1
|
Gupta S, Fink MK, Kempuraj D, Sinha NR, Martin LM, Keele LM, Sinha PR, Giuliano EA, Hesemann NP, Raikwar SP, Chaurasia SS, Mohan RR. Corneal fibrosis abrogation by a localized AAV-mediated inhibitor of differentiation 3 (Id3) gene therapy in rabbit eyes in vivo. Mol Ther 2022; 30:3257-3269. [PMID: 35780298 PMCID: PMC9552811 DOI: 10.1016/j.ymthe.2022.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/18/2022] [Accepted: 06/29/2022] [Indexed: 11/28/2022] Open
Abstract
Previously we found that inhibitor of differentiation 3 (Id3) gene, a transcriptional repressor, efficiently inhibits corneal keratocyte differentiation to myofibroblasts in vitro. This study evaluated the potential of adeno-associated virus 5 (AAV5)-mediated Id3 gene therapy to treat corneal scarring using an established rabbit in vivo disease model. Corneal scarring/fibrosis in rabbit eyes was induced by alkali trauma, and 24 h thereafter corneas were administered with either balanced salt solution AAV5-naked vector, or AAV5-Id3 vector (n = 6/group) via an optimized reported method. Therapeutic effects of AAV5-Id3 gene therapy on corneal pathology and ocular health were evaluated with clinical, histological, and molecular techniques. Localized AAV5-Id3 gene therapy significantly inhibited corneal fibrosis/haze clinically from 2.7 to 0.7 on the Fantes scale in live animals (AAV5-naked versus AAV5-Id3; p < 0.001). Furthermore, AAV5-Id3 treatment significantly reduced profibrotic gene mRNA levels: α-smooth muscle actin (α-SMA) (2.8-fold; p < 0.001), fibronectin (3.2-fold; p < 0.001), collagen I (0.8-fold; p < 0.001), and collagen III (1.4-fold; p < 0.001), as well as protein levels of α-SMA (23.8%; p < 0.001) and collagens (1.8-fold; p < 0.001). The anti-fibrotic activity of AAV5-Id3 is attributed to reduced myofibroblast formation by disrupting the binding of E-box proteins to the promoter of α-SMA, a transforming growth factor-β signaling downstream target gene. In conclusion, these results indicate that localized AAV5-Id3 delivery in stroma caused no clinically relevant ocular symptoms or corneal cellular toxicity in the rabbit eyes.
Collapse
Affiliation(s)
- Suneel Gupta
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Michael K Fink
- Department of Pathology, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Nishant R Sinha
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Lynn M Martin
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Landon M Keele
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Prashant R Sinha
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Elizabeth A Giuliano
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Nathan P Hesemann
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Department of Pathology, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA
| | - Shyam S Chaurasia
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA; Department of Ophthalmology & Visual Sciences, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rajiv R Mohan
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA; Departments of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65211, USA; Mason Eye Institute, School of Medicine, University of Missouri, 1600 East Rollins Street, Columbia, MO 65212, USA.
| |
Collapse
|
2
|
Bu FT, Jia PC, Zhu Y, Yang YR, Meng HW, Bi YH, Huang C, Li J. Emerging therapeutic potential of adeno-associated virus-mediated gene therapy in liver fibrosis. Mol Ther Methods Clin Dev 2022; 26:191-206. [PMID: 35859692 PMCID: PMC9271983 DOI: 10.1016/j.omtm.2022.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Liver fibrosis is a wound-healing response that results from various chronic damages. If the causes of damage are not removed or effective treatments are not given in a timely manner, it will progress to cirrhosis, even liver cancer. Currently, there are no specific medical therapies for liver fibrosis. Adeno-associated virus (AAV)-mediated gene therapy, one of the frontiers of modern medicine, has gained more attention in many fields due to its high safety profile, low immunogenicity, long-term efficacy in mediating gene expression, and increasingly known tropism. Notably, increasing evidence suggests a promising therapeutic potential for AAV-mediated gene therapy in different liver fibrosis models, which helps to correct abnormally changed target genes in the process of fibrosis and improve liver fibrosis at the molecular level. Moreover, the addition of cell-specific promoters to the genome of recombinant AAV helps to limit gene expression in specific cells, thereby producing better therapeutic efficacy in liver fibrosis. However, animal models are considered to be powerless predictive of tissue tropism, immunogenicity, and genotoxic risks in humans. Thus, AAV-mediated gene therapy will face many challenges. This review systemically summarizes the recent advances of AAV-mediated gene therapy in liver fibrosis, especially focusing on cellular and molecular mechanisms of transferred genes, and presents prospective challenges.
Collapse
Affiliation(s)
- Fang-Tian Bu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, Anhui Province 230032, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Peng-Cheng Jia
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, Anhui Province 230032, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yan Zhu
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ya-Ru Yang
- The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hong-Wu Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, Anhui Province 230032, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yi-Hui Bi
- The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, Anhui Province 230032, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, Anhui Province 230032, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
BMP3 inhibits TGFβ2-mediated myofibroblast differentiation during wound healing of the embryonic cornea. NPJ Regen Med 2022; 7:36. [PMID: 35879352 PMCID: PMC9314337 DOI: 10.1038/s41536-022-00232-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
Often acute damage to the cornea initiates drastic tissue remodeling, resulting in fibrotic scarring that disrupts light transmission and precedes vision impairment. Very little is known about the factors that can mitigate fibrosis and promote scar-free cornea wound healing. We previously described transient myofibroblast differentiation during non-fibrotic repair in an embryonic cornea injury model. Here, we sought to elucidate the mechanistic regulation of myofibroblast differentiation during embryonic cornea wound healing. We found that alpha-smooth muscle actin (αSMA)-positive myofibroblasts are superficial and their presence inversely correlates with wound closure. Expression of TGFβ2 and nuclear localization of pSMAD2 were elevated during myofibroblast induction. BMP3 and BMP7 were localized in the corneal epithelium and corresponded with pSMAD1/5/8 activation and absence of myofibroblasts in the healing stroma. In vitro analyses with corneal fibroblasts revealed that BMP3 inhibits the persistence of TGFβ2-induced myofibroblasts by promoting disassembly of focal adhesions and αSMA fibers. This was confirmed by the expression of vinculin and pFAK. Together, these data highlight a mechanism to inhibit myofibroblast persistence during cornea wound repair.
Collapse
|
4
|
Fukihara J, Maiolo S, Kovac J, Sakamoto K, Wakahara K, Hashimoto N, Reynolds PN. Overexpression of bone morphogenetic protein receptor type 2 suppresses transforming growth factor β-induced profibrotic responses in lung fibroblasts. Exp Lung Res 2022; 48:35-51. [PMID: 35037801 DOI: 10.1080/01902148.2021.2024301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
MATERIALS AND METHODS We investigated BMPR2 expression in pulmonary fibrosis and TGF-β/BMP signaling in lung fibroblasts. Then we evaluated the impact of BMPR2 upregulation using adenoviral transduction on TGF-β-induced Smad2/3 phosphorylation and fibronectin production in lung fibroblasts. RESULTS BMPR2 was distributed in airway epithelium and alveolar walls in rat lungs. BMPR2 expression was decreased in fibrotic lesions in the lungs of rats with bleomycin-induced pulmonary fibrosis and in human lung fibroblasts (HLFs) stimulated with TGF-β. Although Smad2/3 phosphorylation and fibronectin production were not suppressed solely by BMPs, phosphorylated Smad2/3 was decreased in BMPR2-transduced cells even without BMP stimulation. Fibronectin was decreased only when BMPR2-transduced HLFs were stimulated with BMP7 (but not BMP4). Similar results were also observed in IPF patient HLFs and rat lung fibroblasts. CONCLUSIONS BMPR2 expression was reduced in fibrotic lungs and lung fibroblasts stimulated with TGF-β. BMPR2 transduction to lung fibroblasts reduced Smad2/3 phosphorylation, and reduced fibronectin production when treated with BMP7. Upregulation of BMPR2 may be a possible strategy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Jun Fukihara
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Suzanne Maiolo
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Jessica Kovac
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Koji Sakamoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Keiko Wakahara
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Paul N Reynolds
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.,Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
5
|
Li S, Zhang H, Chang J, Li D, Cao P. Iron overload and mitochondrial dysfunction orchestrate pulmonary fibrosis. Eur J Pharmacol 2021; 912:174613. [PMID: 34740581 DOI: 10.1016/j.ejphar.2021.174613] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/06/2021] [Accepted: 09/11/2021] [Indexed: 12/26/2022]
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive heterogeneous disease of lung tissues with poor lung function caused by scar tissue. Due to our limited understanding of its mechanism, there is currently no treatment strategy that can prevent the development of PF. In recent years, iron accumulation and mitochondrial damage have been reported to participate in PF, and drugs that reduce iron content and improve mitochondrial function have shown significant efficacy in animal experimental models. Excessive iron leads to mitochondrial impairment, which may be the key cause that results in the dysfunction of various kinds of pulmonary cells and further promotes PF. As an emerging research hotspot, there are few targeted effective therapeutic strategies at present due to limited mechanistic understanding. In this review, the roles of iron homeostasis imbalance and mitochondrial damage in PF are summarized and discussed, highlighting a promising direction for finding truly effective therapeutics for PF.
Collapse
Affiliation(s)
- Shuxin Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Hongmin Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Jing Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China
| | - Dongming Li
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China.
| | - Pengxiu Cao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, 050024, People's Republic of China.
| |
Collapse
|
6
|
Ollewagen T, Powrie YSL, Myburgh KH, Smith C. Unresolved intramuscular inflammation, not diminished skeletal muscle regenerative capacity, is at the root of rheumatoid cachexia: insights from a rat CIA model. Physiol Rep 2021; 9:e15119. [PMID: 34806343 PMCID: PMC8606867 DOI: 10.14814/phy2.15119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 11/24/2022] Open
Abstract
Rheumatoid arthritis targets numerous organs in patients, including the skeletal muscle, resulting in rheumatoid cachexia. In the muscle niche, satellite cells, macrophages, and myofibroblasts may be affected and the factors they release altered. This study aimed to assess these cell types, cytokines, and growth factors and their relationships to muscle fiber size and number in a rodent collagen-induced arthritis (CIA) model, in order to identify new therapeutic targets. Fiber cross-sectional area (CSA) was 57% lower in CIA than controls (p < 0.0001), thus smaller but more fibers visible per field of view. Immunostaining indicated the increased presence of satellite cells, macrophages, myofibroblasts, and myonuclei per field of view in CIA (p < 0.01), but this finding was not maintained when taking fiber number into consideration. Western blots of gastrocnemius samples indicated that tumor necrosis factor-α was significantly elevated (p < 0.01) while interleukin-10 (IL-10) was decreased (p < 0.05) in CIA. This effect was maintained (and heightened for IL-10) when expressed per fiber number. Myogenic regulatory factors (MyoD and myogenin), transforming growth factor-β and inhibitor of differentiation were significantly elevated in CIA muscle and levels correlated significantly with CSA. Several of these factors remained elevated, but bone morphogenetic protein-7 decreased when considering fiber number per area. In conclusion, CIA-muscle demonstrated a good regenerative response. Myoblast numbers per fiber were not elevated, suggesting their activity results from the persistent inflammatory signaling which also significantly hampered maintenance of muscle fiber size. A clearer picture of signaling events at cellular level in arthritis muscle may be derived from expressing data per fiber.
Collapse
Affiliation(s)
- Tracey Ollewagen
- Department Physiological SciencesScience FacultyStellenbosch UniversityStellenboschSouth Africa
| | - Yigael S. L. Powrie
- Division of Clinical PharmacologyDepartment of MedicineFaculty of Medicine and Health SciencesStellenbosch UniversityStellenboschSouth Africa
| | - Kathryn H. Myburgh
- Department Physiological SciencesScience FacultyStellenbosch UniversityStellenboschSouth Africa
| | - Carine Smith
- Division of Clinical PharmacologyDepartment of MedicineFaculty of Medicine and Health SciencesStellenbosch UniversityStellenboschSouth Africa
| |
Collapse
|
7
|
Shu DY, Lovicu FJ. Insights into Bone Morphogenetic Protein-(BMP-) Signaling in Ocular Lens Biology and Pathology. Cells 2021; 10:cells10102604. [PMID: 34685584 PMCID: PMC8533954 DOI: 10.3390/cells10102604] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/23/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are a diverse class of growth factors that belong to the transforming growth factor-beta (TGFβ) superfamily. Although originally discovered to possess osteogenic properties, BMPs have since been identified as critical regulators of many biological processes, including cell-fate determination, cell proliferation, differentiation and morphogenesis, throughout the body. In the ocular lens, BMPs are important in orchestrating fundamental developmental processes such as induction of lens morphogenesis, and specialized differentiation of its fiber cells. Moreover, BMPs have been reported to facilitate regeneration of the lens, as well as abrogate pathological processes such as TGFβ-induced epithelial-mesenchymal transition (EMT) and apoptosis. In this review, we summarize recent insights in this topic and discuss the complexities of BMP-signaling including the role of individual BMP ligands, receptors, extracellular antagonists and cross-talk between canonical and non-canonical BMP-signaling cascades in the lens. By understanding the molecular mechanisms underlying BMP activity, we can advance their potential therapeutic role in cataract prevention and lens regeneration.
Collapse
Affiliation(s)
- Daisy Y. Shu
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA;
| | - Frank J. Lovicu
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
- Correspondence: ; Tel.: +61-2-9351-5170
| |
Collapse
|
8
|
Castillero E, Howsmon DP, Rego BV, Keeney SJ, Driesbaugh KH, Kawashima T, Xue (薛应騛) Y, Camillo C, George I, Gorman RC, Gorman JH, Sacks MS, Levy RJ, Ferrari G. Altered Responsiveness to TGFβ and BMP and Increased CD45+ Cell Presence in Mitral Valves Are Unique Features of Ischemic Mitral Regurgitation. Arterioscler Thromb Vasc Biol 2021; 41:2049-2062. [PMID: 33827255 DOI: 10.1161/atvbaha.121.316111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Estibaliz Castillero
- Department of Surgery, Columbia University Irving Medical Center, New York, NY (E.C., Y.X., C.C., I.G., G.F.)
| | - Daniel P Howsmon
- Department of Biomedical Engineering, James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin (D.P.H., B.V.R., M.S.S.)
| | - Bruno V Rego
- Department of Biomedical Engineering, James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin (D.P.H., B.V.R., M.S.S.)
| | - Samuel J Keeney
- Department of Pediatrics, Children's Hospital of Philadelphia, PA (S.J.K., K.H.D., R.J.L.)
| | - Kathryn H Driesbaugh
- Department of Pediatrics, Children's Hospital of Philadelphia, PA (S.J.K., K.H.D., R.J.L.)
| | - Takayuki Kawashima
- Gorman Cardiovascular Research Group, Department of Surgery, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.K., R.C.G., J.H.G.)
| | - Yingfei Xue (薛应騛)
- Department of Surgery, Columbia University Irving Medical Center, New York, NY (E.C., Y.X., C.C., I.G., G.F.)
| | - Chiara Camillo
- Department of Surgery, Columbia University Irving Medical Center, New York, NY (E.C., Y.X., C.C., I.G., G.F.)
| | - Isaac George
- Department of Surgery, Columbia University Irving Medical Center, New York, NY (E.C., Y.X., C.C., I.G., G.F.)
| | - Robert C Gorman
- Gorman Cardiovascular Research Group, Department of Surgery, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.K., R.C.G., J.H.G.)
| | - Joseph H Gorman
- Gorman Cardiovascular Research Group, Department of Surgery, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia (T.K., R.C.G., J.H.G.)
| | - Michael S Sacks
- Department of Biomedical Engineering, James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin (D.P.H., B.V.R., M.S.S.)
| | - Robert J Levy
- Department of Pediatrics, Children's Hospital of Philadelphia, PA (S.J.K., K.H.D., R.J.L.)
| | - Giovanni Ferrari
- Department of Surgery, Columbia University Irving Medical Center, New York, NY (E.C., Y.X., C.C., I.G., G.F.)
| |
Collapse
|
9
|
Lucas BR, Voegels RL, do Amaral JB, Bachi ALL, Pezato R. BMP-7, MMP-9, and TGF-β tissue remodeling proteins and their correlations with interleukins 6 and 10 in chronic rhinosinusitis. Eur Arch Otorhinolaryngol 2021; 278:4335-4343. [PMID: 33715043 DOI: 10.1007/s00405-021-06722-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/23/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Chronic rhinosinusitis without nasal polyposis (CRSsNP) and Chronic rhinosinusitis with nasal polyposis (CRSwNP) present distinct tissue remodeling processes. The proteins involved in the process of tissue remodeling have their production and activity related to the inflammatory environment they are. This study aimed to evaluate the protein expression of BMP-7, MMP-9, TGF-β in chronic sinusitis with and without nasal polyposis and their relations with IL-6 and IL-10. METHODS Cross-sectional observational study with 86 participants was divided into three groups: patients with CRSwNP (n = 34), patients with CRSsNP (n = 26), and a control group (CG) without inflammatory disease of the nasal mucosa (n = 26). The primary outcomes were the concentrations of BMP-7, MMP-9, TGF-β, IL-6, and IL-10. Secondary outcomes were the correlations of these markers. RESULTS The TGF-β dosage was elevated in the CRSsNP group and reduced in the CSwNP group. The dosage of IL-6 was higher in the CSwNP group, and the IL-10 dosage lower in the groups with sinusitis, and IL-10 was positively correlated with BMP-7 in all groups. There was a negative correlation between IL-6 and IL-10 in all groups observed. The correlation between MMP-9 and interleukins was lost in the CRSsNP group. There was a positive correlation between TGF-β and IL-6 in the CG, and negative in the CRSsNP group. CONCLUSION An inflammation shown in rhinosinusitis with an increase in IL-6 and decrease in IL-10 when compared with the control group; only TGF-β was altered in the tissue remodeling process when compared with BMP-7 and MMP-9 in rhinosinusitis. There is a loss of correlation between tissue remodeling proteins and interleukins studied in CRSsNP.
Collapse
Affiliation(s)
- Bernardo Relvas Lucas
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | | | - Jônatas Bussador do Amaral
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - André Luis Lacerda Bachi
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
- Post-graduation Program in Health Science, Santo Amaro University (UNISA), São Paulo, SP, Brazil
| | - Rogério Pezato
- ENT Research Lab, Department of Otorhinolaryngology-Head and Neck Surgery, Federal University of São Paulo, São Paulo, SP, Brazil.
- Otorhinolaryngology Department, São Paulo University, São Paulo, SP, Brazil.
| |
Collapse
|
10
|
Hall JA, Ramachandran D, Roh HC, DiSpirito JR, Belchior T, Zushin PJH, Palmer C, Hong S, Mina AI, Liu B, Deng Z, Aryal P, Jacobs C, Tenen D, Brown CW, Charles JF, Shulman GI, Kahn BB, Tsai LTY, Rosen ED, Spiegelman BM, Banks AS. Obesity-Linked PPARγ S273 Phosphorylation Promotes Insulin Resistance through Growth Differentiation Factor 3. Cell Metab 2020; 32:665-675.e6. [PMID: 32941798 PMCID: PMC7543662 DOI: 10.1016/j.cmet.2020.08.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 06/05/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022]
Abstract
The thiazolidinediones (TZDs) are ligands of PPARγ that improve insulin sensitivity, but their use is limited by significant side effects. Recently, we demonstrated a mechanism wherein TZDs improve insulin sensitivity distinct from receptor agonism and adipogenesis: reversal of obesity-linked phosphorylation of PPARγ at serine 273. However, the role of this modification hasn't been tested genetically. Here we demonstrate that mice encoding an allele of PPARγ that cannot be phosphorylated at S273 are protected from insulin resistance, without exhibiting differences in body weight or TZD-associated side effects. Indeed, hyperinsulinemic-euglycemic clamp experiments confirm insulin sensitivity. RNA-seq in these mice reveals reduced expression of Gdf3, a BMP family member. Ectopic expression of Gdf3 is sufficient to induce insulin resistance in lean, healthy mice. We find Gdf3 inhibits BMP signaling and insulin signaling in vitro. Together, these results highlight the diabetogenic role of PPARγ S273 phosphorylation and focus attention on a putative target, Gdf3.
Collapse
Affiliation(s)
- Jessica A Hall
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Deepti Ramachandran
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Hyun C Roh
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | | | - Thiago Belchior
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Peter-James H Zushin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Colin Palmer
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Shangyu Hong
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Amir I Mina
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Bingyang Liu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Zhaoming Deng
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Pratik Aryal
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Danielle Tenen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Chester W Brown
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Memphis, TN 38103, USA
| | - Julia F Charles
- Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Linus T Y Tsai
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA 02215, USA
| | - Alexander S Banks
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
11
|
Enhanced asthma-related fibroblast to myofibroblast transition is the result of profibrotic TGF-β/Smad2/3 pathway intensification and antifibrotic TGF-β/Smad1/5/(8)9 pathway impairment. Sci Rep 2020; 10:16492. [PMID: 33020537 PMCID: PMC7536388 DOI: 10.1038/s41598-020-73473-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Airway remodelling with subepithelial fibrosis, which abolishes the physiological functions of the bronchial wall, is a major issue in bronchial asthma. Human bronchial fibroblasts (HBFs) derived from patients diagnosed with asthma display in vitro predestination towards TGF-β1-induced fibroblast-to-myofibroblast transition (FMT), a key event in subepithelial fibrosis. As commonly used anti-asthmatic drugs do not reverse the structural changes of the airways, and the molecular mechanism of enhanced asthma-related TGF-β1-induced FMT is poorly understood, we investigated the balance between the profibrotic TGF-β/Smad2/3 and the antifibrotic TGF-β/Smad1/5/9 signalling pathways and its role in the myofibroblast formation of HBF populations derived from asthmatic and non-asthmatic donors. Our findings showed for the first time that TGF-β-induced activation of the profibrotic Smad2/3 signalling pathway was enhanced, but the activation of the antifibrotic Smad1/5/(8)9 pathway by TGF-β1 was significantly diminished in fibroblasts from asthmatic donors compared to those from their healthy counterparts. The impairment of the antifibrotic TGF-β/Smad1/5/(8)9 pathway in HBFs derived from asthmatic donors was correlated with enhanced FMT. Furthermore, we showed that Smad1 silencing in HBFs from non-asthmatic donors increased the FMT potential in these cells. Additionally, we demonstrated that activation of antifibrotic Smad signalling via BMP7 or isoliquiritigenin [a small-molecule activator of the TGF-β/Smad1/5/(8)9 pathway] administration prevents FMT in HBFs from asthmatic donors through downregulation of profibrotic genes, e.g., α-SMA and fibronectin. Our data suggest that influencing the balance between the antifibrotic and profibrotic TGF-β/Smad signalling pathways using BMP7-mimetic compounds presents an unprecedented opportunity to inhibit subepithelial fibrosis during airway remodelling in asthma.
Collapse
|
12
|
Nishina H, Katou-Ichikawa C, Kuramochi M, Izawa T, Kuwamura M, Yamate J. Participation of Somatic Stem Cells, Recognized by a Unique A3 Antibody, in Mucosal Epithelial Regeneration in Dextran Sulfate Sodium (DSS)-Induced Rat Colonic Lesions. Toxicol Pathol 2020; 48:560-569. [PMID: 32122285 DOI: 10.1177/0192623320906817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A3, generated as a monoclonal antibody against rat malignant fibrous histiocytoma cells, recognizes somatic stem cells in rats. We analyzed the distribution of A3-positive cells in dextran sulfate sodium (DSS)-induced colonic lesions consisting of regenerating mucosa and fibrosis. Male 6-week-old F344 rats were administered 5% DSS in drinking water for 5 to 7 days, and lesions at recovery stage were also examined. In untreated control adult colons, A3-positive cells are localized around the crypts where stem cell niche is formed. Histopathologically, in colons of DSS-administered rats, mucosal atrophy, inflammatory cell infiltration, and fibrosis were observed in the lamina propria; thereafter, mucosal epithelia were desquamated, and crypts were decreased gradually with decrease in surrounding A3-positive cells. At the early recovery stage, crypts showed regeneration with reappearance of A3-positive cells. Interestingly, A3-positive cells aggregated in desquamated mucosa surface of fibrosis. Aggregated A3-positive cells coexpressed with vimentin, Thy-1, and partly CK19 but did not react simultaneously with α-SMA. Likely, aggregated A3-positive cells may be rescue cells with nature of both mesenchymal and epithelial cells to maintain self-renewal after injury in the colon. A3 antibody would become a useful tool to investigate the participation of stem cells in rat colonic lesions.
Collapse
Affiliation(s)
- Hironobu Nishina
- Laboratory of Veterinary Pathology, Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Chisa Katou-Ichikawa
- Laboratory of Veterinary Pathology, Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Mizuki Kuramochi
- Laboratory of Veterinary Pathology, Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Jyoji Yamate
- Laboratory of Veterinary Pathology, Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| |
Collapse
|
13
|
Yin L, Liu MX, Li W, Wang FY, Tang YH, Huang CX. Over-Expression of Inhibitor of Differentiation 2 Attenuates Post-Infarct Cardiac Fibrosis Through Inhibition of TGF-β1/Smad3/HIF-1α/IL-11 Signaling Pathway. Front Pharmacol 2019; 10:1349. [PMID: 31803053 PMCID: PMC6876274 DOI: 10.3389/fphar.2019.01349] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Cardiac fibrosis after myocardial infarction mainly causes cardiac diastolic and systolic dysfunction, which results in fatal arrhythmias or even sudden death. Id2, a transcriptional repressor, has been shown to play an important role in the development of fibrosis in various organs, but its effects on cardiac fibrosis remain unclear. This study aimed to explore the effects of Id2 on cardiac fibrosis after myocardial infarction and its possible mechanisms. Methods: This study was performed in four experimental groups: control group, treatment group (including TGF-β1, hypoxia or MI), treatment+GFP group and treatment+Id2 group. In vitro anoxic and fibrotic models were established by subjecting CFs or NRVMs to a three-gas incubator or TGF-β1, respectively. An animal myocardial infarction model was established by ligating of the left anterior descending coronary artery followed by directly injecting of Id2 adenovirus into the myocardial infarct’s marginal zone. Results: The results showed that Id2 significantly improved cardiac EF and attenuated cardiac hypertrophy. The mRNA and protein levels of α-SMA, Collagen I, Collagen III, MMP2 and TIMP1 were higher in treatment+Id2 group than those in treatment group as well as in treatment+GFP group both in vivo and in vitro. Immunofluorescence revealed that both α-SMA and vimentin were co-expressed in the treatment group and GFP group, but the co-expression were not detected in the control group and Id2 group. Additionally, our findings illustrated that Id2 had protective effects demonstrated by its ability to inhibit the TGF-β1/Smad3/HIF-1α/IL-11 signaling pathways. Besides, over-expression of Id2 reduced cardiomyocytes apoptosis. Conclusion: In conclusion, this study demonstrated that over-expression of Id2 preserved cardiac function and ameliorated adverse cardiac remodeling, which might be a promising treatment target for cardiac fibrosis and apoptosis.
Collapse
Affiliation(s)
- Lin Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ming-Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Feng-Yuan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yan-Hong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
14
|
Kariyawasam HH, Gane SB. Allergen-induced asthma, chronic rhinosinusitis and transforming growth factor-β superfamily signaling: mechanisms and functional consequences. Expert Rev Clin Immunol 2019; 15:1155-1170. [PMID: 31549888 DOI: 10.1080/1744666x.2020.1672538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: Often co-associated, asthma and chronic rhinosinusitis (CRS) are complex heterogeneous disease syndromes. Severity in both is related to tissue inflammation and abnormal repair (termed remodeling). Understanding signaling factors that can modulate, integrate the activation, and regulation of such key processes together is increasingly important. The transforming growth factor (TGF)-β superfamily of ligands comprise a versatile system of immunomodulatory molecules that are gaining recognition as having an essential function in the immunopathogenesis of asthma. Early data suggest an important role in CRS as well. Abnormal or dysregulated signaling may contribute to disease pathogenesis and severity.Areas covered: The essential biology of this complex family of growth factors in relation to the excess inflammation and remodeling that occurs in allergic asthma and CRS is reviewed. The need to understand the integration of signaling pathways together is highlighted. Studies in human airway tissue are evaluated and only selected key animal models relevant to human disease discussed given the highly context-dependent signaling and function of these ligands.Expert opinion: Abnormal or dysregulated TGF-β superfamily signaling may be central to the excess inflammation and tissue remodeling in asthma, and possibly CRS. Therefore, the TGF-β superfamily signaling pathways represent an emerging and attractive therapeutic target.
Collapse
Affiliation(s)
- Harsha H Kariyawasam
- Department of Adult Specialist Allergy and Clinical Immunology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,Department of Rhinology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,University College London, London, UK
| | - Simon B Gane
- Department of Rhinology, Royal National ENT Hospital, University College London Hospitals NHS Foundation Trust, London, UK.,University College London, London, UK
| |
Collapse
|
15
|
Xiao Y, Jiang X, Peng C, Zhang Y, Xiao Y, Liang D, Shi M, Wang Y, Zhang F, Guo B. BMP-7/Smads-induced inhibitor of differentiation 2 (Id2) upregulation and Id2/Twist interaction was involved in attenuating diabetic renal tubulointerstitial fibrosis. Int J Biochem Cell Biol 2019; 116:105613. [PMID: 31539631 DOI: 10.1016/j.biocel.2019.105613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/13/2019] [Accepted: 09/16/2019] [Indexed: 12/15/2022]
Abstract
Renal tubular epithelial-mesenchymal transition (EMT) is the main pathological change in diabetic renal tubulointerstitial fibrosis. Mounting evidence indicates that the inhibitor of differentiation 2 (Id2) protein acts as a negative regulatory factor in organ fibrosis and can inhibit or reverse the process of fibrosis. However, its specific regulatory mechanism is not clear. Diabetes mellitus (DM) rat models were established by injecting rats with streptozotocin and sacrificing them after 16 weeks. Rat renal tubular epithelial cells (NRK-52E) were cultured with normal and high glucose. Immunohistochemical analysis, double immunofluorescence staining, co-immunoprecipitation, Western blot analysis, and real-time polymerase chain reaction were used to determine the expression of Id2, Twist, Smad1/5/8, E-cadherin, α-smooth muscle actin (α-SMA), and collagen Ⅳ. The results showed that bone morphogenetic protein-7 (BMP-7) upregulated the expression of Id2 against high-glucose-induced EMT and extracellular matrix secretion. Moreover, only the simultaneous knockdown of Smad1, Smad5, and Smad8 downregulated the expression of Id2, which was not altered by the individual knockdown of Smad1, Smad5, and Smad8. Basic helix-loop-helix (bHLH) transcription factors were essential for Id2 to regulate the role of downstream target genes, and Twist was a bHLH transcription factor. Therefore, the expression of Twist was examined in this study. Twist was found to be highly expressed in the kidney of DM rats and renal tubular epithelial cells cultured with high glucose. The overexpression of Id2 did not alter the expression of Twist, but the interaction between Id2 and Twist was enhanced. In conclusion, the data showed the specific mechanism underlying Id2 negative regulation in diabetic renal tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Xiaohan Jiang
- Jiangsu Taizhou People's Hospital, Taizhou, Jiangsu 225300, China.
| | - Can Peng
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Yingying Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Yawen Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Dan Liang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases & State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, China.
| |
Collapse
|
16
|
Mao Q, Chen C, Liang H, Zhong S, Cheng X, Li L. Astragaloside IV inhibits excessive mesangial cell proliferation and renal fibrosis caused by diabetic nephropathy via modulation of the TGF-β1/Smad/miR-192 signaling pathway. Exp Ther Med 2019; 18:3053-3061. [PMID: 31572545 PMCID: PMC6755437 DOI: 10.3892/etm.2019.7887] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/12/2019] [Indexed: 12/11/2022] Open
Abstract
Astragaloside IV (ASI) exhibits a wide variety of pharmacological effects in cardiovascular diseases, hepatitis and kidney disease and due to this, ASI has recently become an attractive research target. The present study aimed to determine the effect of ASI on renal fibrosis and the mechanisms underlying its therapeutic effects in diabetic nephropathy (DN). In vitro, ASI was added to rat mesangial cells (RMCs) and cultured with a high level of glucose (HG) to observe the effects exhibited on proliferation and fibrosis-related mRNA and protein expression. In vivo, a DN model was established using streptozotocin administration in rats, and renal injury was evaluated using renal histological examination. The expression levels of related mRNAs and proteins were analyzed using reverse transcription-quantitative PCR, western blot analysis and immunohistochemistry. ASI was demonstrated to downregulate miR-192 expression and inhibit excessive proliferation of RMCs, which was induced by HG, in a dose-dependent manner. Additionally, ASI exhibited a therapeutic effect on DN rats. ASI was also demonstrated to decrease the miR-192 expression and mRNA and protein expression of transforming growth factor-β1 (TGF-β1), Smad3, α-smooth muscle actin (α-SMA) and collagen type 1 (col1), and increase the mRNA and protein expression of Smad7 in vitro and in vivo. These results suggested that ASI exhibited a therapeutic effect on DN, possibly due to the inhibition of excessive mesangial proliferation and renal fibrosis via the TGF-β1/Smad/miR-192 signaling pathway.
Collapse
Affiliation(s)
- Qian Mao
- Department Endocrinology, Hospital of Beihua University, Jilin, Jilin 132013, P.R. China
| | - Cuicui Chen
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, Guangdong 510663, P.R. China
| | - Huankun Liang
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, Guangdong 510663, P.R. China
| | - Shuhai Zhong
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, Guangdong 510663, P.R. China
| | - Xinbo Cheng
- Department of Endocrinology and Metabolism, Hospital of Soochow University, Suzhou, Jiangsu 210506, P.R. China
| | - Laiqing Li
- Guangzhou Youdi Bio-Technology Co., Ltd., Guangzhou, Guangdong 510663, P.R. China
| |
Collapse
|
17
|
Downregulation of Sostdc1 in Testicular Sertoli Cells is Prerequisite for Onset of Robust Spermatogenesis at Puberty. Sci Rep 2019; 9:11458. [PMID: 31391487 PMCID: PMC6686024 DOI: 10.1038/s41598-019-47930-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 07/16/2019] [Indexed: 01/03/2023] Open
Abstract
An alarming decline in sperm count of men from several countries has become a major concern for the world community. Hormones act on testicular Sertoli cells (Sc) to regulate male fertility by governing the division and differentiation of germ cells (Gc). However, there is a limited knowledge about Sc specific gene(s) regulating the spermatogenic output of the testis. Sclerostin domain-containing 1 protein (Sostdc1) is a dual BMP/Wnt regulator is predominantly expressed in the Sc of infant testes which hardly show any sign of spermatogenesis. In order to investigate the role of Sostdc1 in spermatogenic regulation, we have generated transgenic (Tg) rats which induced persistent expression of Sostdc1 in mature Sc causing reduced sperm counts. Although Sc specific Sostdc1 did not affect the function of either Sc or Leydig cells (Lc) in the adult testis of Tg rat, we observed a selective augmentation of the BMP target genes via activated phospho smad 1/5/8 signaling in Gc leading to apoptosis. Here, for the first time, we have demonstrated that Sostdc1 is a negative regulator of spermatogenesis, and provided substantial evidence that down regulation of Sostdc1 during puberty is critically essential for quantitatively and qualitatively normal spermatogenesis governing male fertility.
Collapse
|
18
|
Knickmeyer MD, Mateo JL, Eckert P, Roussa E, Rahhal B, Zuniga A, Krieglstein K, Wittbrodt J, Heermann S. TGFβ-facilitated optic fissure fusion and the role of bone morphogenetic protein antagonism. Open Biol 2019; 8:rsob.170134. [PMID: 29593116 PMCID: PMC5881030 DOI: 10.1098/rsob.170134] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 03/02/2018] [Indexed: 12/25/2022] Open
Abstract
The optic fissure is a transient gap in the developing vertebrate eye, which must be closed as development proceeds. A persisting optic fissure, coloboma, is a major cause for blindness in children. Although many genes have been linked to coloboma, the process of optic fissure fusion is still little appreciated, especially on a molecular level. We identified a coloboma in mice with a targeted inactivation of transforming growth factor β2 (TGFβ2). Notably, here the optic fissure margins must have touched, however failed to fuse. Transcriptomic analyses indicated an effect on remodelling of the extracellular matrix (ECM) as an underlying mechanism. TGFβ signalling is well known for its effect on ECM remodelling, but it is at the same time often inhibited by bone morphogenetic protein (BMP) signalling. Notably, we also identified two BMP antagonists among the downregulated genes. For further functional analyses we made use of zebrafish, in which we found TGFβ ligands expressed in the developing eye, and the ligand binding receptor in the optic fissure margins where we also found active TGFβ signalling and, notably, also gremlin 2b (grem2b) and follistatin a (fsta), homologues of the regulated BMP antagonists. We hypothesized that TGFβ is locally inducing expression of BMP antagonists within the margins to relieve the inhibition from its regulatory capacity regarding ECM remodelling. We tested our hypothesis and found that induced BMP expression is sufficient to inhibit optic fissure fusion, resulting in coloboma. Our findings can likely be applied also to other fusion processes, especially when TGFβ signalling or BMP antagonism is involved, as in fusion processes during orofacial development.
Collapse
Affiliation(s)
- Max D Knickmeyer
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg D-79104, Germany.,Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, Freiburg D-79104, Germany
| | - Juan L Mateo
- Departamento de Informática, Universidad de Oviedo, Jesús Arias de Velasco, Oviedo 33005, Spain
| | - Priska Eckert
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg D-79104, Germany.,Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, Freiburg D-79104, Germany
| | - Eleni Roussa
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg D-79104, Germany
| | - Belal Rahhal
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg D-79104, Germany
| | - Aimee Zuniga
- Developmental Genetics, University of Basel Medical School, Basel CH-4058, Switzerland
| | - Kerstin Krieglstein
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg D-79104, Germany
| | | | - Stephan Heermann
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg D-79104, Germany
| |
Collapse
|
19
|
miR-4739 mediates pleural fibrosis by targeting bone morphogenetic protein 7. EBioMedicine 2019; 41:670-682. [PMID: 30850350 PMCID: PMC6443597 DOI: 10.1016/j.ebiom.2019.02.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pleural fibrosis is defined as excessive depositions of matrix components that result in pleural tissue architecture destruction and dysfunction. In severe cases, the progression of pleural fibrosis leads to lung entrapment, resulting in dyspnea and respiratory failure. However, the mechanism of pleural fibrosis is poorly understood. METHODS miR-4739 levels were detected by miRNA array and real-time PCR. Real-time PCR, western blotting and immunofluorescence were used to identify the expression profile of indicators related to fibrosis. Target gene of miR-4739 and promoter activity assay was measured by using dual-luciferase reporter assay system. In vivo, pleural fibrosis was evaluated by Masson staining and miR-4739 level was detected by In situ hybridization histochemistry. FINDINGS We found that bleomycin induced up-regulation of miR-4739 in pleural mesothelial cells (PMCs). Over-regulated miR-4739 mediated mesothelial-mesenchymal transition and increased collagen-I synthesis in PMCs. Investigation on the clinical specimens revealed that high levels of miR-4739 and low levels of bone morphogenetic protein 7 (BMP-7) associated with pleural fibrosis in patients. Then we next identified that miR-4739 targeted and down-regulated BMP-7 which further resulted in unbalance between Smad1/5/9 and Smad2/3 signaling. Lastly, in vivo studies revealed that miR-4739 over-expression induced pleural fibrosis, and exogenous BMP-7 prevented pleural fibrosis in mice. INTERPRETATION Our data indicated that miR-4739 targets BMP-7 which mediates pleural fibrosis. The miR-4739/BMP-7 axis is a promising therapeutic target for the disease. FUND: The National Natural Science Foundation of China.
Collapse
|
20
|
Liu Y, Pandey PR, Sharma S, Xing F, Wu K, Chittiboyina A, Wu SY, Tyagi A, Watabe K. ID2 and GJB2 promote early-stage breast cancer progression by regulating cancer stemness. Breast Cancer Res Treat 2019; 175:77-90. [PMID: 30725231 DOI: 10.1007/s10549-018-05126-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/27/2018] [Indexed: 01/23/2023]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) is a non-invasive form of breast cancer which could progress to or recur as invasive breast cancer. The underlying molecular mechanism of DCIS progression is yet poorly understood, and appropriate biomarkers to distinguish benign form of DCIS from potentially invasive tumor are urgently needed. METHODS To identify the key regulators of DCIS progression, we performed gene-expression analysis of syngeneic breast cancer cell lines MCF10A, DCIS.com, and MCF10CA and cross-referenced the targets with patient cohort data. RESULTS We identified ID2 as a critical gene for DCIS initiation and found that ID2 promoted DCIS formation by enhancing cancer stemness of pre-malignant cells. ID2 also plays a pivotal role in survival of the aggressive cancer cells. In addition, we identified INHBA and GJB2 as key regulators for the transition of benign DCIS to aggressive phenotype. These two genes regulate migration, colonization, and stemness of invasive cancer cells. Upregulation of ID2 and GJB2 predicts poor prognosis after breast-conserving surgery. Finally, we found a natural compound Helichrysetin as ID2 inhibitor which suppresses DCIS formation in vitro and in vivo. CONCLUSION Our results indicate that ID2 is a key driver of DCIS formation and therefore is considered to be a potential target for prevention of DCIS, while INHBA and GJB2 play vital roles in progression of DCIS to IDC and they may serve as potential prognosis markers.
Collapse
Affiliation(s)
- Yin Liu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, 27151, USA
| | - Puspa R Pandey
- Department of Medical Microbiology, Immunology & Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA.,Lonza Walkersville, Inc, Walkersville, MD, USA
| | - Sambad Sharma
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, 27151, USA
| | - Fei Xing
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, 27151, USA
| | - Kerui Wu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, 27151, USA
| | | | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, 27151, USA
| | - Abhishek Tyagi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, 27151, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, NC, 27151, USA.
| |
Collapse
|
21
|
Sayadi LR, Obagi Z, Banyard DA, Ziegler ME, Prussak J, Tomlinson L, Evans GRD, Widgerow AD. Platelet-Rich Plasma, Adipose Tissue, and Scar Modulation. Aesthet Surg J 2018; 38:1351-1362. [PMID: 29617719 DOI: 10.1093/asj/sjy083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Level of Evidence: 4.
Collapse
Affiliation(s)
- Lohrasb R Sayadi
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| | - Zaidal Obagi
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| | - Derek A Banyard
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| | - Mary E Ziegler
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| | - Jordyne Prussak
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| | - Luke Tomlinson
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| | - Gregory R D Evans
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| | - Alan D Widgerow
- Department of Plastic Surgery Center for Tissue Engineering, University of California, Irvine, Orange, CA
| |
Collapse
|
22
|
Castellano G, Franzin R, Stasi A, Divella C, Sallustio F, Pontrelli P, Lucarelli G, Battaglia M, Staffieri F, Crovace A, Stallone G, Seelen M, Daha MR, Grandaliano G, Gesualdo L. Complement Activation During Ischemia/Reperfusion Injury Induces Pericyte-to-Myofibroblast Transdifferentiation Regulating Peritubular Capillary Lumen Reduction Through pERK Signaling. Front Immunol 2018; 9:1002. [PMID: 29875766 PMCID: PMC5974049 DOI: 10.3389/fimmu.2018.01002] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/23/2018] [Indexed: 12/21/2022] Open
Abstract
Pericytes are one of the principal sources of scar-forming myofibroblasts in chronic kidneys disease. However, the modulation of pericyte-to-myofibroblast transdifferentiation (PMT) in the early phases of acute kidney injury is poorly understood. Here, we investigated the role of complement in inducing PMT after transplantation. Using a swine model of renal ischemia/reperfusion (I/R) injury, we found the occurrence of PMT after 24 h of I/R injury as demonstrated by reduction of PDGFRβ+/NG2+ cells with increase in myofibroblasts marker αSMA. In addition, PMT was associated with significant reduction in peritubular capillary luminal diameter. Treatment by C1-inhibitor (C1-INH) significantly preserved the phenotype of pericytes maintaining microvascular density and capillary lumen area at tubulointerstitial level. In vitro, C5a transdifferentiated human pericytes in myofibroblasts, with increased αSMA expression in stress fibers, collagen I production, and decreased antifibrotic protein Id2. The C5a-induced PMT was driven by extracellular signal-regulated kinases phosphorylation leading to increase in collagen I release that required both non-canonical and canonical TGFβ pathways. These results showed that pericytes are a pivotal target of complement activation leading to a profibrotic maladaptive cellular response. Our studies suggest that C1-INH may be a potential therapeutic strategy to counteract the development of PMT and capillary lumen reduction in I/R injury.
Collapse
Affiliation(s)
- Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Chiara Divella
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Fabio Sallustio
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy.,Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Michele Battaglia
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Marc Seelen
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Mohamed R Daha
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands.,Department of Nephrology, Leiden University Medical Centre, Leiden, Netherlands
| | - Giuseppe Grandaliano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
23
|
Sun Y, Fu J, Xue X, Yang H, Wu L. BMP7 regulates lung fibroblast proliferation in newborn rats with bronchopulmonary dysplasia. Mol Med Rep 2018; 17:6277-6284. [PMID: 29512787 PMCID: PMC5928605 DOI: 10.3892/mmr.2018.8692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 10/03/2017] [Indexed: 01/04/2023] Open
Abstract
The present study investigated the expression of bone morphogenetic protein (BMP) 7 in a newborn rat model of bronchopulmonary dysplasia (BPD) and the biological effects of BMP7 on newborn rat lung fibroblast (LF) cells. For this purpose, a total of 196 newborn rats were randomly and equally assigned to a model group and a control group. Lung tissue was collected at days 3, 7, 14 and 21 for histological analysis. The location and expression of BMP7 was examined by immunohistochemical staining and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) analysis. A total of 38 full‑term newborn rats on the day of birth were sacrificed and LF cells were isolated and treated with BMP7. The biological effects of BMP7 on LF cells were assessed by cell proliferation and cell cycle analysis. The findings demonstrated that abnormal alveolar development due to BPD was gradually intensified in the model group over time. Immunohistochemical staining revealed that the location of BMP7 in lung tissue was altered. Immunohistochemistry and RT‑qPCR assays demonstrated a gradual decrease in BMP7 expression in the model group induced by hyperoxia. MTT assays demonstrated that BMP7 inhibited LF cells and the inhibitory effect was dose‑dependent and time‑dependent. Flow cytometry revealed that the inhibitory effect of BMP7 in LF cells was causing cell cycle arrest at the G1 phase. The present study demonstrated that BMP7 may serve an important role in alveolar development in a BPD model. BMP7 may be involved in abnormal alveolar development through the regulation of LF proliferation.
Collapse
Affiliation(s)
- Yanli Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Haiping Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Linlin Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
24
|
Tumelty KE, Higginson-Scott N, Fan X, Bajaj P, Knowlton KM, Shamashkin M, Coyle AJ, Lu W, Berasi SP. Identification of direct negative cross-talk between the SLIT2 and bone morphogenetic protein-Gremlin signaling pathways. J Biol Chem 2018; 293:3039-3055. [PMID: 29317497 DOI: 10.1074/jbc.m117.804021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 12/06/2017] [Indexed: 12/28/2022] Open
Abstract
Slit guidance ligand 2 (SLIT2) is a large, secreted protein that binds roundabout (ROBO) receptors on multiple cell types, including neurons and kidney podocytes. SLIT2-ROBO-mediated signaling regulates neuronal migration and ureteric bud (UB) outgrowth during kidney development as well as glomerular filtration in adult kidneys. Additionally, SLIT2 binds Gremlin, an antagonist of bone morphogenetic proteins (BMPs), and BMP-Gremlin signaling also regulates UB formation. However, direct cross-talk between the ROBO2-SLIT2 and BMP-Gremlin signaling pathways has not been established. Here, we report the discovery of negative feedback between the SLIT2 and BMP-Gremlin signaling pathways. We found that the SLIT2-Gremlin interaction inhibited both SLIT2-ROBO2 signaling in neurons and Gremlin antagonism of BMP activity in myoblasts and fibroblasts. Furthermore, BMP2 down-regulated SLIT2 expression and promoter activity through canonical BMP signaling. Gremlin treatment, BMP receptor inhibition, and SMAD family member 4 (SMAD4) knockdown rescued BMP-mediated repression of SLIT2. BMP2 treatment of nephron progenitor cells derived from human embryonic stem cells decreased SLIT2 expression, further suggesting an interaction between the BMP2-Gremlin and SLIT2 pathways in human kidney cells. In conclusion, our study has revealed direct negative cross-talk between two pathways, previously thought to be unassociated, that may regulate both kidney development and adult tissue maintenance.
Collapse
Affiliation(s)
- Kathleen E Tumelty
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Nathan Higginson-Scott
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Xueping Fan
- the Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts 02118, and
| | - Piyush Bajaj
- the Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340
| | - Kelly M Knowlton
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Michael Shamashkin
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Anthony J Coyle
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Weining Lu
- the Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts 02118, and
| | - Stephen P Berasi
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139,
| |
Collapse
|
25
|
Herrera B, Addante A, Sánchez A. BMP Signalling at the Crossroad of Liver Fibrosis and Regeneration. Int J Mol Sci 2017; 19:ijms19010039. [PMID: 29295498 PMCID: PMC5795989 DOI: 10.3390/ijms19010039] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Bone Morphogenetic Proteins (BMPs) belong to the Transforming Growth Factor-β (TGF-β) family. Initially identified due to their ability to induce bone formation, they are now known to have multiple functions in a variety of tissues, being critical not only during development for tissue morphogenesis and organogenesis but also during adult tissue homeostasis. This review focus on the liver as a target tissue for BMPs actions, devoting most efforts to summarize our knowledge on their recently recognized and/or emerging roles on regulation of the liver regenerative response to various insults, either acute or chronic and their effects on development and progression of liver fibrosis in different pathological conditions. In an attempt to provide the basis for guiding research efforts in this field both the more solid and more controversial areas of research were highlighted.
Collapse
Affiliation(s)
- Blanca Herrera
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Annalisa Addante
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - Aránzazu Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid (UCM), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| |
Collapse
|
26
|
Hudnall AM, Arthur JW, Lowery JW. Clinical Relevance and Mechanisms of Antagonism Between the BMP and Activin/TGF-β Signaling Pathways. J Osteopath Med 2017; 116:452-61. [PMID: 27367950 DOI: 10.7556/jaoa.2016.089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The transforming growth factor β (TGF-β) superfamily is a large group of signaling molecules that participate in embryogenesis, organogenesis, and tissue homeostasis. These molecules are present in all animal genomes. Dysfunction in the regulation or activity of this superfamily's components underlies numerous human diseases and developmental defects. There are 2 distinct arms downstream of the TGF-β superfamily ligands-the bone morphogenetic protein (BMP) and activin/TGF-β signaling pathways-and these 2 responses can oppose one another's effects, most notably in disease states. However, studies have commonly focused on a single arm of the TGF-β superfamily, and the antagonism between these pathways is unknown in most physiologic and pathologic contexts. In this review, the authors summarize the clinically relevant scenarios in which the BMP and activin/TGF-β pathways reportedly oppose one another and identify several molecular mechanisms proposed to mediate this interaction. Particular attention is paid to experimental findings that may be informative to human pathology to highlight potential therapeutic approaches for future investigation.
Collapse
|
27
|
Shu DY, Wojciechowski MC, Lovicu FJ. Bone Morphogenetic Protein-7 Suppresses TGFβ2-Induced Epithelial-Mesenchymal Transition in the Lens: Implications for Cataract Prevention. Invest Ophthalmol Vis Sci 2017; 58:781-796. [PMID: 28152139 PMCID: PMC5295783 DOI: 10.1167/iovs.16-20611] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose Epithelial-mesenchymal transition (EMT) of lens epithelial cells (LECs) is a key pathologic mechanism underlying cataract. Two members of the transforming growth factor-β (TGFβ) superfamily, TGFβ and bone morphogenetic protein-7 (BMP-7) have functionally distinct roles in EMT. While TGFβ is a potent inducer of EMT, BMP-7 counteracts the fibrogenic activity of TGFβ. We examine the modulating effect of BMP-7 on TGFβ-induced EMT in LECs. Methods Rat lens epithelial explants were treated exogenously with TGFβ2 alone or in combination with BMP-7 for up to 5 days. Expression levels of E-cadherin, β-catenin, α-smooth muscle actin (α-SMA), and phosphorylated downstream Smads were determined using immunofluorescence and Western blotting. Reverse transcriptase quantitative PCR (RT-qPCR) was used to study gene expression levels of EMT markers and downstream BMP target genes, including the Inhibitors of differentiation (Id). Results Transforming growth factor-β2 induced LECs to transdifferentiate into myofibroblastic cells. Addition of BMP-7 suppressed TGFβ2-induced α-SMA protein levels and mesenchymal gene expression, with retention of E-cadherin and β-catenin expression to the cell membrane. Addition of BMP-7 prevented lens capsular wrinkling and cellular loss associated with TGFβ2-induced EMT over the 5-day treatment period. The inhibitory effect of BMP-7 was accompanied by an early induction of pSmad1/5 and suppression of TGFβ2-induced pSmad2/3. Treatment with TGFβ2 alone suppressed gene expression of Id2/3 and addition of BMP-7 restored Id2/3 expression. Conclusions Exogenous administration of BMP-7 abrogated TGFβ2-induced EMT in rat lens epithelial explants. Understanding the complex interplay between the TGFβ- and BMP-7–associated Smad signaling pathways and their downstream target genes holds therapeutic promise in cataract prevention.
Collapse
Affiliation(s)
- Daisy Y Shu
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, New South Wales, Australia 2Save Sight Institute, University of Sydney, New South Wales, Australia
| | - Magdalena C Wojciechowski
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, New South Wales, Australia
| | - Frank J Lovicu
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, New South Wales, Australia 2Save Sight Institute, University of Sydney, New South Wales, Australia
| |
Collapse
|
28
|
Pickup MW, Owens P, Moses HL. TGF-β, Bone Morphogenetic Protein, and Activin Signaling and the Tumor Microenvironment. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022285. [PMID: 28062564 DOI: 10.1101/cshperspect.a022285] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The cellular and noncellular components surrounding the tumor cells influence many aspects of tumor progression. Transforming growth factor β (TGF-β), bone morphogenetic proteins (BMPs), and activins have been shown to regulate the phenotype and functions of the microenvironment and are attractive targets to attenuate protumorigenic microenvironmental changes. Given the pleiotropic nature of the cytokines involved, a full understanding of their effects on numerous cell types in many contexts is necessary for proper clinical intervention. In this review, we will explore the various effects of TGF-β, BMP, and activin signaling on stromal phenotypes known to associate with cancer progression. We will summarize these findings in the context of their tumor suppressive or promoting effects, as well as the molecular changes that these cytokines induce to influence stromal phenotypes.
Collapse
Affiliation(s)
- Michael W Pickup
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| | - Philip Owens
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| | - Harold L Moses
- Department of Cancer Biology and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Tennessee 37232
| |
Collapse
|
29
|
The stellate cell system (vitamin A-storing cell system). Anat Sci Int 2017; 92:387-455. [PMID: 28299597 DOI: 10.1007/s12565-017-0395-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/15/2017] [Indexed: 01/18/2023]
Abstract
Past, present, and future research into hepatic stellate cells (HSCs, also called vitamin A-storing cells, lipocytes, interstitial cells, fat-storing cells, or Ito cells) are summarized and discussed in this review. Kupffer discovered black-stained cells in the liver using the gold chloride method and named them stellate cells (Sternzellen in German) in 1876. Wake rediscovered the cells in 1971 using the same gold chloride method and various modern histological techniques including electron microscopy. Between their discovery and rediscovery, HSCs disappeared from the research history. Their identification, the establishment of cell isolation and culture methods, and the development of cellular and molecular biological techniques promoted HSC research after their rediscovery. In mammals, HSCs exist in the space between liver parenchymal cells (PCs) or hepatocytes and liver sinusoidal endothelial cells (LSECs) of the hepatic lobule, and store 50-80% of all vitamin A in the body as retinyl ester in lipid droplets in the cytoplasm. SCs also exist in extrahepatic organs such as pancreas, lung, and kidney. Hepatic (HSCs) and extrahepatic stellate cells (EHSCs) form the stellate cell (SC) system or SC family; the main storage site of vitamin A in the body is HSCs in the liver. In pathological conditions such as liver fibrosis, HSCs lose vitamin A, and synthesize a large amount of extracellular matrix (ECM) components including collagen, proteoglycan, glycosaminoglycan, and adhesive glycoproteins. The morphology of these cells also changes from the star-shaped HSCs to that of fibroblasts or myofibroblasts.
Collapse
|
30
|
A Survey of Strategies to Modulate the Bone Morphogenetic Protein Signaling Pathway: Current and Future Perspectives. Stem Cells Int 2016; 2016:7290686. [PMID: 27433166 PMCID: PMC4940573 DOI: 10.1155/2016/7290686] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/24/2016] [Indexed: 12/14/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the TGF-β family of ligands and are unequivocally involved in regulating stem cell behavior. Appropriate regulation of canonical BMP signaling is critical for the development and homeostasis of numerous human organ systems, as aberrations in the BMP pathway or its regulation are increasingly associated with diverse human pathologies. In this review, we provide a wide-perspective on strategies that increase or decrease BMP signaling. We briefly outline the current FDA-approved approaches, highlight emerging next-generation technologies, and postulate prospective avenues for future investigation. We also detail how activating other pathways may indirectly modulate BMP signaling, with a particular emphasis on the relationship between the BMP and Activin/TGF-β pathways.
Collapse
|
31
|
Lim RR, Tan A, Liu YC, Barathi VA, Mohan RR, Mehta JS, Chaurasia SS. ITF2357 transactivates Id3 and regulate TGFβ/BMP7 signaling pathways to attenuate corneal fibrosis. Sci Rep 2016; 6:20841. [PMID: 26865052 PMCID: PMC4750002 DOI: 10.1038/srep20841] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 01/08/2016] [Indexed: 12/18/2022] Open
Abstract
Corneal fibrosis is often seen in patients with ocular trauma and infection that compromises corneal transparency resulting in vision loss. Treatment strategies including NSAIDs, steroids, MMC and corneal transplants have shown tremendous success but with several side effects and cellular toxicity. Histone deacetylase inhibitors (HDACi) have been shown to inhibit corneal fibrosis via TGFβ signaling pathway. In this study, we investigated safety, efficacy and mechanism of action of a HDACi, ITF2357 in TGFβ-stimulated in vitro primary human cornea stromal fibroblasts (pHCSFs) and in vivo in a photorefractive keratectomy-treated rabbit model of corneal fibrosis. We found that in vivo ITF2357 decreased collagen I, collagen IV, fibronectin, integrin αVβ3 expression with a reduction in corneal haze. In addition, ITF2357 reduced myofibroblast formation, suppressed phosphorylation of Smad proteins in TGFβ pathway and inhibited key responsive protein, P4HA1 involved in pro-collagen synthesis. Treatment of pHCSFs with ITF2357 activated BMP7 levels and expressed all the members of inhibitor of differentiation proteins (Id1-Id4), however, it failed to rescue TGFβ-driven transdifferentiation of fibroblasts to myofibroblasts in the presence of siRNA specific to Id3. We conclude that ITF2357 is a potential anti-fibrotic drug that exerts its action via activation of Id3, a downstream target of TGFβ/BMP7 signaling pathways.
Collapse
Affiliation(s)
- Rayne R. Lim
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, MO 65211, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Alison Tan
- Singapore Eye Research Institute, 20 College Road, 169856, Singapore
- Institute of Molecular and Cellular Biology, A*Star, Biopolis, Singapore
| | - Yu-Chi Liu
- Singapore Eye Research Institute, 20 College Road, 169856, Singapore
- Singapore National Eye Centre, 11 Third Hospital Avenue, 168751, Singapore
| | - Veluchamy A. Barathi
- Singapore Eye Research Institute, 20 College Road, 169856, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Graduate Medical School, 8 College Rd, 169857, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Rajiv R. Mohan
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, MO 65211, USA
- Harry S. Truman Memorial Veteran Hospital, Columbia, MO 65201, USA
- Mason Eye Institute, University of Missouri, Columbia, MO 65211, USA
| | - Jodhbir S. Mehta
- Singapore Eye Research Institute, 20 College Road, 169856, Singapore
- Institute of Molecular and Cellular Biology, A*Star, Biopolis, Singapore
- Singapore National Eye Centre, 11 Third Hospital Avenue, 168751, Singapore
| | - Shyam S. Chaurasia
- Ocular Immunology and Angiogenesis Lab, Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, MO 65211, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA
- Singapore Eye Research Institute, 20 College Road, 169856, Singapore
- Institute of Molecular and Cellular Biology, A*Star, Biopolis, Singapore
| |
Collapse
|
32
|
Characterization of Inhibitor of differentiation (Id) proteins in human cornea. Exp Eye Res 2015; 146:145-153. [PMID: 26712606 DOI: 10.1016/j.exer.2015.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 11/22/2022]
Abstract
Inhibitor of differentiation (Id) proteins are DNA-binding transcription factors involved in cellular proliferation, migration, inflammation, angiogenesis and fibrosis. However, their expression and role in the cornea is unknown. The present study was undertaken to characterize the expression of Id proteins and their interactions with the pro-fibrotic cytokine Transforming Growth Factor β1 (TGFβ1) and anti-fibrotic cytokine, bone morphogenic protein 7 (BMP7) in human cornea. Human donor corneas procured from Eye Bank were used. Id proteins were localized in human corneal sections using immunofluorescence. Primary cultures of human corneal fibroblasts (HCF) were established and treated with either TGFβ1 (5 ng/ml) or BMP7 (10 ng/ml) for 24 h in serum free medium. Expression of Id's in response to TGFβ1, BMP7 and TGFβ1 + BMP7 was analyzed by quantitative real time PCR (qRT-PCR) and western blot analysis. Id1 and Id2 proteins were ubiquitously expressed in the epithelial cells and stromal keratocytes in human cornea. The Id1 was localized to the basal epithelial cells as seen by immunohistochemistry. HCF expressed all known mammalian Id genes (Id1-Id4). In addition, Id1 and Id2 are selectively expressed in HCF. Treatment of human recombinant TGFβ1 (5 ng/ml) to serum-starved HCF showed a significant increase in Id genes (Id1, Id2 and Id4) at 2 h time point compared to BMP7 treatment, which showed time dependent increase in the expression of Id1-Id3 at 24-48 h. Combined treatment with TGFβ1 + BMP7 to HCF showed a significant increase in Id1 transcript and an increasing trend in Id3 and Id4 expression. The results of this study suggest that Id family of genes (Id1-Id4) are localized in the human cornea and expressed in the corneal fibroblasts. Also, Id's were differentially regulated with TGFβ1 and/or BMP7 in a time dependent manner and might serve as a therapeutic target in corneal fibrosis.
Collapse
|
33
|
Expression of CD24 in Human Bone Marrow-Derived Mesenchymal Stromal Cells Is Regulated by TGFβ3 and Induces a Myofibroblast-Like Genotype. Stem Cells Int 2015; 2016:1319578. [PMID: 26788063 PMCID: PMC4691640 DOI: 10.1155/2016/1319578] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/11/2015] [Accepted: 08/12/2015] [Indexed: 12/17/2022] Open
Abstract
Human bone marrow-derived stromal cells (hBMSCs) derived from the adult organism hold great promise for diverse settings in regenerative medicine. Therefore a more complete understanding of hBMSC biology to fully exploit the cells' potential for clinical settings is important. The protein CD24 has been reported to be involved in a diverse range of processes such as cancer, adaptive immunity, inflammation, and autoimmune diseases in other cell types. Its expression in hBMSCs, which has not yet been analyzed, may add an important aspect in the understanding of hBMSC biology. The present study therefore analyzes the expression, regulation, and functional implication of the surface protein CD24 in hBMSCs. Methods used are stimulation studies with TGF beta as well as shRNA-mediated knockdown and overexpression of CD24 followed by microarray, immunocytochemistry, and flow cytometric analyses. To our knowledge, we demonstrate for the first time that the expression of CD24 is an inherent property of hBMSCs. Importantly, the data links the upregulation of CD24 to the adoption of a myofibroblast-like gene expression pattern in hBMSCs. We demonstrate that CD24 is an important modulator in transforming growth factor beta 3 (TGFβ3) signaling with a reciprocal regulatory relationship between these two proteins.
Collapse
|
34
|
Cuna A, Halloran B, Faye-Petersen O, Kelly D, Crossman DK, Cui X, Pandit K, Kaminski N, Bhattacharya S, Ahmad A, Mariani TJ, Ambalavanan N. Alterations in gene expression and DNA methylation during murine and human lung alveolar septation. Am J Respir Cell Mol Biol 2015; 53:60-73. [PMID: 25387348 DOI: 10.1165/rcmb.2014-0160oc] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
DNA methylation, a major epigenetic mechanism, may regulate coordinated expression of multiple genes at specific time points during alveolar septation in lung development. The objective of this study was to identify genes regulated by methylation during normal septation in mice and during disordered septation in bronchopulmonary dysplasia. In mice, newborn lungs (preseptation) and adult lungs (postseptation) were evaluated by microarray analysis of gene expression and immunoprecipitation of methylated DNA followed by sequencing (MeDIP-Seq). In humans, microarray gene expression data were integrated with genome-wide DNA methylation data from bronchopulmonary dysplasia versus preterm and term lung. Genes with reciprocal changes in expression and methylation, suggesting regulation by DNA methylation, were identified. In mice, 95 genes with inverse correlation between expression and methylation during normal septation were identified. In addition to genes known to be important in lung development (Wnt signaling, Angpt2, Sox9, etc.) and its extracellular matrix (Tnc, Eln, etc.), genes involved with immune and antioxidant defense (Stat4, Sod3, Prdx6, etc.) were also observed. In humans, 23 genes were differentially methylated with reciprocal changes in expression in bronchopulmonary dysplasia compared with preterm or term lung. Genes of interest included those involved with detoxifying enzymes (Gstm3) and transforming growth factor-β signaling (bone morphogenetic protein 7 [Bmp7]). In terms of overlap, 20 genes and three pathways methylated during mouse lung development also demonstrated changes in methylation between preterm and term human lung. Changes in methylation correspond to altered expression of a number of genes associated with lung development, suggesting that DNA methylation of these genes may regulate normal and abnormal alveolar septation.
Collapse
Affiliation(s)
- Alain Cuna
- 1 University of Missouri-Kansas City, Kansas City, Missouri
| | - Brian Halloran
- 2 University of Alabama at Birmingham, Birmingham, Alabama
| | | | - David Kelly
- 2 University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Xiangqin Cui
- 2 University of Alabama at Birmingham, Birmingham, Alabama
| | - Kusum Pandit
- 3 University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | | | - Ausaf Ahmad
- 5 University of Rochester Medical Center, Rochester, New York
| | | | | |
Collapse
|
35
|
Verhamme FM, Bracke KR, Joos GF, Brusselle GG. Transforming growth factor-β superfamily in obstructive lung diseases. more suspects than TGF-β alone. Am J Respir Cell Mol Biol 2015; 52:653-62. [PMID: 25396302 DOI: 10.1165/rcmb.2014-0282rt] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Asthma and chronic obstructive pulmonary disease are respiratory disorders and a major global health problem with increasing incidence and severity. Genes originally associated with lung development could be relevant in the pathogenesis of chronic obstructive pulmonary disease/asthma, owing to either an early-life origin of adult complex diseases or their dysregulation in adulthood upon exposure to environmental stressors (e.g., smoking). The transforming growth factor (TGF)-β superfamily is conserved through evolution and is involved in a range of biological processes, both during development and in adult tissue homeostasis. TGF-β1 has emerged as an important regulator of lung and immune system development. However, considerable evidence has been presented for a role of many of the other ligands of the TGF-β superfamily in lung pathology, including activins, bone morphogenetic proteins, and growth differentiation factors. In this review, we summarize the current knowledge on the mechanisms by which activin, bone morphogenetic protein, and growth differentiation factor signaling contribute to the pathogenesis of obstructive airway diseases.
Collapse
Affiliation(s)
- Fien M Verhamme
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | | | | |
Collapse
|
36
|
Calle EA, Mendez JJ, Ghaedi M, Leiby KL, Bove PF, Herzog EL, Sundaram S, Niklason LE. Fate of distal lung epithelium cultured in a decellularized lung extracellular matrix. Tissue Eng Part A 2015; 21:1916-28. [PMID: 25789725 DOI: 10.1089/ten.tea.2014.0511] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Type II cells are the defenders of the alveolus. They produce surfactant to prevent alveolar collapse, they actively transport water to prevent filling of the air sacs that would otherwise prevent gas exchange, and they differentiate to type I epithelial cells. They are an indispensable component of functional lung tissue. To understand the functionality of type II cells in isolation, we sought to track their fate in decellularized matrices and to assess their ability to contribute to barrier function by differentiation to type I alveolar epithelial cells. Rat type II cells were isolated from neonatal rat lungs by labeling with the RTII-70 surface marker and separation using a magnetic column. This produced a population of ∼50% RTII-70-positive cells accompanied by few type I epithelial cells or α-actin-positive mesenchymal cells. This population was seeded into decellularized rat lung matrices and cultured for 1 or 7 days. Culture in Dulbecco's modified Eagle's medium +10% fetal bovine serum (FBS) resulted in reduced expression of epithelial markers and increased expression of mesenchymal markers. By 7 days, no epithelial markers were visible by immunostaining; nearly all cells were α-actin positive. Gene expression for the mesenchymal markers, α-actin, vimentin, and TGF-βR, was significantly upregulated on day 1 (p=0.0005, 0.0005, and 2.342E-5, respectively). Transcript levels of α-actin and TGF-βR remained high at 7 days (p=1.364E-10 and 0.0002). Interestingly, human type II cells cultured under the same conditions showed a similar trend in the loss of epithelial markers, but did not display high expression of mesenchymal markers. Rat cells additionally showed the ability to produce and degrade the basement membrane and extracellular matrix components, such as fibronectin, collagen IV, and collagen I. Quantitative real-time reverse transcription polymerase chain reaction (RT-PCR) showed significant increases in expression of the fibronectin and matrix metalloprotease-2 (MMP-2) genes after 1 day in culture (p=0.0135 and 0.0128, respectively) and elevated collagen I expression at 7 days (p=0.0016). These data suggest that the original type II-enriched population underwent a transition to increased expression of mesenchymal markers, perhaps as part of a survival or wound-healing program. These results suggest that additional medium components and/or the application of physiologically appropriate stimuli such as ventilation may be required to promote lung-specific epithelial phenotypes.
Collapse
Affiliation(s)
- Elizabeth A Calle
- 1Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Julio J Mendez
- 2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| | - Mahboobe Ghaedi
- 2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| | - Katherine L Leiby
- 1Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Peter F Bove
- 3Cystic Fibrosis/Pulmonary Research Treatment Center, University of North Carolina, Chapel Hill, North Carolina
| | - Erica L Herzog
- 4Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sumati Sundaram
- 2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| | - Laura E Niklason
- 1Department of Biomedical Engineering, Yale University, New Haven, Connecticut.,2Department of Anesthesia, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
37
|
De Langhe E, Cailotto F, De Vooght V, Aznar-Lopez C, Vanoirbeek JA, Luyten FP, Lories RJU. Enhanced endogenous bone morphogenetic protein signaling protects against bleomycin induced pulmonary fibrosis. Respir Res 2015; 16:38. [PMID: 25849157 PMCID: PMC4364322 DOI: 10.1186/s12931-015-0202-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 03/04/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Effective treatments for fibrotic diseases such as idiopathic pulmonary fibrosis are largely lacking. Transforming growth factor beta (TGFβ) plays a central role in the pathophysiology of fibrosis. We hypothesized that bone morphogenetic proteins (BMP), another family within the TGFβ superfamily of growth factors, modulate fibrogenesis driven by TGFβ. We therefore studied the role of endogenous BMP signaling in bleomycin induced lung fibrosis. METHODS Lung fibrosis was induced in wild-type or noggin haploinsufficient (Nog +/LacZ ) mice by intratracheal instillation of bleomycin, or phosphate buffered saline as a control. Invasive pulmonary function tests were performed using the flexiVent® SCIREQ system. The mice were sacrificed and lung tissue was collected for analysis using histopathology, collagen quantification, immunohistochemistry and gene expression analysis. RESULTS Nog +/LacZ mice are a known model of increased BMP signaling and were partially protected from bleomycin-induced lung fibrosis with reduced Ashcroft score, reduced collagen content and preservation of pulmonary compliance. In bleomycin-induced lung fibrosis, TGFβ and BMP signaling followed an inverse course, with dynamic activation of TGFβ signaling and repression of BMP signaling activity. CONCLUSIONS Upon bleomycin exposure, active BMP signaling is decreased. Derepression of BMP signaling in Nog +/LacZ mice protects against bleomycin-induced pulmonary fibrosis. Modulating the balance between BMP and TGFβ, in particular increasing endogenous BMP signals, may therefore be a therapeutic target in fibrotic lung disease.
Collapse
|
38
|
Gogiraju R, Xu X, Bochenek ML, Steinbrecher JH, Lehnart SE, Wenzel P, Kessel M, Zeisberg EM, Dobbelstein M, Schäfer K. Endothelial p53 deletion improves angiogenesis and prevents cardiac fibrosis and heart failure induced by pressure overload in mice. J Am Heart Assoc 2015; 4:jah3850. [PMID: 25713289 PMCID: PMC4345879 DOI: 10.1161/jaha.115.001770] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Cardiac dysfunction developing in response to chronic pressure overload is associated with apoptotic cell death and myocardial vessel rarefaction. We examined whether deletion of tumor suppressor p53 in endothelial cells may prevent the transition from cardiac hypertrophy to heart failure. Methods and Results Mice with endothelial‐specific deletion of p53 (End.p53‐KO) were generated by crossing p53fl/fl mice with mice expressing Cre recombinase under control of an inducible Tie2 promoter. Cardiac hypertrophy was induced by transverse aortic constriction. Serial echocardiography measurements revealed improved cardiac function in End.p53‐KO mice that also exhibited better survival. Cardiac hypertrophy was associated with increased p53 levels in End.p53‐WT controls, whereas banded hearts of End.p53‐KO mice exhibited lower numbers of apoptotic endothelial and non‐endothelial cells and altered mRNA levels of genes regulating cell cycle progression (p21), apoptosis (Puma), or proliferation (Pcna). A higher cardiac capillary density and improved myocardial perfusion was observed, and pharmacological inhibition or genetic deletion of p53 also promoted endothelial sprouting in vitro and new vessel formation following hindlimb ischemia in vivo. Hearts of End.p53‐KO mice exhibited markedly less fibrosis compared with End.p53‐WT controls, and lower mRNA levels of p53‐regulated genes involved in extracellular matrix production and turnover (eg, Bmp‐7, Ctgf, or Pai‐1), or of transcription factors involved in controlling mesenchymal differentiation were observed. Conclusions Our analyses reveal that accumulation of p53 in endothelial cells contributes to blood vessel rarefaction and fibrosis during chronic cardiac pressure overload and suggest that endothelial cells may be a therapeutic target for preserving cardiac function during hypertrophy.
Collapse
Affiliation(s)
- Rajinikanth Gogiraju
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (R.G., X.X., J.H.S., S.E.L., E.M.Z., K.S.)
| | - Xingbo Xu
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (R.G., X.X., J.H.S., S.E.L., E.M.Z., K.S.)
| | - Magdalena L Bochenek
- Division of Cardiology, Department of Medicine 2, University Medical Center Mainz, Germany (M.L.B., P.W., K.S.) Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (M.L.B., P.W.)
| | - Julia H Steinbrecher
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (R.G., X.X., J.H.S., S.E.L., E.M.Z., K.S.)
| | - Stephan E Lehnart
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (R.G., X.X., J.H.S., S.E.L., E.M.Z., K.S.) German Center for Cardiovascular Research (DZHK), University of Maryland Baltimore, MD (S.E.L., E.M.Z., K.S.) Center for Biomedical Engineering and Technology, University of Maryland Baltimore, MD (S.E.L.)
| | - Philip Wenzel
- Division of Cardiology, Department of Medicine 2, University Medical Center Mainz, Germany (M.L.B., P.W., K.S.) Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (M.L.B., P.W.)
| | - Michael Kessel
- Department of Developmental Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany (M.K.)
| | - Elisabeth M Zeisberg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (R.G., X.X., J.H.S., S.E.L., E.M.Z., K.S.) German Center for Cardiovascular Research (DZHK), University of Maryland Baltimore, MD (S.E.L., E.M.Z., K.S.)
| | - Matthias Dobbelstein
- Institute of Molecular Oncology, University Medical Center Göttingen, Germany (M.D.)
| | - Katrin Schäfer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Germany (R.G., X.X., J.H.S., S.E.L., E.M.Z., K.S.) Division of Cardiology, Department of Medicine 2, University Medical Center Mainz, Germany (M.L.B., P.W., K.S.) German Center for Cardiovascular Research (DZHK), University of Maryland Baltimore, MD (S.E.L., E.M.Z., K.S.)
| |
Collapse
|
39
|
Lowery JW, Intini G, Gamer L, Lotinun S, Salazar VS, Ote S, Cox K, Baron R, Rosen V. Loss of BMPR2 leads to high bone mass due to increased osteoblast activity. J Cell Sci 2015; 128:1308-15. [PMID: 25663702 DOI: 10.1242/jcs.156737] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Imbalances in the ratio of bone morphogenetic protein (BMP) versus activin and TGFβ signaling are increasingly associated with human diseases yet the mechanisms mediating this relationship remain unclear. The type 2 receptors ACVR2A and ACVR2B bind BMPs and activins but the type 2 receptor BMPR2 only binds BMPs, suggesting that type 2 receptor utilization might play a role in mediating the interaction of these pathways. We tested this hypothesis in the mouse skeleton, where bone mass is reciprocally regulated by BMP signaling and activin and TGFβ signaling. We found that deleting Bmpr2 in mouse skeletal progenitor cells (Bmpr2-cKO mice) selectively impaired activin signaling but had no effect on BMP signaling, resulting in an increased bone formation rate and high bone mass. Additionally, activin sequestration had no effect on bone mass in Bmpr2-cKO mice but increased bone mass in wild-type mice. Our findings suggest a novel model whereby BMPR2 availability alleviates receptor-level competition between BMPs and activins and where utilization of ACVR2A and ACVR2B by BMPs comes at the expense of activins. As BMP and activin pathway modulation are of current therapeutic interest, our findings provide important mechanistic insight into the relationship between these pathways in human health.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Department of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, USA Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Giuseppe Intini
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Laura Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sutada Lotinun
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Valerie S Salazar
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Satoshi Ote
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Karen Cox
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Roland Baron
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| |
Collapse
|
40
|
Oncostatin M overexpression induces matrix deposition, STAT3 activation, and SMAD1 Dysregulation in lungs of fibrosis-resistant BALB/c mice. J Transl Med 2014; 94:1003-16. [PMID: 24933422 DOI: 10.1038/labinvest.2014.81] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 05/02/2014] [Accepted: 05/15/2014] [Indexed: 02/06/2023] Open
Abstract
Adverse health outcomes in pulmonary fibrosis are associated with extracellular matrix (ECM) accumulation. Although transforming growth factor-β (TGF-β) has been reported to be an important regulator of fibrosis pathogenesis, TGF-β-independent pathways may also be involved. Here, we investigated responses of putative relatively fibrosis-resistant BALB/c mice to transient pulmonary overexpression of oncostatin M (OSM) using an adenovirus vector encoding OSM (AdOSM) and compared responses with the relatively fibrosis-prone C57Bl/6 strain. Interestingly, BALB/c mice showed similar ECM accumulation and collagen 1A1 and 3A1 mRNA elevation to C57Bl/6 mice 7 days after endotracheal administration of AdOSM. TGF-β1 mRNA levels and pSMAD2 signal were not regulated in either strain in total lung extracts. In contrast to C57Bl/6 mice, BALB/c mice lacked eosinophil, Th2 cytokine, and pro-inflammatory cytokine elevation in the broncholveolar space. OSM overexpression induced STAT3 activation and SMAD1/5/8 signaling suppression in lung from both mice strains, which was associated with a downregulation of BMPR2 and BMP ligands, and increased expression of the BMP antagonist gremlin. Although we also observed STAT3 activation and SMAD1/5/8 signaling suppression in mouse lung fibroblast cultures in vitro upon OSM stimulation, immunohistochemistry analyses indicated that the AdOSM-induced pSMAD1/5/8 signal suppression was primarily localized to the airway epithelium. Other gp130 cytokines including IL-6, LIF, CT-1, but not IL-31, also induced STAT3 activation and SMAD1/5/8 signaling suppression in C10 mouse lung epithelial cells and BEAS 2B bronchial epithelial cells, and we found that pharmacological inhibition of STAT3 activation reversed OSM-induced SMAD1/5/8 signaling suppression in vitro. The results demonstrate that OSM induces ECM accumulation in fibrosis-resistant BALB/c mouse lung in the absence of Th2 inflammation or TGF-β signaling, and highlight a dichotomy of STAT3 activation versus SMAD1 suppression in this process.
Collapse
|
41
|
Hasslund S, Dadali T, Ulrich-Vinther M, Søballe K, Schwarz EM, Awad HA. Freeze-dried allograft-mediated gene or protein delivery of growth and differentiation factor 5 reduces reconstructed murine flexor tendon adhesions. J Tissue Eng 2014; 5:2041731414528736. [PMID: 24812579 PMCID: PMC4014079 DOI: 10.1177/2041731414528736] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 02/24/2014] [Indexed: 01/04/2023] Open
Abstract
Advances in allograft processing have opened new horizons for clinical adaptation of flexor tendon allografts as delivery scaffolds for antifibrotic therapeutics. Recombinant adeno-associated-virus (rAAV) gene delivery of the growth and differentiation factor 5 (GDF-5) has been previously associated with antifibrotic effects in a mouse model of flexor tendoplasty. In this study, we compared the effects of loading freeze-dried allografts with different doses of GDF-5 protein or rAAV-Gdf5 on flexor tendon healing and adhesions. We first optimized the protein and viral loading parameters using reverse transcription polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and in vivo bioluminescent imaging. We then reconstructed flexor digitorum longus (FDL) tendons of the mouse hindlimb with allografts loaded with low and high doses of recombinant GDF-5 protein and rAAV-Gdf5 and evaluated joint flexion and biomechanical properties of the reconstructed tendon. In vitro optimization studies determined that both the loading time and concentration of the growth factor and viral vector had dose-dependent effects on their retention on the freeze-dried allograft. In vivo data suggest that protein and gene delivery of GDF-5 had equivalent effects on improving joint flexion function, in the range of doses used. Within the doses tested, the lower doses of GDF-5 had more potent effects on suppressing adhesions without adversely affecting the strength of the repair. These findings indicate equivalent antifibrotic effects of Gdf5 gene and protein delivery, but suggest that localized delivery of this potent factor should also carefully consider the dosage used to eliminate untoward effects, regardless of the delivery mode.
Collapse
Affiliation(s)
- Sys Hasslund
- The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Tulin Dadali
- The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Kjeld Søballe
- Department of Orthopedics, Aarhus University Hospital, Aarhus, Denmark
| | - Edward M Schwarz
- The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Hani A Awad
- The Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
42
|
Upton PD, Davies RJ, Tajsic T, Morrell NW. Transforming growth factor-β(1) represses bone morphogenetic protein-mediated Smad signaling in pulmonary artery smooth muscle cells via Smad3. Am J Respir Cell Mol Biol 2014; 49:1135-45. [PMID: 23937428 DOI: 10.1165/rcmb.2012-0470oc] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Previous studies of pulmonary arterial hypertension (PAH) have implicated excessive transforming growth factor (TGF)-β1 signaling and reduced bone morphogenetic protein (BMP) signaling in the disease pathogenesis. Reduced BMP signaling in pulmonary artery smooth muscle cells (PASMCs) from patients with heritable PAH is a consequence of germline mutations in the BMP type II receptor (BMPR-II). We sought to establish whether the TGF-β1 and BMP4 pathways interact in PASMCs, and if this is altered in cells with BMPR-II mutations. Control PASMCs or from patients with PAH harboring BMPR-II mutations were treated with BMP4, TGF-β1, or cotreated with both ligands. Signaling was assessed by examination of Smad phosphorylation, luciferase reporters, and the transcription of BMP4 or TGF-β1-responsive genes. TGF-β1 attenuated BMP4-mediated inhibitors of differentiation 1/2 induction and abolished the response in BMPR-II mutant PASMCs, whereas BMP4 did not alter TGF-β1-mediated transcription. Activin-like kinase 5 inhibition blocked this effect, whereas cycloheximide or pharmacological inhibitors of TGF-β-activated kinase 1, extracellular signal-regulated kinase 1/2, or p38 mitogen-activated protein kinase were ineffective. BMP4 and TGF-β1 cotreatment did not alter the activation or nuclear translocation of their respective Smad signaling proteins. Small interfering RNA for Smad3, but not Smad2, Smad6, or Smad7, reversed the inhibition by TGF-β1. In addition, TGF-β-activated kinase 1 inhibition blocked Smad3 phosphorylation, implying that C-terminal Smad3 phosphorylation is not required for the inhibition of BMP4 signaling by TGF-β1. TGF-β1 reduces BMP4 signaling in PASMCs, a response that is exacerbated on the background of reduced BMP responsiveness due to BMPR-II mutations. These data provide a rationale for therapeutic inhibition of TGF-β1 signaling in PAH.
Collapse
Affiliation(s)
- Paul D Upton
- 1 Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
43
|
Yasmin N, Bauer T, Modak M, Wagner K, Schuster C, Köffel R, Seyerl M, Stöckl J, Elbe-Bürger A, Graf D, Strobl H. Identification of bone morphogenetic protein 7 (BMP7) as an instructive factor for human epidermal Langerhans cell differentiation. ACTA ACUST UNITED AC 2013; 210:2597-610. [PMID: 24190429 PMCID: PMC3832935 DOI: 10.1084/jem.20130275] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Bone morphogenetic protein 7 (BMP7) promotes the differentiation of Langerhans cells in the epidermis during prenatal development. Human Langerhans cell (LC) precursors populate the epidermis early during prenatal development and thereafter undergo massive proliferation. The prototypic antiproliferative cytokine TGF-β1 is required for LC differentiation from human CD34+ hematopoietic progenitor cells and blood monocytes in vitro. Similarly, TGF-β1 deficiency results in LC loss in vivo. However, immunohistology studies revealed that human LC niches in early prenatal epidermis and adult basal (germinal) keratinocyte layers lack detectable TGF-β1. Here we demonstrated that these LC niches express high levels of bone morphogenetic protein 7 (BMP7) and that Bmp7-deficient mice exhibit substantially diminished LC numbers, with the remaining cells appearing less dendritic. BMP7 induces LC differentiation and proliferation by activating the BMP type-I receptor ALK3 in the absence of canonical TGF-β1–ALK5 signaling. Conversely, TGF-β1–induced in vitro LC differentiation is mediated via ALK3; however, co-induction of ALK5 diminished TGF-β1–driven LC generation. Therefore, selective ALK3 signaling by BMP7 promotes high LC yields. Within epidermis, BMP7 shows an inverse expression pattern relative to TGF-β1, the latter induced in suprabasal layers and up-regulated in outer layers. We observed that TGF-β1 inhibits microbial activation of BMP7-generated LCs. Therefore, TGF-β1 in suprabasal/outer epidermal layers might inhibit LC activation, resulting in LC network maintenance.
Collapse
Affiliation(s)
- Nighat Yasmin
- Institute of Pathophysiology and Immunology, Center for Molecular Medicine and 2 Center for Medical Research, Medical University Graz, A-8036 Graz, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Quarto N, Li S, Renda A, Longaker MT. Exogenous activation of BMP-2 signaling overcomes TGFβ-mediated inhibition of osteogenesis in Marfan embryonic stem cells and Marfan patient-specific induced pluripotent stem cells. Stem Cells 2013; 30:2709-19. [PMID: 23037987 DOI: 10.1002/stem.1250] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 08/23/2012] [Indexed: 01/10/2023]
Abstract
Marfan syndrome (MFS) is a hereditary disease caused by mutations in the gene encoding Fibrillin-1 (FBN1) and characterized by a number of skeletal abnormalities, aortic root dilatation, and sometimes ectopia lentis. Although the molecular pathogenesis of MFS was attributed initially to a structural weakness of the fibrillin-rich microfibrils within the extracellular matrix, more recent results have documented that many of the pathogenic abnormalities in MFS are the result of alterations in TGFβ signaling. Mutations in FBN1 are therefore associated with increased activity and bioavailability of TGF-β1, which is suspected to be the basis for phenotypical similarities of FBN1 mutations in MFS and mutations in the receptors for TGFβ in Marfan syndrome-related diseases. We have previously demonstrated that unique skeletal phenotypes observed in human embryonic stem cells carrying the monogenic FBN1 mutation (MFS cells) are faithfully phenocopied by cells differentiated from induced pluripotent-stem cells (MFSiPS) derived independently from MFS patient fibroblasts. In this study, we aimed to determine further the biochemical features of transducing signaling(s) in MFS stem cells and MFSiPS cells highlighting a crosstalk between TGFβ and BMP signaling. Our results revealed that enhanced activation of TGFβ signaling observed in MFS cells decreased their endogenous BMP signaling. Moreover, exogenous BMP antagonized the enhanced TGFβ signaling in both MFS stem cells and MFSiPS cells therefore, rescuing their ability to undergo osteogenic differentiation. This study advances our understanding of molecular mechanisms underlying the pathogenesis of bone loss/abnormal skeletogenesis in human diseases caused by mutations in FBN1.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, California 94305, USA.
| | | | | | | |
Collapse
|
45
|
Wen W, Wang AM, Liu DL, Zhang YB, Yao LQ, Lai GX. Expression of connective tissue growth factor and bone morphogenetic protein-7 in Pseudomonas aeruginosa-induced chronic obstructive pulmonary disease in rats. COPD 2013; 10:657-66. [PMID: 23848567 DOI: 10.3109/15412555.2013.814625] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Repeated intratracheal injection of Pseudomonas aeruginosa (PA) in male Wistar rats was used to investigate the role of chronic infection in the development of chronic obstructive pulmonary disease (COPD) and the possible involvement of connective tissue growth factor (CTGF) and bone morphogenetic protein-7 (BMP-7) in this process. Injections of PA or normal saline solution were given for 8 weeks and the rats observed for a further 8 weeks. In addition to arterial blood gas, lung function and lung pathology measurement during this time period, protein and mRNA expression of CTGF and BMP-7 were measured, and the correlation of expression of CTGF and BMP-7 with pathological changes in the lung was evaluated. Repeated intratracheal PA infection in rats caused reduction in body weight, hypoxia, carbon dioxide retention, compromised lung function, chronic inflammation, thickening of the tracheal and arterial walls, and emphysema, changes consistent with those of COPD. Rats with PA infection also had increased CTGF and decreased BMP-7 expression, suggesting that both CTGF and BMP-7 are involved in the occurrence and development of airway remodeling. Our findings suggest that repeated airway infection is not only a factor resulting in deterioration of COPD, but is also a risk factor for its development, and that CTGF and BMP-7 are involved in the pathogenesis of this condition.
Collapse
Affiliation(s)
- Wen Wen
- 1Department of Respiratory and Critical Care Medicine, Fuzhou General Hospital of Nanjing Commond, Dongfang Hospital, Xiamen University, Fuzhou, China
| | | | | | | | | | | |
Collapse
|
46
|
Zhao L, Yee M, O'Reilly MA. Transdifferentiation of alveolar epithelial type II to type I cells is controlled by opposing TGF-β and BMP signaling. Am J Physiol Lung Cell Mol Physiol 2013; 305:L409-18. [PMID: 23831617 DOI: 10.1152/ajplung.00032.2013] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Alveolar epithelial type II (ATII) cells are essential for maintaining normal lung homeostasis because they produce surfactant, express innate immune proteins, and can function as progenitors for alveolar epithelial type I (ATI) cells. Although autocrine production of transforming growth factor (TGF)-β1 has been shown to promote the transdifferentiation of primary rat ATII to ATI cells in vitro, mechanisms controlling this process still remain poorly defined. Here, evidence is provided that Tgf-β1, -2, -3 mRNA and phosphorylated SMAD2 and SMAD3 significantly increase as primary cultures of mouse ATII cells transdifferentiate to ATI cells. Concomitantly, bone morphogenetic protein (Bmp)-2 and -4 mRNA, and phosphorylated SMAD1/5/8 expression decrease. Exogenously supplied recombinant human TGF-β1 inhibited BMP signaling and enhanced transdifferentiation by promoting the loss of ATII cell-specific gene expression and weakly stimulating ATI cell-specific gene expression. On the other hand, exogenously supplied recombinant human BMP-4 inhibited TGF-β signaling and delayed transdifferentiation by inhibiting the gain in ATI cell-specific gene expression and weakly delaying the loss of ATII cell-specific gene expression. In mouse lung epithelial (MLE15) cells, small-interfering RNA (siRNA) knockdown of TGF-β receptor type-1 enhanced basal expression of ATII genes while siRNA RNA knockdown of BMP receptors type-1a and -1b enhanced basal expression of ATI genes. Together, these results suggest that the rate of ATII cell transdifferentiation is controlled by the opposing actions of BMP and TGF-β signaling that switch during the process of transdifferentiation.
Collapse
Affiliation(s)
- Lan Zhao
- Dept. of Pediatrics, Box 850, The Univ. of Rochester, School of Medicine and Dentistry, 601 Elmwood Ave., Rochester NY 14642.
| | | | | |
Collapse
|
47
|
Tan HC, Poh CK, Cai Y, Wang W. Anti-fibrosis effect of BMP-7 peptide functionalization on cobalt chromium alloy. J Orthop Res 2013; 31:983-90. [PMID: 23456668 DOI: 10.1002/jor.22313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 12/20/2012] [Indexed: 02/04/2023]
Abstract
Orthopedic metallic prosthetic implants are commonly made of cobalt chromium (CoCr) alloys. However, such metal-based implants are susceptible to fibrous capsule formation on the implant surface after implantation. At the bone-implant interface, this capsule can prevent implant integration, resulting in loosening and failure. Minimizing the development of such a capsule on the CoCr surface would improve direct bone-implant bonding leading to long-term implant functionality. We evaluated the anti-fibrosis effect of bone morphogenic protein-7 (BMP-7) peptide covalently bonded to CoCr alloy. This peptide, a biomimetic derivation of the knuckle epitope of BMP-7, was conjugated at the N-terminus with a cysteine amino acid. X-ray photoelectron spectroscopy (XPS) and probe binding assay were used to evaluate different stages of grafting and surface functionalization using polydopamine coating. Cellular functions were studied using fibroblast attachment, cell proliferation, and MTT assays. Fibroblasts were grown on functionalized and pristine CoCr substrates, and the efficacy of BMP-7 peptide on anti-fibrosis was analyzed via gene expression and protein expression of fibrosis markers ACTA2, Collagen 1A1, and fibronectin. The peptide functionalized substrates showed significant reduction of fibrosis markers expression after 1 week of incubation compared to controls. BMP-7 signaling pathway activation was shown by the presence of phosphorylation of Smad1/5/8. These findings may contribute to the improvement of CoCr implants in orthopedic surgery applications.
Collapse
Affiliation(s)
- Hark Chuan Tan
- Department of Orthopaedic Surgery, National University of Singapore, Kent Ridge, Singapore 119074, Singapore
| | | | | | | |
Collapse
|
48
|
Ohba T, Wada H, Yoshino I, Yoshida S, Tagawa T, Shoji F, Yamazaki K, Maehara Y. Increase of bone morphogenetic protein-7 expressing pulmonary resident cells in pneumonectomized rats. Surg Today 2013; 44:324-31. [PMID: 23640479 DOI: 10.1007/s00595-013-0604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 12/09/2012] [Indexed: 11/28/2022]
Abstract
PURPOSE Compensatory lung growth (CLG) is recognized in rodents subjected to major pulmonary resection; however, the source of cells constituting regenerated tissues during the CLG is still unknown. We investigated the differentiation of lung resident cells and the participation of bone marrow (BM)-derived cells in the remnant lung of pneumonectomized rats. METHODS After left pneumonectomy, the right remnant lung of Wistar rats was subjected to morphologic and molecular experiments at several time points. We studied the expression of bone morphogenic protein 7 (BMP-7), an accelerator of epithelial differentiation, based on the gene expression profile data of the remnant lung. Next, we evaluated the presence of GFP-positive cells in the remnant lung of Wistar rats that had received BM transplantation from green fluorescent protein (GFP) gene-transgenic Wistar rats prior to left pneumonectomy. RESULTS We observed progression of emphysematous change, modulation of gene expression profile, and proliferating cellular nuclear antigen-positive cells in the alveoli of the remnant lungs. BMP-7 protein positive cells were detected in the alveolar septa, which increased significantly over time with the progression of emphysematous change. No bone marrow-derived cells were detected in the right remnant lung of the GFP-BM transferred rats by fluorescence microscopy, immunohistochemistry, or polymerase chain reaction at any time. CONCLUSION Lung resident cells appear to contribute to CLG, possibly via a trans-differentiation pathway.
Collapse
Affiliation(s)
- Taro Ohba
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Upton PD, Morrell NW. The transforming growth factor-β-bone morphogenetic protein type signalling pathway in pulmonary vascular homeostasis and disease. Exp Physiol 2013; 98:1262-6. [PMID: 23645549 DOI: 10.1113/expphysiol.2012.069104] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Germ-line mutations in the bone morphogenetic protein type II receptor (BMPR2; BMPR-II) gene, a transforming growth factor-β (TGFβ) receptor superfamily member, cause the majority of cases of heritable pulmonary arterial hypertension (PAH). Pulmonary arterial hypertension is a subset of pulmonary hypertension (PH) disorders, which also encompass hypoxia-related lung diseases. Bone morphogenetic proteins (BMPs), via BMPR-II, activate the canonical Smad1/5/9 pathway, whereas TGFβs (TGFβ1-3) activate the Smad2/3 pathway via the ALK5 receptor. Dysregulated TGFβ1 signalling is pathogenic in fibrotic diseases. We compared two rat PH models, monocrotaline-induced PAH (MCT-PAH) and chronic normobaric hypoxia (fractional inspired O2 10%), to address whether BMPR-II loss is common to PH and permits pathogenic TGFβ1 signalling. Both models exhibited reduced lung BMPR-II expression, but increased TGFβ1 signalling and decreased BMP signalling were observed only in MCT-PAH. Furthermore, a pharmacological ALK5 inhibitor prevented disease progression in the MCT-PAH model, but not in hypoxia. In vitro studies using human pulmonary artery smooth muscle cells showed that TGFβ1 directly inhibits BMP-Smad signalling. In conclusion, BMPR-II loss is common to the hypoxic and MCT-PAH models, but systemic ALK5 inhibition is effective only in the MCT model, highlighting a specific role for TGFβ1 in vascular remodelling in MCT-PAH, potentially via direct inhibition of BMP signalling.
Collapse
Affiliation(s)
- Paul D Upton
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge, School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, UK.
| | | |
Collapse
|
50
|
Hori M, Juniantito V, Izawa T, Ichikawa C, Tanaka M, Tanaka K, Takenaka S, Kuwamura M, Yamate J. Distribution of Cells Labelled by a Novel Somatic Stem Cell-recognizing Antibody (A3) in Pulmonary Genesis and Bleomycin induced Pulmonary Fibrosis in Rats. J Comp Pathol 2013; 148:385-95. [DOI: 10.1016/j.jcpa.2012.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 08/27/2012] [Accepted: 09/03/2012] [Indexed: 11/16/2022]
|