151
|
Inhibition of IL-18 reduces renal fibrosis after ischemia-reperfusion. Biomed Pharmacother 2018; 106:879-889. [PMID: 30119258 DOI: 10.1016/j.biopha.2018.07.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 01/20/2023] Open
Abstract
Acute kidney injury induced by ischemia-reperfusion injury (IRI) is a high risk factor in the progression towards chronic kidney disease, which is featured by renal interstitial fibrosis. Interleukin (IL)-18 is produced by T cells and macrophages and has been involved in the pathophysiology of IRI. However, the role of IL-18 in IRI-induced renal fibrosis is poorly understood. In the present study, we showed that interleukin (IL)-18 was significantly up-regulated after IRI stress. Mice treated with IL-18 Bp, a natural inhibitor of IL-18, presented less severe fibrotic response in the kidneys following IRI compared with vehicle-treated mice. Inhibition of IL-18 decreased myofibroblasts formation in the kidneys in response to IRI, which was associated with reduction of fibronectin and collagenⅠproteins. Moreover, inhibition of IL-18 impaired infiltration of CD3+ T cells and F4/80+ macrophages in the kidneys of mice after IRI. Treatment with IL-18 Bp reduces the levels of profibrotic molecules in the kidneys of mice following IRI. Finally, administration of IL-18 Bp impedes the transition of M2 macrophages to myofibroblasts and suppressed the accumulation of bone marrow-derived M2 macrophages. Adoptive transfer of M2 macrophages abolished the anti-fibrotic effect of IL-18 Bp. In summary, our results suggest that IL-18 plays an important role in the progression of IRI-induced renal fibrosis via modulating inflammation cells infiltration, the expression of inflammatory cytokines and chemokines, and the transition of bone marrow-derived M2 macrophages to myofibroblasts.
Collapse
|
152
|
The preventive and therapeutic implication for renal fibrosis by targetting TGF-β/Smad3 signaling. Clin Sci (Lond) 2018; 132:1403-1415. [PMID: 29875262 DOI: 10.1042/cs20180243] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 01/04/2023]
Abstract
It is well established that Smad3 is a key downstream effector of transforming growth factor-β (TGF-β) signaling in tissue fibrogenesis. We reported here that targetting Smad3 specifically with a Smad3 inhibitor SIS3 is able to prevent or halt the progression of renal fibrosis in a mouse model of unilateral ureteral obstructive nephropathy (UUO). We found that preventive treatment with SIS3 at the time of disease induction largely suppressed progressive renal fibrosis by inhibiting α-smooth muscle actin (α-SMA) + myofibroblast accumulation and extracellular matrix (collagen I (Col.I) and fibronectin (FN)) production. Importantly, we also found that treatment with SIS3 on established mouse model of UUO from day 4 after UUO nephropathy halted the progression of renal fibrosis. Mechanistically, the preventive and therapeutic effects of SIS3 on renal fibrosis were associated with the inactivation of Smad3 signaling and inhibition of TGF-β1 expression in the UUO kidney. In conclusion, results from the present study suggest that targetting Smad3 may be a specific and effective therapy for renal fibrosis.
Collapse
|
153
|
The correlation between pulmonary fibrosis and methylation of peripheral Smad3 in cases of pigeon breeder's lung in a Chinese Uygur population. Oncotarget 2018; 8:43104-43113. [PMID: 28562330 PMCID: PMC5522131 DOI: 10.18632/oncotarget.17763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 04/06/2017] [Indexed: 11/25/2022] Open
Abstract
Smad3 is a key protein in the transforming growth factor-beta (TGF-β)/Smad signaling pathway, which is involved in fibrosis in many organs. We investigated the relationship between Smad3 gene methylation and pulmonary fibrosis in pigeon breeder's lung (PBL). Twenty Uygur PBL patients with pulmonary fibrosis in Kashi between October 2015 and March 2016 were enrolled. Twenty PBL-free pigeon breeders and 20 healthy non-pigeon breeders enrolled during the same period constituted the negative and normal control groups, respectively. Participants’ data and peripheral blood samples were collected, and three Smad3 CpG loci were examined. Distributions of CpG_2 and CpG_4 methylation rates did not differ across groups, whereas distributions of CpG_3 methylation rates were significantly different among the three groups. The CpG_3 methylation rate was significantly lower in the patient group than in the negative control group. Smad3 mRNA expression was significantly higher in the patient group than in the negative control group but did not differ between the two control groups. TGF-βlevels were significantly higher in the patient group than in either control group (both P<0.01). Smad3 gene methylation and Smad3 mRNA expression were negatively correlated, with a correlation coefficient of -0.84. The number of pigeons bred during the preceding three months was positively correlated with Smad3 mRNA expression, with a correlation coefficient of 0.77. Smad3 gene hypomethylation might promote pulmonary fibrosis in Uygur PBL patients via increased Smad3 mRNA expression. Smad3 methylation, Smad3 mRNA expression and TGF-β level were correlated with the number of pigeons bred by patients.
Collapse
|
154
|
Kramann R, Machado F, Wu H, Kusaba T, Hoeft K, Schneider RK, Humphreys BD. Parabiosis and single-cell RNA sequencing reveal a limited contribution of monocytes to myofibroblasts in kidney fibrosis. JCI Insight 2018; 3:99561. [PMID: 29720573 DOI: 10.1172/jci.insight.99561] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Fibrosis is the common final pathway of virtually all chronic injury to the kidney. While it is well accepted that myofibroblasts are the scar-producing cells in the kidney, their cellular origin is still hotly debated. The relative contribution of proximal tubular epithelium and circulating cells, including mesenchymal stem cells, macrophages, and fibrocytes, to the myofibroblast pool remains highly controversial. Using inducible genetic fate tracing of proximal tubular epithelium, we confirm that the proximal tubule does not contribute to the myofibroblast pool. However, in parabiosis models in which one parabiont is genetically labeled and the other is unlabeled and undergoes kidney fibrosis, we demonstrate that a small fraction of genetically labeled renal myofibroblasts derive from the circulation. Single-cell RNA sequencing confirms this finding but indicates that these cells are circulating monocytes, express few extracellular matrix or other myofibroblast genes, and express many proinflammatory cytokines. We conclude that this small circulating myofibroblast progenitor population contributes to renal fibrosis by paracrine rather than direct mechanisms.
Collapse
Affiliation(s)
- Rafael Kramann
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Flavia Machado
- Division of Nephrology, Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Haojia Wu
- Division of Nephrology, Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tetsuro Kusaba
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Konrad Hoeft
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Rebekka K Schneider
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands.,Division of Hematology, RWTH Aachen University, Aachen, Germany
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine and Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
155
|
Jiang Y, Wang Y, Ma P, An D, Zhao J, Liang S, Ye Y, Lu Y, Zhang P, Liu X, Han H, Qin H. Myeloid-specific targeting of Notch ameliorates murine renal fibrosis via reduced infiltration and activation of bone marrow-derived macrophage. Protein Cell 2018; 10:196-210. [PMID: 29644573 PMCID: PMC6338623 DOI: 10.1007/s13238-018-0527-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/28/2018] [Indexed: 12/25/2022] Open
Abstract
Macrophages play critical roles in renal fibrosis. However, macrophages exhibit ontogenic and functional heterogeneities, and which population of macrophages contributes to renal fibrosis and the underlying mechanisms remain unclear. In this study, we genetically targeted Notch signaling by disrupting the transcription factor recombination signal binding protein-Jκ (RBP-J), to reveal its role in regulation of macrophages during the unilateral ureteral obstruction (UUO)-induced murine renal fibrosis. Myeloid-specific disruption of RBP-J attenuated renal fibrosis with reduced extracellular matrix deposition and myofibroblast activation, as well as attenuated epithelial-mesenchymal transition, likely owing to the reduced expression of TGF-β. Meanwhile, RBP-J deletion significantly hampered macrophage infiltration and activation in fibrotic kidney, although their proliferation appeared unaltered. By using macrophage clearance experiment, we found that kidney resident macrophages made negligible contribution, but bone marrow (BM)-derived macrophages played a major role in renal fibrogenesis. Further mechanistic analyses showed that Notch blockade reduced monocyte emigration from BM by down-regulating CCR2 expression. Finally, we found that myeloid-specific Notch activation aggravated renal fibrosis, which was mediated by CCR2+ macrophages infiltration. In summary, our data have unveiled that myeloid-specific targeting of Notch could ameliorate renal fibrosis by regulating BM-derived macrophages recruitment and activation, providing a novel strategy for intervention of this disease.
Collapse
Affiliation(s)
- Yali Jiang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #15, Xi'an, 710032, China
| | - Yuanyuan Wang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China.,Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #15, Xi'an, 710032, China
| | - Pengfei Ma
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Dongjie An
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Junlong Zhao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Shiqian Liang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuchen Ye
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Yingying Lu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #15, Xi'an, 710032, China
| | - Peng Zhang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #15, Xi'an, 710032, China
| | - Xiaowei Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #15, Xi'an, 710032, China.
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Hongyan Qin
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
156
|
Luong VH, Chino T, Oyama N, Matsushita T, Sasaki Y, Ogura D, Niwa SI, Biswas T, Hamasaki A, Fujita M, Okamoto Y, Otsuka M, Ihn H, Hasegawa M. Blockade of TGF-β/Smad signaling by the small compound HPH-15 ameliorates experimental skin fibrosis. Arthritis Res Ther 2018; 20:46. [PMID: 29544542 PMCID: PMC5855969 DOI: 10.1186/s13075-018-1534-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 02/04/2018] [Indexed: 12/20/2022] Open
Abstract
Background Transforming growth factor-β (TGF-β)/Smad signaling is well known to play a critical role in the pathogenesis of systemic sclerosis (SSc). We previously developed an artificial molecule, the histidine-pyridine-histidine ligand derivative HPH-15, which may have an antifibrotic effect. The purpose of the present study was to clarify the effects of this drug in human skin fibroblasts and in a preclinical model of SSc. Methods The effects of HPH-15 on expression of extracellular matrix components and TGF-β signaling in human dermal fibroblasts were analyzed. The antifibrotic properties of HPH-15 and its mechanisms were also examined in a bleomycin-induced skin fibrosis mouse model. Results HPH-15 suppressed the TGF-β-induced phosphorylation of Smad3 and inhibited the expression of collagen I, fibronectin 1, connective tissue growth factor, and α-smooth muscle actin induced by TGF-β in cultured human skin fibroblasts. In the bleomycin-induced skin fibrosis model, oral administration of HPH-15 protected against the development of skin fibrosis and ameliorated established skin fibrosis. Additionally, HPH-15 suppressed the phosphorylation of Smad3 in various cells, including macrophages in the bleomycin-injected skin. Further, in the treated mice, dermal infiltration of proinflammatory macrophages (CD11b+Ly6Chi) and M2 profibrotic macrophages (CD11b+CD204+ or CD11b+CD206+) was significantly decreased during the early and late stages, respectively. HPH-15 treatment resulted in decreased messenger RNA (mRNA) expression of the M2 macrophage markers arginase 1 and Ym-1 in the skin, whereas it inversely augmented expression of Friend leukemia integration 1 and Krüppel-like factor 5 mRNAs, the transcription factors that repress collagen synthesis. No apparent adverse effects of HPH-15 were found during the treatment. Conclusions HPH-15 may inhibit skin fibrosis by inhibiting the phosphorylation of Smad3 in dermal fibroblasts and possibly in macrophages. Our results demonstrate several positive qualities of HPH-15, including oral bioavailability, a good safety profile, and therapeutic effectiveness. Thus, this TGF-β/Smad inhibitor is a potential candidate therapeutic for SSc clinical trials. Electronic supplementary material The online version of this article (10.1186/s13075-018-1534-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vu Huy Luong
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Takenao Chino
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Noritaka Oyama
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Takashi Matsushita
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | | | | | | | - Tanima Biswas
- Department of Bioorganic Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Akiyuki Hamasaki
- Department of Bioorganic Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Mikako Fujita
- Research Institute for Drug Discovery, Kumamoto University, Kumamoto, Japan
| | - Yoshinari Okamoto
- Department of Bioorganic Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Masami Otsuka
- Department of Bioorganic Medicinal Chemistry, Kumamoto University, Kumamoto, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Minoru Hasegawa
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| |
Collapse
|
157
|
Li Y, Liu TM. Discovering Macrophage Functions Using In Vivo Optical Imaging Techniques. Front Immunol 2018; 9:502. [PMID: 29599778 PMCID: PMC5863475 DOI: 10.3389/fimmu.2018.00502] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/26/2018] [Indexed: 12/27/2022] Open
Abstract
Macrophages are an important component of host defense and inflammation and play a pivotal role in immune regulation, tissue remodeling, and metabolic regulation. Since macrophages are ubiquitous in human bodies and have versatile physiological functions, they are involved in virtually every disease, including cancer, diabetes, multiple sclerosis, and atherosclerosis. Molecular biological and histological methods have provided critical information on macrophage biology. However, many in vivo dynamic behaviors of macrophages are poorly understood and yet to be discovered. A better understanding of macrophage functions and dynamics in pathogenesis will open new opportunities for better diagnosis, prognostic assessment, and therapeutic intervention. In this article, we will review the advances in macrophage tracking and analysis with in vivo optical imaging in the context of different diseases. Moreover, this review will cover the challenges and solutions for optical imaging techniques during macrophage intravital imaging.
Collapse
Affiliation(s)
- Yue Li
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Tzu-Ming Liu
- Faculty of Health Sciences, University of Macau, Macao, China
| |
Collapse
|
158
|
Sraeyes S, Pham DH, Gee TW, Hua J, Butcher JT. Monocytes and Macrophages in Heart Valves: Uninvited Guests or Critical Performers? CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 5:82-89. [PMID: 30276357 PMCID: PMC6162070 DOI: 10.1016/j.cobme.2018.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Monocytes and macrophages are critical components of the myeloid niche of the innate immune system. In addition to traditional roles as phagocytes, this subsection of innate immunity has been implicated in its ability to regulate tissue homeostasis and inflammation across diverse physiological systems. Recent emergence of discriminatory features within the monocyte/macrophage niche within the last 5 years has helped to clarify specific function(s) of the subpopulations of these cells. It is becoming increasingly aware that these cells are likely implicated in valve development and disease. This review seeks to use current literature and opinions to show the diverse roles and potential contributions of this niche throughout valvulogenic processes, adult homeostatic function, valve disease mechanisms, and tissue engineering approaches.
Collapse
Affiliation(s)
- Sridhar Sraeyes
- Nancy E. and Peter C. Meinig School of Biomedical Engineering
| | - Duc H Pham
- Nancy E. and Peter C. Meinig School of Biomedical Engineering
| | - Terence W Gee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering
| | - Joanna Hua
- Nancy E. and Peter C. Meinig School of Biomedical Engineering
| | | |
Collapse
|
159
|
MicroRNA-27a promotes renal tubulointerstitial fibrosis via suppressing PPARγ pathway in diabetic nephropathy. Oncotarget 2018; 7:47760-47776. [PMID: 27351287 PMCID: PMC5216977 DOI: 10.18632/oncotarget.10283] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/12/2016] [Indexed: 12/24/2022] Open
Abstract
MicroRNA-27a (miR-27a) upregulation has been identified in diabetes, but the pathogenesis of miR-27a in renal tubulointerstitial fibrosis (TIF) in diabetic nephropathy (DN) has not been elucidated. Herein, we found that high glucose stimulated miR-27a expression, which directly inhibited PPARγ and promoted fibrosis in NRK-52E cells. The functional relevance of miR-27a-dependent PPARγ decrease was proven by inhibition or overexpression of miR-27a both in vitro and in streptozotocin-induced diabetic rats. MiR-27a, via repression of PPARγ, activates the TGF-β/Smad3 signaling and contributes to the expressional changes of connective tissue growth factor (CTGF), Fibronectin and Collagen I, key mediators of fibrosis. Furthermore, we provide evidences that plasma miR-27a upregulation contributed to unfavorable renal function and increased TIF in renal tissues of diabetic rats and DN patients. Notably, miR-27a exhibited clinical and biological relevance as it was linked to elevated serum creatinine, proteinuria, urinary N-acetyl-β-D-glucosaminidase (NAG), and reduced estimated glomerular filtration rate (eGFR). Thus, we propose a novel role of the miR-27a-PPARγ axis in fostering the progression toward more deteriorated renal TIF in DN. Monitoring plasma miR-27a level and its association with PPARγ can be used to reflect the severity of renal TIF. Targeting miR-27a could be evaluated as a potential therapeutic approach for DN.
Collapse
|
160
|
Shrishrimal S, Kosmacek EA, Chatterjee A, Tyson MJ, Oberley-Deegan RE. The SOD Mimic, MnTE-2-PyP, Protects from Chronic Fibrosis and Inflammation in Irradiated Normal Pelvic Tissues. Antioxidants (Basel) 2017; 6:antiox6040087. [PMID: 29113120 PMCID: PMC5745497 DOI: 10.3390/antiox6040087] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023] Open
Abstract
Pelvic radiation for cancer therapy can damage a variety of normal tissues. In this study, we demonstrate that radiation causes acute changes to pelvic fibroblasts such as the transformation to myofibroblasts and the induction of senescence, which persist months after radiation. The addition of the manganese porphyrin, MnTE-2-PyP, resulted in protection of these acute changes in fibroblasts and this protection persisted months following radiation exposure. Specifically, at two months post-radiation, MnTE-2-PyP inhibited the number of α-smooth muscle actin positive fibroblasts induced by radiation and at six months post-radiation, MnTE-2-PyP significantly reduced collagen deposition (fibrosis) in the skin and bladder tissues of irradiated mice. Radiation also resulted in changes to T cells. At two months post-radiation, there was a reduction of Th1-producing splenocytes, which resulted in reduced Th1:Th2 ratios. MnTE-2-PyP maintained Th1:Th2 ratios similar to unirradiated mice. At six months post-radiation, increased T cells were observed in the adipose tissues. MnTE-2-PyP treatment inhibited this increase. Thus, MnTE-2-PyP treatment maintains normal fibroblast function and T cell immunity months after radiation exposure. We believe that one of the reasons MnTE-2-PyP is a potent radioprotector is due to its protection of multiple cell types from radiation damage.
Collapse
Affiliation(s)
- Shashank Shrishrimal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - McDonald J Tyson
- Department of Physics & Cancer Research Center, Hampton University, Hampton, VA 23668, USA.
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
161
|
Kong W, Fu J, Liu N, Jiao C, Guo G, Luan J, Wang H, Yao L, Wang L, Yamamoto M, Pi J, Zhou H. Nrf2 deficiency promotes the progression from acute tubular damage to chronic renal fibrosis following unilateral ureteral obstruction. Nephrol Dial Transplant 2017; 33:771-783. [PMID: 29126308 DOI: 10.1093/ndt/gfx299] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/04/2017] [Indexed: 01/24/2023] Open
Affiliation(s)
- Weiwei Kong
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Nan Liu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Congcong Jiao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Guangying Guo
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Junjun Luan
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Huihui Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Lining Wang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University of Medicine, Sendai, Japan
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Hua Zhou
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
162
|
Tang PMK, Zhou S, Li CJ, Liao J, Xiao J, Wang QM, Lian GY, Li J, Huang XR, To KF, Ng CF, Chong CCN, Ma RCW, Lee TL, Lan HY. The proto-oncogene tyrosine protein kinase Src is essential for macrophage-myofibroblast transition during renal scarring. Kidney Int 2017; 93:173-187. [PMID: 29042082 DOI: 10.1016/j.kint.2017.07.026] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/18/2017] [Accepted: 07/27/2017] [Indexed: 02/05/2023]
Abstract
Src activation has been associated with fibrogenesis after kidney injury. Macrophage-myofibroblast transition is a newly identified process to generate collagen-producing myofibroblasts locally in the kidney undergoing fibrosis in a TGF-β/Smad3-dependent manner. The potential role of the macrophage-myofibroblast transition in Src-mediated renal fibrosis is unknown. In studying this by RNA sequencing at single-cell resolution, we uncovered a unique Src-centric regulatory gene network as a key underlying mechanism of macrophage-myofibroblast transition. A total of 501 differentially expressed genes associated with macrophage-myofibroblast transition were identified. However, Smad3-knockout largely reduced the transcriptome diversity. More importantly, inhibition of Src largely suppresses ureteral obstruction-induced macrophage-myofibroblast transition in the injured kidney in vivo along with transforming growth factor-β1-induced elongated fibroblast-like morphology, α-smooth muscle actin expression and collagen production in bone marrow derived macrophages in vitro. Unexpectedly, we further uncovered that Src serves as a direct Smad3 target gene and also specifically up-regulated in macrophages during macrophage-myofibroblast transition. Thus, macrophage-myofibroblast transition contributes to Src-mediated tissue fibrosis. Hence, targeting Src may represent as a precision therapeutic strategy for macrophage-myofibroblast transition-driven fibrotic diseases.
Collapse
Affiliation(s)
- Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuang Zhou
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China; Clinical Translational Research Center, Shanghai Pulmonary Hospital, and Department of Histology and Embryology, Tongji University School of Medicine, Tongji University Cancer Institute, Shanghai, China
| | - Chun-Jie Li
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Jinyue Liao
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Jun Xiao
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing-Ming Wang
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Guang-Yu Lian
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jinhong Li
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao-Ru Huang
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chi-Fai Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Ronald Ching-Wa Ma
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tin-Lap Lee
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Hui-Yao Lan
- Li Ka Shing Institute of Health Sciences, and Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
163
|
Cheng Y, Zhu Y, Zhang J, Duan X, Zhang Y. Large Accumulation of Collagen and Increased Activation of Mast Cells in Hearts of Mice with Hyperlipidemia. Arq Bras Cardiol 2017; 109:404-409. [PMID: 28977053 PMCID: PMC5729775 DOI: 10.5935/abc.20170143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hyperlipidemia, which is characterized by an elevation of lipids in the bloodstream, is a major risk factor for cardiac disease. OBJECTIVES The present study investigated the role of fibrosis in the progression of hyperlipidemia in the mice heart, and whether mast cell activation was associated with the fibrosis process. METHODS Hyperlipidemia was produced in C57BL / 6 mice by feeding them on a high-fat diet for 8 weeks.To assess tissue fibrosis, picrosirius red staining was performed. Hematoxylin & eosin (H&E) staining was performed to identify the histopathological changes in the hearts. Immunohistochemistry was also accomplished to determine the localization of transforming growth factor (TGF)-β and α-smooth muscle actin (α-SMA). Western blotting was performed to analyze the expression of chymase, tryptase, TGF-β, α-SMA and activity of Wnt/β-catenin pathway. At the end, serum total cholesterol (TC) and triglycerides (TG) levels were measured. All the values were expressed as means ± SD, the statistical significance level adopted was 5%. RESULTS Hyperlipidemia mice showed significantly increased collagen deposition in the hearts compared with normal mice. In addition, H&E staining showed significant cellular degeneration. Cardiac muscle was arranged in disorder with fracture in mice of the model group. Immunohistochemistry and western blot analysis revealed that expression levels of tryptase, chymase, β-catenin, TGF-β and α-SMA were significantly increased in the hyperlipidemia mice compared with the control group. CONCLUSIONS The results indicated that mast cell activation might induce cardiac fibrosis by tryptase and chymase in hyperlipidemia, which had a close relationship with the increased activity of TGF-β/Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yunpeng Cheng
- Department of Cardiology - the First Affiliated Hospital of Dalian Medical University
| | - Yanqiu Zhu
- Department of Ultrasonics - Affiliated Zhongshan Hospital of Dalian University
| | - Jiashu Zhang
- College of Pharmacy - Dalian Medical University - China
| | - Xingping Duan
- College of Pharmacy - Dalian Medical University - China
| | - Ying Zhang
- Department of Cardiology - the First Affiliated Hospital of Dalian Medical University
| |
Collapse
|
164
|
FGF23 activates injury-primed renal fibroblasts via FGFR4-dependent signalling and enhancement of TGF-β autoinduction. Int J Biochem Cell Biol 2017; 92:63-78. [PMID: 28919046 DOI: 10.1016/j.biocel.2017.09.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/29/2017] [Accepted: 09/14/2017] [Indexed: 01/15/2023]
Abstract
Bone-derived fibroblast growth factor 23 (FGF23) is an important endocrine regulator of mineral homeostasis with effects transduced by cognate FGF receptor (FGFR)1-α-Klotho complexes. Circulating FGF23 levels rise precipitously in patients with kidney disease and portend worse renal and cardiovascular outcomes. De novo expression of FGF23 has been found in the heart and kidney following injury but its significance remains unclear. Studies showing that exposure to chronically high FGF23 concentrations activates hypertrophic gene programs in the cardiomyocyte has spawned intense interest in other pathological off-target effects of FGF23 excess. In the kidney, observational evidence points to a concordance of ectopic renal FGF23 expression and the activation of local transforming growth factor (TGF)-β signalling. Although we have previously shown that FGF23 activates injury-primed renal fibroblasts in vitro, our understanding of the mechanism underpinning these effects was incomplete. Here we show that in the absence of α-Klotho, FGF23 augments pro-fibrotic signalling cascades in injury-primed renal fibroblasts via activation of FGFR4 and upregulation of the calcium transporter, transient receptor potential cation channel 6. The resultant rise in intracellular calcium and production of mitochondrial reactive oxygen species induced expression of NFAT responsive-genes and enhanced TGF-β1 autoinduction through non-canonical JNK-dependent pathways. Reconstitution with transmembrane α-Klotho, or its soluble ectodomain, restored classical Egr signalling and antagonised FGF23-driven myofibroblast differentiation. Thus, renal FGF23 may amplify local myofibroblast activation in injury and perpetuate pro-fibrotic signalling. These findings strengthen the rationale for exploring therapeutic inhibition of FGFR4 or restoration of α-Klotho as upstream regulators of off-target FGF23 effects.
Collapse
|
165
|
Gao P, Qiao XH, Gou LM, Huang Y, Li QH, Li LJ, Wang XY, Li CJ. TGF-β1 attenuated branching morphogenesis of embryonic murine submandibular gland through Smad3 activation. Anat Histol Embryol 2017; 46:600-605. [PMID: 28884513 DOI: 10.1111/ahe.12295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/31/2017] [Indexed: 02/05/2023]
Abstract
Transforming growth factor-β1 (TGF-β1) plays several crucial regulatory roles in multiple physiological and pathological processes. The aim of this work was to investigate the role of TGF-β1 in branching morphogenesis of salivary gland. We harvested and cultured submandibular salivary glands (SMGs) from murine embryos, which were then treated with exogenous TGF-β1, or its neutralized antibody, Smad3 inhibitor, or Smad3 small interfering RNA (siRNA). Our results suggested that TGF-β1 attenuated branching morphogenesis of embryonic murine SMG via Smad3 activation, thus playing a negative regulatory role in salivary gland development.
Collapse
Affiliation(s)
- P Gao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X-H Qiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L-M Gou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Huang
- Department of Oral and Maxillofacial Surgery, Sichuan Provincial Hospital, University of Electronic Science and Technology, Chengdu, China
| | - Q-H Li
- Chinese and Western Medicine Hospital of Panzhihua, Panzhihua, China
| | - L-J Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X-Y Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - C-J Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
166
|
Lin L, He Y, Zhang J, Liu Q, Wang L. The Effects and Possible Mechanisms of Puerarin to Treat Uterine Fibrosis Induced by Ischemia-Reperfusion Injury in Rats. Med Sci Monit 2017; 23:3404-3411. [PMID: 28704345 PMCID: PMC5521046 DOI: 10.12659/msm.902227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Tissues fibrosis is caused by ischemia-reperfusion injury (IRI) and results in organ dysfunction. In this study, we aimed to investigate whether fibrosis occurs after uterine ischemia-reperfusion injury, and to investigate the effects of puerarin (Pur) on the fibrosis process in rats. Material/Methods Twenty-four female Wistar rats were randomly divided into three groups (8 in each group): the control group rats only received operation without uterine ischemic, the IRI group and the IRI + Pur group rats received 30-minutes ischemia and 2-weeks of reperfusion. Pur was orally administered at the onset of reperfusion. Picrosirius red staining was used to assess uterine fibrosis. Immunohistochemistry was used to detect the expression levels of transforming growth factor (TGF)-β and α-smooth muscle actin (α-SMA). Western blotting was used to evaluate the expression of chymase, TGF-β, α-SMA, and the activity of the Wnt/β-catenin pathway. Results Uterine fibrosis in the IRI+Pur group was significantly decreased compared with the IRI group. In addition, immunohistochemistry reveals that TGF-β and α-SMA were decreased in the IRI+Pur group compared with the IRI group. Western blotting results showed that Pur significantly suppresses the increase in chymase, α-SMA, TGF-β, and β-catenin expression levels induced by IRI. Conclusions The results indicated that IRI could induce uterine fibrosis and that Pur had an improvement effect on IRI-induced uterine fibrosis by downregulating the activity of mast cell chymase, TGF-β, α-SMA, and the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Lin Lin
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Yi He
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Jiashu Zhang
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Qi Liu
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (mainland)
| | - Lixia Wang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China (mainland)
| |
Collapse
|
167
|
Smad7 protects against acute kidney injury by rescuing tubular epithelial cells from the G1 cell cycle arrest. Clin Sci (Lond) 2017; 131:1955-1969. [PMID: 28566468 DOI: 10.1042/cs20170127] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 12/16/2022]
Abstract
Smad7 plays a protective role in chronic kidney disease; however, its role in acute kidney injury (AKI) remains unexplored. Here, we report that Smad7 protects against AKI by rescuing the G1 cell cycle arrest of tubular epithelial cells (TECs) in ischemia/reperfusion-induced AKI in mice in which Smad7 gene is disrupted or restored locally into the kidney. In Smad7 gene knockout (KO) mice, more severe renal impairment including higher levels of serum creatinine and massive tubular necrosis was developed at 48 h after AKI. In contrast, restored renal Smad7 gene locally into the kidney of Smad7 KO mice protected against AKI by promoting TEC proliferation identified by PCNA+ and BrdU+ cells. Mechanistic studies revealed that worsen AKI in Smad7 KO mice was associated with a marked activation of TGF-β/Smad3-p21/p27 signaling and a loss of CDK2/cyclin E activities, thereby impairing TEC regeneration at the G1 cell cycle arrest. In contrast, restored Smad7 locally into the kidneys of Smad7 KO mice protected TECs from the G1 cell cycle arrest and promoted TEC G1/S transition via a CDK2/cyclin E-dependent mechanism. In conclusion, Smad7 plays a protective role in AKI. Blockade of TGF-β/Smad3-p21/p27-induced G1 cell cycle arrest may be a key mechanism by which Smad7 treatment inhibits AKI. Thus, Smad7 may be a novel therapeutic agent for AKI.
Collapse
|
168
|
Protection of tubular epithelial cells during renal injury via post-transcriptional control of BMP7. Mol Cell Biochem 2017; 435:141-148. [PMID: 28526933 DOI: 10.1007/s11010-017-3063-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/04/2017] [Indexed: 10/19/2022]
Abstract
Severe injury of renal tubular epithelial cells may cause acute renal failure, the progression of which results in renal fibrosis, and obstructive nephropathy. Transforming growth factor β 1 and bone morphogenic protein 7 (BMP7) play contradicting roles in and coordinate the process of epithelial-to-mesenchymal transition of renal tubular epithelial cells, but the molecular regulation of BMP7 remains ill-defined. Here, we addressed this question. We found that after induction of unilateral ureteral obstruction (UUO) in mice, the increases in BMP7 mRNA were much more pronounced than BMP7 protein in kidney, suggesting the presence of post-transcriptional control of BMP7. Moreover, significant increases in a BMP7-targeting microRNA, miR-384-5p, were detected in the mouse kidney post UUO. Overexpression of miR-384-5p significantly decreased BMP7 protein, while depletion of miR-384-5p significantly increased BMP7 protein in renal epithelial cells. Bioinformatics study showed that miR-384-5p appeared to suppress BMP7 protein translation, through its direct binding to the 3'-UTR of BMP7 mRNA. Furthermore, suppression of miR-384-5p in vivo attenuated severity of renal injury by UUO. Together, our study sheds light on miR-384-5p as a crucial factor that regulates the fibrosis-related pathogenesis after renal injury, and points to miR-384-5p as a promising innovative therapeutic target for prevention of renal fibrosis.
Collapse
|
169
|
Zhang S, Li J, Li Y, Liu Y, Guo H, Xu X. Nitric Oxide Synthase Activity Correlates with OGG1 in Ozone-Induced Lung Injury Animal Models. Front Physiol 2017; 8:249. [PMID: 28496412 PMCID: PMC5406453 DOI: 10.3389/fphys.2017.00249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/07/2017] [Indexed: 12/12/2022] Open
Abstract
Background: NO is an important cellular signaling molecule which is derived from L-arginine by nitric oxide synthase (NOS) and the effects of NOS signaling in lung injury is conflicting. The present study was designed to observe the effect of NOS and Arginase signaling in the occurrence and development of lung injury and its mechanism. Methods: An ozone-stressed lung injury animal model was established by exposure to 2.0 ppm O3 for 30 min every day for consecutive 12 day with or without the administration of NO precursor L-arginine or non-selective NOS inhibitor N-nitro-L-arginine methyl ester (L-NAME). Then, the lung histopathology, the releases of inflammatory mediators and the production of ROS were assayed by immunohistochemistry, ELISA and flow cytometry respectively. The activities and expression of NOS and Arginase were assayed by biochemical methods and western blot. Correspondingly, the release of 8-oxoguanine glycosylase 1(8-OxoG) and 8-oxoguanine glycosylase 1 (OGG1) were assayed by ELISA and western blot. The correlation between NOS/Arginase signaling with 8-OxoG/ OGG1 was also analyzed by Pearson correlation coefficients and immunofluorescence in NOS deficient bronchial epithelial cells. Results: In ozone-induced rat lung injury models, lung inflammation as well as lung architecture was disrupted in a time dependent manner. Ozone treatment with L-arginine showed a substantial attenuation of adverse lung histopathological changes and treatment with L-NAME promoted the inflammation and remodeling. Importantly, the expression of NOS was promoted by L-arginine and inhibited by L-NAME and the expression of Arginase was promoted by L-NAME treatment. Further, we observed significantly higher levels of 8-OxoG and lower levels of OGG1 in ozone group which was reversed by L-arginine and promoted by L-NAME. The expression of NOS is closely related with 8-OxoG /OCG1. Conclusion: These findings give further evidence that the NOS signaling is related with base excise repair.
Collapse
Affiliation(s)
- Suqin Zhang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Jianhua Li
- Department of General Surgery, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Yuqin Li
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Yufeng Liu
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Hongxiang Guo
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Xiaoli Xu
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| |
Collapse
|
170
|
Sun YY, Li XF, Meng XM, Huang C, Zhang L, Li J. Macrophage Phenotype in Liver Injury and Repair. Scand J Immunol 2017; 85:166-174. [PMID: 27491503 DOI: 10.1111/sji.12468] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/19/2016] [Indexed: 12/14/2022]
Abstract
Macrophages hold a critical position in the pathogenesis of liver injury and repair, in which their infiltrations is regarded as a main feature for both acute and chronic liver diseases. It is noted that, based on the distinct phenotypes and origins, hepatic macrophages are capable of clearing pathogens, promoting/or inhibiting liver inflammation, while regulating liver fibrosis and fibrolysis through interplaying with hepatocytes and hepatic stellate cells (HSC) via releasing different types of pro- or anti-inflammatory cytokines and growth factors. Macrophages are typically categorized into M1 or M2 phenotypes by adapting to local microenvironment during the progression of liver injury. In most occasions, M1 macrophages play a pro-inflammatory role in liver injury, while M2 macrophages exert an anti-inflammatory or pro-fibrotic role during liver repair and fibrosis. In this review, we focused on the up-to-date information about the phenotypic and functional plasticity of the macrophages and discussed the detailed mechanisms through which the phenotypes and functions of macrophages are regulated in different stages of liver injury and repair. Moreover, their roles in determining the fate of liver diseases were also summarized. Finally, the macrophage-targeted therapies against liver diseases were also be evaluated.
Collapse
Affiliation(s)
- Y-Y Sun
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Hefei, China
| | - X-F Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Hefei, China
| | - X-M Meng
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Hefei, China
| | - C Huang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Hefei, China
| | - L Zhang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Hefei, China
| | - J Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, China.,Institute for Liver Diseases of Anhui Medical University (AMU), Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Hefei, China
| |
Collapse
|
171
|
Tang PMK, Tang PCT, Chung JYF, Lan HY. TGF-β1 signaling in kidney disease: From Smads to long non-coding RNAs. Noncoding RNA Res 2017; 2:68-73. [PMID: 30159422 PMCID: PMC6096420 DOI: 10.1016/j.ncrna.2017.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/14/2017] [Accepted: 04/06/2017] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1) has an essential role in the development of kidney diseases. However, targeting TGF-β1 is not a good strategy for fibrotic diseases due to its multifunctional characteristic in physiology. A precise therapeutic target maybe identified by further resolving the underlying TGF-β1 driven mechanisms in renal inflammation and fibrosis. Smad signaling is uncovered as a key pathway of TGF-β1-mediated renal injury, where Smad3 is hyper-activated but Smad7 is suppressed. Mechanistic studies revealed that TGF-β1/Smad3 is capable of promoting renal inflammation and fibrosis via regulating non-coding RNAs. More importantly, involvement of disease- and tissue-specific TGF-β1-dependent long non-coding RNAs (lncRNA) have been recently recognized in a number of kidney diseases. In this review, current understanding of TGF-β1 driven lncRNAs in the pathogenesis of kidney injury, diabetic nephropathy and renal cell carcinoma will be intensively discussed.
Collapse
Affiliation(s)
- Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Philip Chiu-Tsun Tang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jeff Yat-Fai Chung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hui-Yao Lan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
172
|
Zhong B, Zhang C, Guo S, Zhang C. Rational design of cyclic peptides to disrupt TGF-Β/SMAD7 signaling in heterotopic ossification. J Mol Graph Model 2017; 72:25-31. [DOI: 10.1016/j.jmgm.2016.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/17/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022]
|
173
|
Wang YY, Jiang H, Pan J, Huang XR, Wang YC, Huang HF, To KF, Nikolic-Paterson DJ, Lan HY, Chen JH. Macrophage-to-Myofibroblast Transition Contributes to Interstitial Fibrosis in Chronic Renal Allograft Injury. J Am Soc Nephrol 2017; 28:2053-2067. [PMID: 28209809 DOI: 10.1681/asn.2016050573] [Citation(s) in RCA: 304] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 12/14/2016] [Indexed: 01/04/2023] Open
Abstract
Interstitial fibrosis is an important contributor to graft loss in chronic renal allograft injury. Inflammatory macrophages are associated with fibrosis in renal allografts, but how these cells contribute to this damaging response is not clearly understood. Here, we investigated the role of macrophage-to-myofibroblast transition in interstitial fibrosis in human and experimental chronic renal allograft injury. In biopsy specimens from patients with active chronic allograft rejection, we identified cells undergoing macrophage-to-myofibroblast transition by the coexpression of macrophage (CD68) and myofibroblast (α-smooth muscle actin [α-SMA]) markers. CD68+/α-SMA+ cells accounted for approximately 50% of the myofibroblast population, and the number of these cells correlated with allograft function and the severity of interstitial fibrosis. Similarly, in C57BL/6J mice with a BALB/c renal allograft, cells coexpressing macrophage markers (CD68 or F4/80) and α-SMA composed a significant population in the interstitium of allografts undergoing chronic rejection. Fate-mapping in Lyz2-Cre/Rosa26-Tomato mice showed that approximately half of α-SMA+ myofibroblasts in renal allografts originated from recipient bone marrow-derived macrophages. Knockout of Smad3 protected against interstitial fibrosis in renal allografts and substantially reduced the number of macrophage-to-myofibroblast transition cells. Furthermore, the majority of macrophage-to-myofibroblast transition cells in human and experimental renal allograft rejection coexpressed the M2-type macrophage marker CD206, and this expression was considerably reduced in Smad3-knockout recipients. In conclusion, our studies indicate that macrophage-to-myofibroblast transition contributes to interstitial fibrosis in chronic renal allograft injury. Moreover, the transition of bone marrow-derived M2-type macrophages to myofibroblasts in the renal allograft is regulated via a Smad3-dependent mechanism.
Collapse
Affiliation(s)
- Ying-Ying Wang
- Kidney Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and The Chinese University of Hong Kong, Shenzhen Research Institute, and
| | - Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Pan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and The Chinese University of Hong Kong, Shenzhen Research Institute, and.,Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and The Chinese University of Hong Kong, Shenzhen Research Institute, and
| | - Yu-Cheng Wang
- Kidney Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and The Chinese University of Hong Kong, Shenzhen Research Institute, and
| | - Hong-Feng Huang
- Kidney Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China; and
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Center and Monash University Department of Medicine, Clayton, Melbourne, Australia
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, and The Chinese University of Hong Kong, Shenzhen Research Institute, and
| | - Jiang-Hua Chen
- Kidney Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China,
| |
Collapse
|
174
|
Oyarzún C, Garrido W, Alarcón S, Yáñez A, Sobrevia L, Quezada C, San Martín R. Adenosine contribution to normal renal physiology and chronic kidney disease. Mol Aspects Med 2017; 55:75-89. [PMID: 28109856 DOI: 10.1016/j.mam.2017.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 12/12/2022]
Abstract
Adenosine is a nucleoside that is particularly interesting to many scientific and clinical communities as it has important physiological and pathophysiological roles in the kidney. The distribution of adenosine receptors has only recently been elucidated; therefore it is likely that more biological roles of this nucleoside will be unveiled in the near future. Since the discovery of the involvement of adenosine in renal vasoconstriction and regulation of local renin production, further evidence has shown that adenosine signaling is also involved in the tubuloglomerular feedback mechanism, sodium reabsorption and the adaptive response to acute insults, such as ischemia. However, the most interesting finding was the increased adenosine levels in chronic kidney diseases such as diabetic nephropathy and also in non-diabetic animal models of renal fibrosis. When adenosine is chronically increased its signaling via the adenosine receptors may change, switching to a state that induces renal damage and produces phenotypic changes in resident cells. This review discusses the physiological and pathophysiological roles of adenosine and pays special attention to the mechanisms associated with switching homeostatic nucleoside levels to increased adenosine production in kidneys affected by CKD.
Collapse
Affiliation(s)
- Carlos Oyarzún
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Wallys Garrido
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Sebastián Alarcón
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Alejandro Yáñez
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory (CMPL), Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD 4029, Queensland, Australia
| | - Claudia Quezada
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Rody San Martín
- Institute of Biochemistry and Microbiology, Science Faculty, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
175
|
The cellular and signalling alterations conducted by TGF-β contributing to renal fibrosis. Cytokine 2016; 88:115-125. [DOI: 10.1016/j.cyto.2016.08.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 01/08/2023]
|
176
|
Guiteras R, Flaquer M, Cruzado JM. Macrophage in chronic kidney disease. Clin Kidney J 2016; 9:765-771. [PMID: 27994852 PMCID: PMC5162417 DOI: 10.1093/ckj/sfw096] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/22/2016] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) has become a major health problem worldwide. This review describes the role of macrophages in CKD and highlights the importance of anti-inflammatory M2 macrophage activation in both renal fibrosis and wound healing processes. Furthermore, the mechanisms by which M2 macrophages induce renal repair and regeneration are still under debate and currently demand more attention. The M1/M2 macrophage balance is related to the renal microenvironment and could influence CKD progression. In fact, an inflammatory renal environment and M2 plasticity can be the major hurdles to establishing macrophage cell-based therapies in CKD. M2 macrophage cell-based therapy is promising if the M2 phenotype remains stable and is 'fixed' by in vitro manipulation. However, a greater understanding of phenotype polarization is still required. Moreover, better strategies and targets to induce reparative macrophages in vivo should guide future investigations in order to abate kidney diseases.
Collapse
Affiliation(s)
- Roser Guiteras
- Experimental Nephrology, Departament de Ciències Clíniques, Universitat de Barcelona, Institut d'Investigació biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Maria Flaquer
- Experimental Nephrology, Departament de Ciències Clíniques, Universitat de Barcelona, Institut d'Investigació biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M. Cruzado
- Experimental Nephrology, Departament de Ciències Clíniques, Universitat de Barcelona, Institut d'Investigació biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Nephrology Department, Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
177
|
Inflammatory macrophages can transdifferentiate into myofibroblasts during renal fibrosis. Cell Death Dis 2016; 7:e2495. [PMID: 27906172 PMCID: PMC5261004 DOI: 10.1038/cddis.2016.402] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 01/06/2023]
Abstract
Myofibroblasts play a central role in renal fibrosis although the origin of these cells remains controversial. We recently reported that bone marrow-derived macrophages can give rise to myofibroblasts through macrophage to myofibroblast transition (MMT). However, several important issues remain to be addressed, including whether MMT occurs in human kidney disease and verification of the MMT process through lineage tracing. Biopsies from a cohort of 58 patients with various forms of kidney disease were examined for MMT cells that co-express macrophage (CD68) and myofibroblast (α-smooth muscle actin, α-SMA) markers. MMT cells were evident in active fibrotic lesions, but were largely absent in acute inflammatory or sclerotic lesions, suggesting that MMT cells contribute to progressive renal fibrosis. Fate-mapping studies in LysMCreTomato mice identified substantial numbers of Tomato+ myeloid cells with F4/80+ macrophage phenotype expressing α-SMA and collagen I in the unilateral ureteral obstructive model of renal fibrosis, providing direct evidence for the MMT process during the development of renal fibrosis. In addition, MMT cells had a predominant M2 phenotype in both human and mouse renal fibrosis. Finally, selective depletion of myeloid cells via diphtheria toxin in LysMCreiDTR mice largely abolished macrophage infiltration and MMT cells in the obstructed kidney and substantially reduced accumulation of α-SMA+ myofibroblasts and collagen deposition, revealing a pathogenic role for inflammatory macrophages in MMT and tissue fibrosis. In conclusion, these findings provide substantial new data to support the postulate that macrophages can directly transdifferentiate into collagen-producing myofibroblasts in human and experimental kidney disease.
Collapse
|
178
|
Afik R, Zigmond E, Vugman M, Klepfish M, Shimshoni E, Pasmanik-Chor M, Shenoy A, Bassat E, Halpern Z, Geiger T, Sagi I, Varol C. Tumor macrophages are pivotal constructors of tumor collagenous matrix. J Exp Med 2016; 213:2315-2331. [PMID: 27697834 PMCID: PMC5068227 DOI: 10.1084/jem.20151193] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/29/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages are pivotal constructors of the tumoral ECM structure and molecular composition. In particular, they orchestrate the buildup of the tumorigenic collagenous ECM niche. Tumor-associated macrophages (TAMs) promote tumor development, invasion, and dissemination by various mechanisms. In this study, using an orthotopic colorectal cancer (CRC) model, we found that monocyte-derived TAMs advance tumor development by the remodeling of its extracellular matrix (ECM) composition and structure. Unbiased transcriptomic and proteomic analyses of (a) TAM-abundant and -deficient tumor tissues and (b) sorted tumor-associated and -resident colonic macrophage subpopulations defined a distinct TAM-induced ECM molecular signature composed of an ensemble of matricellular proteins and remodeling enzymes they provide to the tumor microenvironment. Remarkably, many of these ECM proteins are specifically increased in human CRC versus healthy colon. Specifically, we demonstrate that although differentiating into TAMs, monocytes up-regulate matrix-remodeling programs associated with the synthesis and assembly of collagenous ECM, specifically collagen types I, VI, and XIV. This finding was further established by advanced imaging showing that TAMs instruct the deposition, cross-linking, and linearization of collagen fibers during tumor development, especially at areas of tumor invasiveness. Finally, we show that cancer-associated fibroblasts are significantly outnumbered by TAMs in this model and that their expression of collagen XIV and I is reduced by TAM deficiency. Here, we outline a novel TAM protumoral function associated with building of the collagenous ECM niche.
Collapse
Affiliation(s)
- Ran Afik
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ehud Zigmond
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Milena Vugman
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Mordehay Klepfish
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elee Shimshoni
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anjana Shenoy
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Elad Bassat
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Zamir Halpern
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Chen Varol
- Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
179
|
Lovisa S, Zeisberg M, Kalluri R. Partial Epithelial-to-Mesenchymal Transition and Other New Mechanisms of Kidney Fibrosis. Trends Endocrinol Metab 2016; 27:681-695. [PMID: 27372267 DOI: 10.1016/j.tem.2016.06.004] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/06/2016] [Accepted: 06/06/2016] [Indexed: 12/24/2022]
Abstract
Kidney fibrosis is the unavoidable consequence of chronic kidney disease irrespective of the primary underlying insult. It is a complex phenomenon governed by the interplay between different cellular components and intricate networks of signaling pathways, which together lead to loss of renal functionality and replacement of kidney parenchyma with scar tissue. An immense effort has recently been made to understand the molecular and cellular mechanisms leading to kidney fibrosis. The cellular protagonists of this process include myofibroblasts, tubular epithelial cells, endothelial cells, and immune cells. We discuss here the most recent findings, including partial epithelial-to-mesenchymal transition (EMT), in the initiation and progression of tissue fibrosis and chronic kidney disease (CKD). A deep understanding of these mechanisms will allow the development of effective therapies.
Collapse
Affiliation(s)
- Sara Lovisa
- Department of Cancer Biology, Metastasis Research Center, University of Texas M.D. Anderson Cancer Center, Houston, TX 77054, USA
| | - Michael Zeisberg
- Department of Nephrology and Rheumatology, Göttingen University Medical Center, Georg August University, Göttingen, Germany
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas M.D. Anderson Cancer Center, Houston, TX 77054, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
180
|
TGF-β1/Smads and miR-21 in Renal Fibrosis and Inflammation. Mediators Inflamm 2016; 2016:8319283. [PMID: 27610006 PMCID: PMC5005604 DOI: 10.1155/2016/8319283] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/15/2016] [Accepted: 07/26/2016] [Indexed: 12/14/2022] Open
Abstract
Renal fibrosis, irrespective of its etiology, is a final common stage of almost all chronic kidney diseases. Increased apoptosis, epithelial-to-mesenchymal transition, and inflammatory cell infiltration characterize the injured kidney. On the molecular level, transforming growth factor-β1 (TGF-β1)-Smad3 signaling pathway plays a central role in fibrotic kidney disease. Recent findings indicate the prominent role of microRNAs, small noncoding RNA molecules that inhibit gene expression through the posttranscriptional repression of their target mRNAs, in different pathologic conditions, including renal pathophysiology. miR-21 was also shown to play a dynamic role in inflammatory responses and in accelerating injury responses to promote organ failure and fibrosis. Understanding the cellular and molecular bases of miR-21 involvement in the pathogenesis of kidney diseases, including inflammatory reaction, could be crucial for their early diagnosis. Moreover, the possibility of influencing miR-21 level by specific antagomirs may be considered as an approach for treatment of renal diseases.
Collapse
|
181
|
Abstract
Transforming growth factor-β (TGF-β) is the primary factor that drives fibrosis in most, if not all, forms of chronic kidney disease (CKD). Inhibition of the TGF-β isoform, TGF-β1, or its downstream signalling pathways substantially limits renal fibrosis in a wide range of disease models whereas overexpression of TGF-β1 induces renal fibrosis. TGF-β1 can induce renal fibrosis via activation of both canonical (Smad-based) and non-canonical (non-Smad-based) signalling pathways, which result in activation of myofibroblasts, excessive production of extracellular matrix (ECM) and inhibition of ECM degradation. The role of Smad proteins in the regulation of fibrosis is complex, with competing profibrotic and antifibrotic actions (including in the regulation of mesenchymal transitioning), and with complex interplay between TGF-β/Smads and other signalling pathways. Studies over the past 5 years have identified additional mechanisms that regulate the action of TGF-β1/Smad signalling in fibrosis, including short and long noncoding RNA molecules and epigenetic modifications of DNA and histone proteins. Although direct targeting of TGF-β1 is unlikely to yield a viable antifibrotic therapy due to the involvement of TGF-β1 in other processes, greater understanding of the various pathways by which TGF-β1 controls fibrosis has identified alternative targets for the development of novel therapeutics to halt this most damaging process in CKD.
Collapse
|
182
|
Yan J, Zhang Z, Jia L, Wang Y. Role of Bone Marrow-Derived Fibroblasts in Renal Fibrosis. Front Physiol 2016; 7:61. [PMID: 26941655 PMCID: PMC4766307 DOI: 10.3389/fphys.2016.00061] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/09/2016] [Indexed: 01/13/2023] Open
Abstract
Renal fibrosis represents a common pathway leading to progression of chronic kidney disease. Renal interstitial fibrosis is characterized by extensive fibroblast activation and excessive production and deposition of extracellular matrix (ECM), which leads to progressive loss of kidney function. There is no effective therapy available clinically to halt or even reverse renal fibrosis. Although activated fibroblasts/myofibroblasts are responsible for the excessive production and deposition of ECM, their origin remains controversial. Recent evidence suggests that bone marrow-derived fibroblast precursors contribute significantly to the pathogenesis of renal fibrosis. Understanding the molecular signaling mechanisms underlying the recruitment and activation of the bone marrow-derived fibroblast precursors will lead to novel therapy for the treatment of chronic kidney disease. In this review, we summarize recent advances in our understanding of the recruitment and activation of bone marrow-derived fibroblast precursors in the kidney and the development of renal fibrosis and highlights new insights that may lead to novel therapies to prevent or reverse the development of renal fibrosis.
Collapse
Affiliation(s)
- Jingyin Yan
- Selzman Institute for Kidney Health and Section of Nephrology, Department of Medicine, Baylor College of Medicine Houston, TX, USA
| | - Zhengmao Zhang
- Selzman Institute for Kidney Health and Section of Nephrology, Department of Medicine, Baylor College of Medicine Houston, TX, USA
| | - Li Jia
- Selzman Institute for Kidney Health and Section of Nephrology, Department of Medicine, Baylor College of Medicine Houston, TX, USA
| | - Yanlin Wang
- Selzman Institute for Kidney Health and Section of Nephrology, Department of Medicine, Baylor College of MedicineHouston, TX, USA; Renal Section, Michael E. DeBakey Veterans Affairs Medical CenterHouston, TX, USA
| |
Collapse
|