treatment of IPF. This chapter will describe commonly used techniques for induction of the model by endotracheal administration of bleomycin through surgical and nonsurgical (transoral instillation).
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | | | - Sem H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
21
|
Ding L, Liu T, Wu Z, Hu B, Nakashima T, Ullenbruch M, Gonzalez De Los Santos F, Phan SH. Bone Marrow CD11c+ Cell-Derived Amphiregulin Promotes Pulmonary Fibrosis. THE JOURNAL OF IMMUNOLOGY 2016; 197:303-12. [PMID: 27206766 DOI: 10.4049/jimmunol.1502479] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/27/2016] [Indexed: 12/30/2022]
Abstract
Amphiregulin (AREG), an epidermal growth factor receptor ligand, is implicated in tissue repair and fibrosis, but its cellular source and role in regeneration versus fibrosis remain unclear. In this study, we hypothesize that AREG induced in bone marrow-derived CD11c(+) cells is essential for pulmonary fibrosis. Thus, the objectives were to evaluate the importance and role of AREG in pulmonary fibrosis, identify the cellular source of AREG induction, and analyze its regulation of fibroblast function and activation. The results showed that lung AREG expression was significantly induced in bleomycin-induced pulmonary fibrosis. AREG deficiency in knockout mice significantly diminished pulmonary fibrosis. Analysis of AREG expression in major lung cell types revealed induction in fibrotic lungs predominantly occurred in CD11c(+) cells. Moreover, depletion of bone marrow-derived CD11c(+) cells suppressed both induction of lung AREG expression and pulmonary fibrosis. Conversely, adoptive transfer of bone marrow-derived CD11c(+) cells from bleomycin-treated donor mice exacerbated pulmonary fibrosis, but not if the donor cells were made AREG deficient prior to transfer. CD11c(+) cell-conditioned media or coculture stimulated fibroblast proliferation, activation, and myofibroblast differentiation in an AREG-dependent manner. Furthermore, recombinant AREG induced telomerase reverse transcriptase, which appeared to be essential for the proliferative effect. Finally, AREG significantly enhanced fibroblast motility, which was associated with increased expression of α6 integrin. These findings suggested that induced AREG specifically in recruited bone marrow-derived CD11c(+) cells promoted bleomycin-induced pulmonary fibrosis by activation of fibroblast telomerase reverse transcriptase-dependent proliferation, motility, and indirectly, myofibroblast differentiation.
Collapse
Affiliation(s)
- Lin Ding
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Taku Nakashima
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | | | - Sem H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
22
|
Affiliation(s)
- Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James E Loyd
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
23
|
Zhang Z, Yu X, Fang X, Liang A, Yu Z, Gu P, Zeng Y, He J, Zhu H, Li S, Fan D, Han F, Zhang L, Yi X. Preventive effects of vitamin D treatment on bleomycin-induced pulmonary fibrosis. Sci Rep 2015; 5:17638. [PMID: 26627341 PMCID: PMC4667265 DOI: 10.1038/srep17638] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 11/03/2015] [Indexed: 01/21/2023] Open
Abstract
Patients with pulmonary fibrosis often have low vitamin D levels, the effects of which are largely unknown. We here report that early vitamin D supplementation significantly reduced the severity of pulmonary fibrosis and inflammatory cell accumulationin in the bleomycin-induced pulmonary fibrosis mouse model on supplementary days 14, 21 and 28 (P < 0.001). Vitamin D supplementation also prevented some ultrastructural changes in response to bleomycin administration, including basement membrane thickening, interstitial fibrin deposition and microvilli flattening or disappearance on days 14, 21 and 28, and lamellar body swelling or vacuolation on days 21 and 28. The bleomycin group had rising hydroxyproline level on days 14, 21 and 28, whereas the vitamin D treatment group showed consistently lower hydroxyproline level but still higher than that of the control group (P < 0.001). Our immunohistochemistry and densitometry analyses showed less staining for α-smooth muscle actin, a myofibroblast marker, in the vitamin D group compared to the bleomycin group (P < 0.001). Thus, vitamin D treatment could prevent bleomycin-induced pulmonary fibrosis by delaying or suppressing ultrastructural changes, as well as attenuating hydroxyproline accumulation and inhibiting myofibroblastic proliferation. These data further our understanding of the roles of vitamin D in pulmonary fibrogenesis and in the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Zongmei Zhang
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Pathology, Tumor Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Yunnan, 650000, China
| | - Xiaoting Yu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xia Fang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zhang Yu
- Electron Microscopy Core Laboratory, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Pan Gu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yu Zeng
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Jian He
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Hailong Zhu
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Shuai Li
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Desheng Fan
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Fei Han
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton at Plainsboro, Plainsboro, NJ, USA
- Department of Chemical Biology, Ernest Mario School of Pharmacy
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School and Piscataway, NJ, USA
- Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Xianghua Yi
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| |
Collapse
|
24
|
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
|
25
|
Chen R, Zhang K, Chen H, Zhao X, Wang J, Li L, Cong Y, Ju Z, Xu D, Williams BRG, Jia J, Liu JP. Telomerase Deficiency Causes Alveolar Stem Cell Senescence-associated Low-grade Inflammation in Lungs. J Biol Chem 2015; 290:30813-29. [PMID: 26518879 DOI: 10.1074/jbc.m115.681619] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Indexed: 12/16/2022] Open
Abstract
Mutations of human telomerase RNA component (TERC) and telomerase reverse transcriptase (TERT) are associated with a subset of lung aging diseases, but the mechanisms by which TERC and TERT participate in lung diseases remain unclear. In this report, we show that knock-out (KO) of the mouse gene Terc or Tert causes pulmonary alveolar stem cell replicative senescence, epithelial impairment, formation of alveolar sacs, and characteristic inflammatory phenotype. Deficiency in TERC or TERT causes a remarkable elevation in various proinflammatory cytokines, including IL-1, IL-6, CXCL15 (human IL-8 homolog), IL-10, TNF-α, and monocyte chemotactic protein 1 (chemokine ligand 2 (CCL2)); decrease in TGF-β1 and TGFβRI receptor in the lungs; and spillover of IL-6 and CXCL15 into the bronchoalveolar lavage fluids. In addition to increased gene expressions of α-smooth muscle actin and collagen 1α1, suggesting myofibroblast differentiation, TERC deficiency also leads to marked cellular infiltrations of a mononuclear cell population positive for the leukocyte common antigen CD45, low-affinity Fc receptor CD16/CD32, and pattern recognition receptor CD11b in the lungs. Our data demonstrate for the first time that telomerase deficiency triggers alveolar stem cell replicative senescence-associated low-grade inflammation, thereby driving pulmonary premature aging, alveolar sac formation, and fibrotic lesion.
Collapse
Affiliation(s)
- Ruping Chen
- From the Department of Microbiology/Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, Shandong Province 250012, China, the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Kexiong Zhang
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Hao Chen
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Xiaoyin Zhao
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Jianqiu Wang
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Li Li
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Yusheng Cong
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Zhenyu Ju
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China
| | - Dakang Xu
- the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China, the Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia, the Department of Molecular and Translational Science, Faculty of Medicine, Monash University, Clayton, Victoria 3168, Australia, and
| | - Bryan R G Williams
- the Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia, the Department of Molecular and Translational Science, Faculty of Medicine, Monash University, Clayton, Victoria 3168, Australia, and
| | - Jihui Jia
- From the Department of Microbiology/Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, Shandong Province 250012, China
| | - Jun-Ping Liu
- From the Department of Microbiology/Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, Shandong Province 250012, China, the Institute of Aging Research, Hangzhou Normal University, School of Medicine, Hangzhou, Zhejiang Province 311121, China, the Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia, the Department of Molecular and Translational Science, Faculty of Medicine, Monash University, Clayton, Victoria 3168, Australia, and the Department of Immunology, Faculty of Medicine, Central Clinical School, Monash University, Prahran, Victoria 3018, Australia
| |
Collapse
|
26
|
Sontake V, Shanmukhappa SK, DiPasquale BA, Reddy GB, Medvedovic M, Hardie WD, White ES, Madala SK. Fibrocytes Regulate Wilms Tumor 1-Positive Cell Accumulation in Severe Fibrotic Lung Disease. THE JOURNAL OF IMMUNOLOGY 2015; 195:3978-91. [PMID: 26371248 DOI: 10.4049/jimmunol.1500963] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/04/2015] [Indexed: 02/06/2023]
Abstract
Collagen-producing myofibroblast transdifferentiation is considered a crucial determinant in the formation of scar tissue in the lungs of patients with idiopathic pulmonary fibrosis. Multiple resident pulmonary cell types and bone marrow-derived fibrocytes have been implicated as contributors to fibrotic lesions because of the transdifferentiation potential of these cells into myofibroblasts. In this study, we assessed the expression of Wilms tumor 1 (WT1), a known marker of mesothelial cells, in various cell types in normal and fibrotic lungs. We demonstrate that WT1 is expressed by both mesothelial and mesenchymal cells in idiopathic pulmonary fibrosis lungs but has limited or no expression in normal human lungs. We also demonstrate that WT1(+) cells accumulate in fibrotic lung lesions, using two different mouse models of pulmonary fibrosis and WT1 promoter-driven fluorescent reporter mice. Reconstitution of bone marrow cells into a TGF-α transgenic mouse model demonstrated that fibrocytes do not transform into WT1(+) mesenchymal cells, but they do augment accumulation of WT1(+) cells in severe fibrotic lung disease. Importantly, the number of WT1(+) cells in fibrotic lesions was correlated with severity of lung disease as assessed by changes in lung function, histology, and hydroxyproline levels in mice. Finally, inhibition of WT1 expression was sufficient to attenuate collagen and other extracellular matrix gene production by mesenchymal cells from both murine and human fibrotic lungs. Thus, the results of this study demonstrate a novel association between fibrocyte-driven WT1(+) cell accumulation and severe fibrotic lung disease.
Collapse
Affiliation(s)
- Vishwaraj Sontake
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; Department of Biochemistry, National Institute of Nutrition, Hyderabad 500007, India
| | - Shiva K Shanmukhappa
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Betsy A DiPasquale
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Geereddy B Reddy
- Department of Biochemistry, National Institute of Nutrition, Hyderabad 500007, India
| | - Mario Medvedovic
- Laboratory for Statistical Genomics and Systems Biology, University of Cincinnati, Cincinnati, OH 45267; and
| | - William D Hardie
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Eric S White
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109
| | - Satish K Madala
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229;
| |
Collapse
|
27
|
Kropski JA, Blackwell TS, Loyd JE. The genetic basis of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45:1717-27. [PMID: 25837031 DOI: 10.1183/09031936.00163814] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 03/17/2015] [Indexed: 02/06/2023]
Abstract
Throughout the past decade, there have been substantial advances in understanding the pathogenesis of idiopathic pulmonary fibrosis (IPF). Recently, several large genome-wide association and linkage studies have identified common genetic variants in more than a dozen loci that appear to contribute to IPF risk. In addition, family-based studies have led to the identification of rare genetic variants in genes related to surfactant function and telomere biology, and mechanistic studies suggest pathophysiological derangements associated with these rare genetic variants are also found in sporadic cases of IPF. Current evidence suggests that rather than existing as distinct syndromes, sporadic and familial cases of IPF (familial interstitial pneumonia) probably reflect a continuum of genetic risk. Rapidly evolving bioinformatic and molecular biology techniques, combined with next-generation sequencing technologies, hold great promise for developing a comprehensive, integrated approach to defining the fundamental molecular mechanisms that underlie IPF pathogenesis.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA Dept of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA Dept of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA Department of Veterans Affairs Medical Center, Nashville, TN, USA
| | - James E Loyd
- Division of Allergy, Pulmonary and Critical Care Medicine, Dept of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
28
|
Abstract
Ageing is the main risk factor for major non-communicable chronic lung diseases, including chronic obstructive pulmonary disease, most forms of lung cancer and idiopathic pulmonary fibrosis. While the prevalence of these diseases continually increases with age, their respective incidence peaks at different times during the lifespan, suggesting specific effects of ageing on the onset and/or pathogenesis of chronic obstructive pulmonary disease, lung cancer and idiopathic pulmonary fibrosis. Recently, the nine hallmarks of ageing have been defined as cell-autonomous and non-autonomous pathways involved in ageing. Here, we review the available evidence for the involvement of each of these hallmarks in the pathogenesis of chronic obstructive pulmonary disease, lung cancer, or idiopathic pulmonary fibrosis. Importantly, we propose an additional hallmark, “dysregulation of the extracellular matrix”, which we argue acts as a crucial modifier of cell-autonomous changes and functions, and as a key feature of the above-mentioned lung diseases.
Collapse
|
29
|
Abstract
The extracellular matrix regulates tissue development and homeostasis, and its dysregulation contributes to neoplastic progression. The extracellular matrix serves not only as the scaffold upon which tissues are organized but provides critical biochemical and biomechanical cues that direct cell growth, survival, migration and differentiation and modulate vascular development and immune function. Thus, while genetic modifications in tumor cells undoubtedly initiate and drive malignancy, cancer progresses within a dynamically evolving extracellular matrix that modulates virtually every behavioral facet of the tumor cells and cancer-associated stromal cells. Hanahan and Weinberg defined the hallmarks of cancer to encompass key biological capabilities that are acquired and essential for the development, growth and dissemination of all human cancers. These capabilities include sustained proliferation, evasion of growth suppression, death resistance, replicative immortality, induced angiogenesis, initiation of invasion, dysregulation of cellular energetics, avoidance of immune destruction and chronic inflammation. Here, we argue that biophysical and biochemical cues from the tumor-associated extracellular matrix influence each of these cancer hallmarks and are therefore critical for malignancy. We suggest that the success of cancer prevention and therapy programs requires an intimate understanding of the reciprocal feedback between the evolving extracellular matrix, the tumor cells and its cancer-associated cellular stroma.
Collapse
Affiliation(s)
- Michael W Pickup
- Department of Surgery, Center for Bioengineering and Tissue Regeneration UCSF, San Francisco, CA, USA
| | - Janna K Mouw
- Department of Surgery, Center for Bioengineering and Tissue Regeneration UCSF, San Francisco, CA, USA
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration UCSF, San Francisco, CA, USA Departments of Anatomy, Bioengineering and Therapeutic Sciences, UCSF, San Francisco, CA, USA Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research UCSF, San Francisco, CA, USA UCSF Helen Diller Comprehensive Cancer Center UCSF, San Francisco, CA, USA
| |
Collapse
|
30
|
Kropski JA, Mitchell DB, Markin C, Polosukhin VV, Choi L, Johnson JE, Lawson WE, Phillips JA, Cogan JD, Blackwell TS, Loyd JE. A novel dyskerin (DKC1) mutation is associated with familial interstitial pneumonia. Chest 2014; 146:e1-e7. [PMID: 24504062 DOI: 10.1378/chest.13-2224] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Short telomeres are frequently identified in patients with idiopathic pulmonary fibrosis (IPF) and its inherited form, familial interstitial pneumonia (FIP). We identified a kindred with FIP with short telomeres who did not carry a mutation in known FIP genes TERT or hTR . We performed targeted sequencing of other telomere-related genes to identify the genetic basis of FIP in this kindred. The proband was a 69 year-old man with dyspnea, restrictive pulmonary function test results, and reticular changes on high-resolution CT scan. An older male sibling had died from IPF. The proband had markedly shortened telomeres in peripheral blood and undetectably short telomeres in alveolar epithelial cells. Polymerase chain reaction-based sequencing of NOP10 , TINF2 , NHP2 , and DKC1 revealed that both affected siblings shared a novel A to G 1213 transition in DKC1 near the hTR binding domain that is predicted to encode a Thr405Ala amino acid substitution. hTR levels were decreased out of proportion to DKC1 expression in the T405A DKC1 proband, suggesting this mutation destabilizes hTR and impairs telomerase function. This DKC1 variant represents the third telomere-related gene identified as a genetic cause of FIP. Further investigation into the mechanism by which dyskerin contributes to the development of lung fibrosis is warranted.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University School of Medicine, Nashville, TN.
| | - Daphne B Mitchell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Cheryl Markin
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Vasiliy V Polosukhin
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Leena Choi
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN
| | - Joyce E Johnson
- Department of Pediatrics, Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - William E Lawson
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University School of Medicine, Nashville, TN; Department of Veterans Affairs Medical Center, Vanderbilt University School of Medicine, Nashville, TN
| | - John A Phillips
- Division of Medical Genetics and Genomic Medicine, Vanderbilt University School of Medicine, Nashville, TN; Department of Pediatrics, Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Joy D Cogan
- Division of Medical Genetics and Genomic Medicine, Vanderbilt University School of Medicine, Nashville, TN
| | - Timothy S Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University School of Medicine, Nashville, TN; Department of Veterans Affairs Medical Center, Vanderbilt University School of Medicine, Nashville, TN; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - James E Loyd
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
31
|
Parra ER, Pincelli MS, Teodoro WR, Velosa APP, Martins V, Rangel MP, Barbas-Filho JV, Capelozzi VL. Modeling pulmonary fibrosis by abnormal expression of telomerase/apoptosis/collagen V in experimental usual interstitial pneumonia. ACTA ACUST UNITED AC 2014; 47:567-75. [PMID: 24919172 PMCID: PMC4123836 DOI: 10.1590/1414-431x20143522] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 03/06/2014] [Indexed: 11/22/2022]
Abstract
Limitations on tissue proliferation capacity determined by telomerase/apoptosis
balance have been implicated in pathogenesis of idiopathic pulmonary fibrosis. In
addition, collagen V shows promise as an inductor of apoptosis. We evaluated the
quantitative relationship between the telomerase/apoptosis index, collagen V
synthesis, and epithelial/fibroblast replication in mice exposed to butylated
hydroxytoluene (BHT) at high oxygen concentration. Two groups of mice were analyzed:
20 mice received BHT, and 10 control mice received corn oil. Telomerase expression,
apoptosis, collagen I, III, and V fibers, and hydroxyproline were evaluated by
immunohistochemistry, in situ detection of apoptosis, electron
microscopy, immunofluorescence, and histomorphometry. Electron microscopy confirmed
the presence of increased alveolar epithelial cells type 1 (AEC1) in apoptosis.
Immunostaining showed increased nuclear expression of telomerase in AEC type 2 (AEC2)
between normal and chronic scarring areas of usual interstitial pneumonia (UIP).
Control lungs and normal areas from UIP lungs showed weak green birefringence of type
I and III collagens in the alveolar wall and type V collagen in the basement membrane
of alveolar capillaries. The increase in collagen V was greater than collagens I and
III in scarring areas of UIP. A significant direct association was found between
collagen V and AEC2 apoptosis. We concluded that telomerase, collagen V fiber
density, and apoptosis evaluation in experimental UIP offers the potential to control
reepithelization of alveolar septa and fibroblast proliferation. Strategies aimed at
preventing high rates of collagen V synthesis, or local responses to high rates of
cell apoptosis, may have a significant impact in pulmonary fibrosis.
Collapse
Affiliation(s)
- E R Parra
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M S Pincelli
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - W R Teodoro
- Disciplina de Reumatologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - A P P Velosa
- Disciplina de Reumatologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - V Martins
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M P Rangel
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - J V Barbas-Filho
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - V L Capelozzi
- Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
32
|
Volckaert T, De Langhe S. Lung epithelial stem cells and their niches: Fgf10 takes center stage. FIBROGENESIS & TISSUE REPAIR 2014; 7:8. [PMID: 24891877 PMCID: PMC4041638 DOI: 10.1186/1755-1536-7-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 04/04/2014] [Indexed: 12/20/2022]
Abstract
Throughout life adult animals crucially depend on stem cell populations to maintain and repair their tissues to ensure life-long organ function. Stem cells are characterized by their capacity to extensively self-renew and give rise to one or more differentiated cell types. These powerful stem cell properties are key to meet the changing demand for tissue replacement during normal lung homeostasis and regeneration after lung injury. Great strides have been made over the last few years to identify and characterize lung epithelial stem cells as well as their lineage relationships. Unfortunately, knowledge on what regulates the behavior and fate specification of lung epithelial stem cells is still limited, but involves communication with their microenvironment or niche, a local tissue environment that hosts and influences the behaviors or characteristics of stem cells and that comprises other cell types and extracellular matrix. As such, an intimate and dynamic epithelial-mesenchymal cross-talk, which is also essential during lung development, is required for normal homeostasis and to mount an appropriate regenerative response after lung injury. Fibroblast growth factor 10 (Fgf10) signaling in particular seems to be a well-conserved signaling pathway governing epithelial-mesenchymal interactions during lung development as well as between different adult lung epithelial stem cells and their niches. On the other hand, disruption of these reciprocal interactions leads to a dysfunctional epithelial stem cell-niche unit, which may culminate in chronic lung diseases such as chronic obstructive pulmonary disease (COPD), chronic asthma and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
- Thomas Volckaert
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA ; The Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, 9052 Ghent, Belgium ; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| | - Stijn De Langhe
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA ; Department of Cellular and Developmental Biology, School of Medicine, University of Colorado Denver, 12605 E 16th Avenue, Aurora CO 80045, USA
| |
Collapse
|
33
|
Abstract
Telomeres are ribonucleoprotein structures capping the end of every linear chromosome. In all vertebrates, they are composed of TTAGGG repeats coated with specific protecting proteins. Telomeres shorten with each mitotic cell division, but telomerase, a reverse transcriptase, elongate telomeres in very specific cells, such as embryonic and adult stem cells. Although telomere sequence is identical in mice and humans and telomeres serve the same role of protecting chromosomes and genetic information from damage and erosion in both species, abnormalities in telomere maintenance and in telomerase function do not coincide in phenotype in humans and mice. The telomeres of most laboratory mice are 5 to 10 times longer than in humans, but their lifespan is 30 times shorter. Complete absence of telomerase has little expression in phenotype over several generations in mice, whereas heterozygosity for telomerase mutations in humans is sufficient to result in organ regeneration defect and cancer development. Patients with telomerase deficiency and very short telomeres may develop aplastic anemia, pulmonary fibrosis, or cirrhosis, whereas telomerase-null murine models display only modest hematopoietic deficiency and develop emphysema when exposed to cigarette smoke. In summary, telomerase deficiency in both humans and mice accelerate telomere shortening, but its consequences in the different organs and in the organism diverge, mainly due to telomere length differences.
Collapse
|
34
|
Liu T, Yu H, Ullenbruch M, Jin H, Ito T, Wu Z, Liu J, Phan SH. The in vivo fibrotic role of FIZZ1 in pulmonary fibrosis. PLoS One 2014; 9:e88362. [PMID: 24516640 PMCID: PMC3916640 DOI: 10.1371/journal.pone.0088362] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 01/10/2014] [Indexed: 01/15/2023] Open
Abstract
FIZZ (found in inflammatory zone) 1, a member of a cysteine-rich secreted protein family, is highly induced in lung allergic inflammation and bleomycin induced lung fibrosis, and primarily expressed by airway and type II alveolar epithelial cells. This novel mediator is known to stimulate α-smooth muscle actin and collagen expression in lung fibroblasts. The objective of this study was to investigate the in vivo effects of FIZZ1 on the development of lung fibrosis by evaluating bleomycin-induced pulmonary fibrosis in FIZZ1 deficient mice. FIZZ1 knockout mice exhibited no detectable abnormality. When these mice were treated with bleomycin they exhibited significantly impaired pulmonary fibrosis relative to wild type mice, along with impaired proinflammatory cytokine/chemokine expression. Deficient lung fibroblast activation was also noted in the FIZZ1 knockout mice. Moreover, recruitment of bone marrow-derived cells to injured lung was deficient in FIZZ1 knockout mice. Interestingly in vitro FIZZ1 was shown to have chemoattractant activity for bone marrow cells, including bone marrow-derived dendritic cells. Finally, overexpression of FIZZ1 exacerbated fibrosis. These findings suggested that FIZZ1 exhibited profibrogenic properties essential for bleomycin induced pulmonary fibrosis, as reflected by its ability to induce myofibroblast differentiation and recruit bone marrow-derived cells.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Hongfeng Yu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Hong Jin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Toshihiro Ito
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Jianhua Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Sem H. Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
35
|
Choi M, Ban T, Rhim T. Therapeutic use of stem cell transplantation for cell replacement or cytoprotective effect of microvesicle released from mesenchymal stem cell. Mol Cells 2014; 37:133-9. [PMID: 24598998 PMCID: PMC3935626 DOI: 10.14348/molcells.2014.2317] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/12/2013] [Accepted: 12/13/2013] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and severe type of idiopathic interstitial pneumonias (IIP), and which is currently no method was developed to restore normal structure and function. There are several reports on therapeutic effects of adult stem cell transplantations in animal models of pulmonary fibrosis. However, little is known about how mesenchymal stem cell (MSC) can repair the IPF. In this study, we try to provide the evidence to show that transplanted mesenchymal stem cells directly replace fibrosis with normal lung cells using IPF model mice. As results, transplanted MSC successfully integrated and differentiated into type II lung cell which express surfactant protein. In the other hand, we examine the therapeutic effects of microvesicle treatment, which were released from mesenchymal stem cells. Though the therapeutic effects of MV treatment is less than that of MSC treatment, MV treatment meaningfully reduced the symptom of IPF, such as collagen deposition and inflammation. These data suggest that stem cell transplantation may be an effective strategy for the treatment of pulmonary fibrosis via replacement and cytoprotective effect of microvesicle released from MSCs.
Collapse
Affiliation(s)
- Moonhwan Choi
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791,
Korea
| | - Taehyun Ban
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791,
Korea
| | - Taiyoun Rhim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 133-791,
Korea
| |
Collapse
|
36
|
Gardet A, Zheng TS, Viney JL. Genetic architecture of human fibrotic diseases: disease risk and disease progression. Front Pharmacol 2013; 4:159. [PMID: 24391588 PMCID: PMC3866586 DOI: 10.3389/fphar.2013.00159] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/03/2013] [Indexed: 12/12/2022] Open
Abstract
Genetic studies of human diseases have identified multiple genetic risk loci for various fibrotic diseases. This has provided insights into the myriad of biological pathways potentially involved in disease pathogenesis. These discoveries suggest that alterations in immune responses, barrier function, metabolism and telomerase activity may be implicated in the genetic risks for fibrotic diseases. In addition to genetic disease-risks, the identification of genetic disease-modifiers associated with disease complications, severity or prognosis provides crucial insights into the biological processes implicated in disease progression. Understanding the biological processes driving disease progression may be critical to delineate more effective strategies for therapeutic interventions. This review provides an overview of current knowledge and gaps regarding genetic disease-risks and genetic disease-modifiers in human fibrotic diseases.
Collapse
|
37
|
Stenmark KR, Nozik-Grayck E, Gerasimovskaya E, Anwar A, Li M, Riddle S, Frid M. The adventitia: Essential role in pulmonary vascular remodeling. Compr Physiol 2013; 1:141-61. [PMID: 23737168 DOI: 10.1002/cphy.c090017] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A rapidly emerging concept is that the vascular adventitia acts as a biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. It is the most complex compartment of the vessel wall and comprises a variety of cells including fibroblasts, immunomodulatory cells, resident progenitor cells, vasa vasorum endothelial cells, and adrenergic nerves. In response to vascular stress or injury, resident adventitial cells are often the first to be activated and reprogrammed to then influence tone and structure of the vessel wall. Experimental data indicate that the adventitial fibroblast, the most abundant cellular constituent of adventitia, is a critical regulator of vascular wall function. In response to vascular stresses such as overdistension, hypoxia, or infection, the adventitial fibroblast is activated and undergoes phenotypic changes that include proliferation, differentiation, and production of extracellular matrix proteins and adhesion molecules, release of reactive oxygen species, chemokines, cytokines, growth factors, and metalloproteinases that, collectively, affect medial smooth muscle cell tone and growth directly and that stimulate recruitment and retention of circulating inflammatory and progenitor cells to the vessel wall. Resident dendritic cells also participate in "sensing" vascular stress and actively communicate with fibroblasts and progenitor cells to simulate repair processes that involve expansion of the vasa vasorum, which acts as a conduit for further delivery of inflammatory/progenitor cells. This review presents the current evidence demonstrating that the adventitia acts as a key regulator of pulmonary vascular wall function and structure from the "outside in."
Collapse
Affiliation(s)
- Kurt R Stenmark
- University of Colorado Denver - Pediatric Critical Care, Aurora, Colorado, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
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
|
39
|
B Moore B, Lawson WE, Oury TD, Sisson TH, Raghavendran K, Hogaboam CM. Animal models of fibrotic lung disease. Am J Respir Cell Mol Biol 2013; 49:167-79. [PMID: 23526222 DOI: 10.1165/rcmb.2013-0094tr] [Citation(s) in RCA: 315] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Interstitial lung fibrosis can develop as a consequence of occupational or medical exposure, as a result of genetic defects, and after trauma or acute lung injury leading to fibroproliferative acute respiratory distress syndrome, or it can develop in an idiopathic manner. The pathogenesis of each form of lung fibrosis remains poorly understood. They each result in a progressive loss of lung function with increasing dyspnea, and most forms ultimately result in mortality. To better understand the pathogenesis of lung fibrotic disorders, multiple animal models have been developed. This review summarizes the common and emerging models of lung fibrosis to highlight their usefulness in understanding the cell-cell and soluble mediator interactions that drive fibrotic responses. Recent advances have allowed for the development of models to study targeted injuries of Type II alveolar epithelial cells, fibroblastic autonomous effects, and targeted genetic defects. Repetitive dosing in some models has more closely mimicked the pathology of human fibrotic lung disease. We also have a much better understanding of the fact that the aged lung has increased susceptibility to fibrosis. Each of the models reviewed in this report offers a powerful tool for studying some aspect of fibrotic lung disease.
Collapse
Affiliation(s)
- Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109-2200, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Thannickal VJ. Mechanistic links between aging and lung fibrosis. Biogerontology 2013; 14:609-15. [PMID: 23929205 DOI: 10.1007/s10522-013-9451-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 08/02/2013] [Indexed: 01/25/2023]
Abstract
Our understanding of the biology of aging has advanced significantly in recent years. This has resulted in the recent formulation of the "hallmarks of aging" that include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease that results from the accumulation of scar tissue in the lungs of affected individuals. IPF is a disease of aging that most commonly affects human subjects older than 60 years of age. While progress has been made in elucidating key pathological processes in IPF, the relationship of these processes to those that occur during aging are not well defined. In this review, we explore existing and emerging paradigms in the pathogenesis of IPF in light of the recently defined hallmarks of aging.
Collapse
Affiliation(s)
- Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, 429 THT, 1900 University Blvd., Birmingham, AL, 35294-0006, USA,
| |
Collapse
|
41
|
Kropski JA, Lawson WE, Young LR, Blackwell TS. Genetic studies provide clues on the pathogenesis of idiopathic pulmonary fibrosis. Dis Model Mech 2013; 6:9-17. [PMID: 23268535 PMCID: PMC3529334 DOI: 10.1242/dmm.010736] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and often fatal lung disease for which there is no known treatment. Although the traditional paradigm of IPF pathogenesis emphasized chronic inflammation as the primary driver of fibrotic remodeling, more recent insights have challenged this view. Linkage analysis and candidate gene approaches have identified four genes that cause the inherited form of IPF, familial interstitial pneumonia (FIP). These four genes encode two surfactant proteins, surfactant protein C (encoded by SFTPC) and surfactant protein A2 (SFTPA2), and two components of the telomerase complex, telomerase reverse transcriptase (TERT) and the RNA component of telomerase (TERC). In this review, we discuss how investigating these mutations, as well as genetic variants identified in other inherited disorders associated with pulmonary fibrosis, are providing new insights into the pathogenesis of common idiopathic interstitial lung diseases, particularly IPF. Studies in this area have highlighted key roles for epithelial cell injury and dysfunction in the development of lung fibrosis. In addition, genetic approaches have uncovered the importance of several processes – including endoplasmic reticulum stress and the unfolded protein response, DNA-damage and -repair pathways, and cellular senescence – that might provide new therapeutic targets in fibrotic lung diseases.
Collapse
Affiliation(s)
- Jonathan A Kropski
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
42
|
Ding L, Dolgachev V, Wu Z, Liu T, Nakashima T, Wu Z, Ullenbruch M, Lukacs NW, Chen Z, Phan SH. Essential role of stem cell factor-c-Kit signalling pathway in bleomycin-induced pulmonary fibrosis. J Pathol 2013; 230:205-14. [PMID: 23401096 DOI: 10.1002/path.4177] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/22/2013] [Accepted: 02/01/2013] [Indexed: 01/13/2023]
Abstract
Stem cell factor (SCF) and its receptor c-Kit have been implicated in tissue remodelling and fibrosis. Alveolar fibroblasts from patients with diffuse interstitial fibrosis secrete more SCF. However, its precise role remains unclear. In this study the potential role of the SCF-c-Kit axis in pulmonary fibrosis was examined. Fibrosis was induced by intratracheal instillation of bleomycin (BLM), which caused increased SCF levels in plasma, bronchoalveolar lavage fluid (BALF) and lung tissue, as well as increased expression by lung fibroblasts. These changes were accompanied by increased numbers of bone marrow-derived c-Kit(+) cells in the lung, with corresponding depletion in bone marrow. Both recombinant SCF and lung extracts from BLM-treated animals induced bone-marrow cell migration, which was blocked by c-Kit inhibitor. The migrated cells promoted myofibroblast differentiation when co-cultured with fibroblasts, suggesting a paracrine pathogenic role. Interestingly, lung fibroblast cultures contained a subpopulation of cells that expressed functionally active c-Kit, which were significantly greater and more responsive to SCF induction when isolated from fibrotic lungs, including those from patients with idiopathic pulmonary fibrosis (IPF). This c-Kit(+) subpopulation was αSMA-negative and expressed lower levels of collagen I but significantly higher levels of TGFβ than c-Kit-negative cells. SCF deficiency achieved by intratracheal treatment with neutralizing anti-SCF antibody or by use of Kitl(Sl)/Kitl(Sl-d) mutant mice in vivo resulted in significant reduction in pulmonary fibrosis. Taken together, the SCF-c-Kit pathway was activated in BLM-injured lung and might play a direct role in pulmonary fibrosis by the recruitment of bone marrow progenitor cells capable of promoting lung myofibroblast differentiation.
Collapse
Affiliation(s)
- Lin Ding
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Degryse AL, Xu XC, Newman JL, Mitchell DB, Tanjore H, Polosukhin VV, Jones BR, McMahon FB, Gleaves LA, Phillips JA, Cogan JD, Blackwell TS, Lawson WE. Telomerase deficiency does not alter bleomycin-induced fibrosis in mice. Exp Lung Res 2012; 38:124-34. [PMID: 22394286 DOI: 10.3109/01902148.2012.658148] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by interstitial lung infiltrates, dyspnea, and progressive respiratory failure. Reports linking telomerase mutations to familial interstitial pneumonia (FIP) suggest that telomerase activity and telomere length maintenance are important in disease pathogenesis. To investigate the role of telomerase in lung fibrotic remodeling, intratracheal bleomycin was administered to mice deficient in telomerase reverse transcriptase (TERT) or telomerase RNA component (TERC) and to wild-type controls. TERT-deficient and TERC-deficient mice were interbred to the F6 and F4 generation, respectively, when they developed skin manifestations and infertility. Fibrosis was scored using a semiquantitative scale and total lung collagen was measured using a hydroxyprolinemicroplate assay. Telomere lengths were measured in peripheral blood leukocytes and isolated type II alveolar epithelial cells (AECs). Telomerase activity in type II AECs was measured using a real-time polymerase chain reaction (PCR)-based system. Following bleomycin, TERT-deficient and TERC-deficient mice developed an equivalent inflammatory response and similar lung fibrosis (by scoring of lung sections and total lung collagen content) compared to controls, a pattern seen in both early (F1) and later (F6 TERT and F4 TERC) generations. Telomere lengths were reduced in peripheral blood leukocytes and isolated type II AECs from F6 TERT-deficient and F4 TERC-deficient mice compared to controls. Telomerase deficiency in a murine model leads to telomere shortening, but does not predispose to enhanced bleomycin-induced lung fibrosis. Additional genetic or environmental factors may be necessary for development of fibrosis in the presence of telomerase deficiency.
Collapse
Affiliation(s)
- Amber L Degryse
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-2650, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Short telomeres result in chromosomal instability in hematopoietic cells and precede malignant evolution in human aplastic anemia. Leukemia 2011; 26:700-7. [PMID: 22005790 DOI: 10.1038/leu.2011.272] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In cell and animal models, telomere erosion promotes chromosomal instability via breakage-fusion-bridge cycles, contributing to the early stages of tumorigenesis. However, evidence involving short telomeres in cancer development in humans is scarce, epidemiological and indirect. Here we directly implicate telomere shortening as a critical molecular event for malignant evolution in aplastic anemia (AA). Patients' telomere lengths at diagnosis of AA, while comparable to age-matched controls, inversely correlated with the probability of developing a cytogenetically abnormal clone. A significantly increased number of telomere signal-free chromosomal ends and chromosomal numerical and structural abnormalities were observed in bone marrow cells of patients with shorter telomeres in comparison with patients with longer telomeres and healthy subjects. The proportion of monosomy-7 cells in the bone marrow at diagnosis of AA inversely correlated with telomere length, years before the emergence of an autonomous and clinically detectable abnormal clone. Marrow cells of clinically healthy individuals carrying loss-of-function telomerase mutations and with extremely short telomeres also showed chromosomal instability in vitro. These results provide the first clinical direct evidence in humans that short telomeres in hematopoietic cells are dysfunctional, mediate chromosomal instability and predispose to malignant transformation in a human disease.
Collapse
|
48
|
C/EBPβ-Thr217 phosphorylation signaling contributes to the development of lung injury and fibrosis in mice. PLoS One 2011; 6:e25497. [PMID: 21998664 PMCID: PMC3187778 DOI: 10.1371/journal.pone.0025497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 09/05/2011] [Indexed: 11/19/2022] Open
Abstract
Background Although C/EBPβko mice are refractory to Bleomycin-induced lung fibrosis the molecular mechanisms remain unknown. Here we show that blocking the ribosomal S-6 kinase (RSK) phosphorylation of the CCAAT/Enhancer Binding Protein (C/EBP)-β on Thr217 (a RSK phosphoacceptor) with either a single point mutation (Ala217), dominant negative transgene or a blocking peptide containing the mutated phosphoacceptor ameliorates the progression of lung injury and fibrosis induced by Bleomycin in mice. Methodology/Principal Findings Mice expressing the non-phosphorylatable C/EBPβ-Ala217 transgene had a marked reduction in lung injury on day-13 after Bleomycin exposure, compared to C/EBPβwt mice, judging by the decrease of CD68+ activated monocytes/macrophages, bone marrow-derived CD45+ cells and lung cytokines as well as by the normal surfactant protein-C expression by lung pneumocytes. On day-21 after Bleomycin treatment, C/EBPβwt mice but not mice expressing the dominant negative C/EBPβ-Ala217 transgene developed severe lung fibrosis as determined by quantitative collagen assays. All mice were of identical genetic background and back-crossed to the parental wild-type inbreed FVB mice for at least ten generations. Treatment of C/EBPβwt mice with a cell permeant, C/EBPβ peptide that inhibits phosphorylation of C/EBPβ on Thr217 (40 µg instilled intracheally on day-2 and day-6 after the single Bleomycin dose) also blocked the progression of lung injury and fibrosis induced by Bleomycin. Phosphorylation of human C/EBPβ on Thr266 (human homologue phosphoacceptor) was induced in collagen-activated human lung fibroblasts in culture as well as in activated lung fibroblasts in situ in lungs of patients with severe lung fibrosis but not in control lungs, suggesting that this signaling pathway may be also relevant in human lung injury and fibrosis. Conclusions/Significance These data suggest that the RSK-C/EBPβ phosphorylation pathway may contribute to the development of lung injury and fibrosis.
Collapse
|
49
|
Liu T, Baek HA, Yu H, Lee HJ, Park BH, Ullenbruch M, Liu J, Nakashima T, Choi YY, Wu GD, Chung MJ, Phan SH. FIZZ2/RELM-β induction and role in pulmonary fibrosis. THE JOURNAL OF IMMUNOLOGY 2011; 187:450-61. [PMID: 21602491 DOI: 10.4049/jimmunol.1000964] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Found in inflammatory zone (FIZZ) 2, also known as resistin-like molecule (RELM)-β, belongs to a novel cysteine-rich secreted protein family named FIZZ/RELM. Its function is unclear, but a closely related family member, FIZZ1, has profibrotic activities. The human ortholog of rodent FIZZ1 has not been identified, but human FIZZ2 has significant sequence homology to both rodent FIZZ2 (59%) and FIZZ1 (50%). Given the greater homology to rodent FIZZ2, analyzing the role of FIZZ2 in a rodent model of bleomycin-induced pulmonary fibrosis would be of greater potential relevance to human fibrotic lung disease. The results showed that FIZZ2 was highly induced in lungs of rodents with bleomycin-induced pulmonary fibrosis and of human patients with idiopathic pulmonary fibrosis. FIZZ2 expression was induced in rodent and human lung epithelial cells by Th2 cytokines, which was mediated via STAT6 signaling. The FIZZ2 induction in murine lungs was found to be essential for pulmonary fibrosis, as FIZZ2 deficiency significantly suppressed pulmonary fibrosis and associated enhanced extracellular matrix and cytokine gene expression. In vitro analysis indicated that FIZZ2 could stimulate type I collagen and α-smooth muscle actin expression in lung fibroblasts. Furthermore, FIZZ2 was shown to have chemoattractant activity for bone marrow (BM) cells, especially BM-derived CD11c(+) dendritic cells. Notably, lung recruitment of BM-derived cells was impaired in FIZZ2 knockout mice. These findings suggest that FIZZ2 is a Th2-associated multifunctional mediator with potentially important roles in the pathogenesis of fibrotic lung diseases.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Eaton KA, Opp JS, Gray BM, Bergin IL, Young VB. Ulcerative typhlocolitis associated with Helicobacter mastomyrinus in telomerase-deficient mice. Vet Pathol 2010; 48:713-25. [PMID: 20926734 DOI: 10.1177/0300985810383876] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Telomerase deficiency induces early senescence and defects in proliferating cell populations, but in mice it has not been associated with inflammatory bowel disease. Genetically engineered mice lacking either telomerase reverse transcriptase (TERT) or telomerase RNA were examined for chronic diarrhea and wasting. Affected mice had pasty stools, thickened nondistensible colon walls, and contracted ceca. Histologically, the cecal mucosa was largely replaced by inflammatory infiltrate consisting of plasma cells, neutrophils, lymphocytes, and macrophages with marked widespread fibrosis and ulceration. Remaining epithelium was disorganized and hyperplastic, with multifocal dysplasia. Colonic mucosa was markedly hyperplastic with similar inflammation and epithelial dysplasia. Multifocal adenomatous hyperplasia, but no inflammation, was present in the small intestine. Microaerophilic spiral bacteria with 16S rRNA gene sequences identical to Helicobacter mastomyrinus were isolated from the colon and cecum. Severe granulomatous typhlocolitis without epithelial dysplasia developed in germ-free recombination-activating gene (RAG) knockout (KO) recipients of CD4+ T cells and inoculated with cecal contents from affected TERT KO mice and in specific pathogen-free recipient RAG KO mice and interleukin-10 KO mice inoculated with H mastomyrinus. Typhlocolitis in mice given H mastomyrinus was more severe than in mice given Helicobacter hepaticus. Telomerase-deficient mice are susceptible to helicobacter-associated typhlocolitis. H mastomyrinus causes severe disease in susceptible mouse strains.
Collapse
Affiliation(s)
- K A Eaton
- University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|