1
|
Liao YR, Tsai YC, Hsieh TH, Tsai MT, Lin FY, Lin SJ, Lin CC, Chiang HY, Chu PH, Li SY. FHL2 in arterial medial calcification in chronic kidney disease. Nephrol Dial Transplant 2024; 39:2025-2039. [PMID: 38664060 PMCID: PMC11596093 DOI: 10.1093/ndt/gfae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Arterial medial calcification (AMC) is a common complication in individuals with chronic kidney disease (CKD), which can lead to cardiovascular morbidity and mortality. The progression of AMC is controlled by a key transcription factor called runt-related transcription factor 2 (RUNX2), which induces vascular smooth muscle cells (VSMCs) transdifferentiation into an osteogenic phenotype. However, RUNX2 has not been targeted for therapy due to its essential role in bone development. The objective of our study was to discover a RUNX2 coactivator that is highly expressed in arterial VSMCs as a potential therapy for AMC. METHODS We employed transcriptomic analysis of human data and an animal reporter system to pinpoint four and a half LIM domains 2 (FHL2) as a potential target. Subsequently, we investigated the mRNA and protein expression patterns of FHL2 in the aortas of both human and animal subjects with CKD. To examine the role of FHL2 in the RUNX2 transcription machinery, we conducted coimmunoprecipitation and chromatin immunoprecipitation experiments. Next, we manipulated FHL2 expression in cultured VSMCs to examine its impact on high phosphate-induced transdifferentiation. Finally, we employed FHL2-null mice to confirm the role of FHL2 in the development of AMC in vivo. RESULTS Among all the potential RUNX2 cofactors, FHL2 displays selective expression within the cardiovascular system. In the context of CKD subjects, FHL2 undergoes upregulation and translocation from the cytosol to the nucleus of arterial VSMCs. Once in the nucleus, FHL2 interacts structurally and functionally with RUNX2, acting as a coactivator of RUNX2. Notably, the inhibition of FHL2 expression averts transdifferentiation of VSMCs into an osteogenic phenotype and mitigates aortic calcification in uremic animals, without causing any detrimental effects on the skeletal system. CONCLUSION These observations provide evidence that FHL2 is a promising target for treating arterial calcification in patients with CKD.
Collapse
MESH Headings
- Animals
- LIM-Homeodomain Proteins/metabolism
- LIM-Homeodomain Proteins/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/complications
- Humans
- Mice
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Transcription Factors/metabolism
- Transcription Factors/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/etiology
- Vascular Calcification/genetics
- Male
- Cell Transdifferentiation
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Knockout
Collapse
Affiliation(s)
- Yuan-Ru Liao
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Cheng Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yen Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hou-Yu Chiang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Science, College of Medicine, Chang Guang University, Taoyuan, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taiwan
| | - Szu-Yuan Li
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
2
|
Li A, Wang Y, Wang Y, Xiong Y, Li Y, Liu W, Zhu J, Lin Y. Effects of the FHL2 gene on the development of subcutaneous and intramuscular adipocytes in goats. BMC Genomics 2024; 25:850. [PMID: 39261767 PMCID: PMC11389066 DOI: 10.1186/s12864-024-10755-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Adipose tissue affects not only the meat quality of domestic animals, but also human health. Adipocyte differentiation is regulated by a series of regulatory genes and cyclins. Four and half-LIM protein (FHL2) is positively correlated with the hypertrophy of adipocytes and can cause symptoms such as obesity and diabetes. RESULT In the transcriptome sequencing analysis of intramuscular adipocytes after three days of differentiation, the differentially expressed gene FHL2 was found. To further explore the biological significance of the differentially expressed gene FHL2, which was downregulated in the mature adipocytes. We revealed the function of FHL2 in adipogenesis through the acquisition and loss of function of FHL2. The results showed that the overexpression of FHL2 significantly increased the expression of adipogenic genes (PPARγ, C/EBPβ) and the differentiation of intramuscular and subcutaneous adipocytes. However, silencing FHL2 significantly inhibited adipocyte differentiation. The overexpression of FHL2 increased the number of adipocytes stained with crystal violet and increased the mRNA expression of proliferation marker genes such as CCNE, PCNA, CCND and CDK2. In addition, it significantly increased the rate of EdU positive cells. In terms of apoptosis, overexpression of FHL2 significantly inhibited the expression of P53 and BAX in both intramuscular and subcutaneous adipocytes, which are involved in cell apoptosis. However, overexpression of FHL2 promoted the expression of BCL, but was rescued by the silencing of FHL2. CONCLUSIONS In summary, FHL2 may be a positive regulator of intramuscular and subcutaneous adipocyte differentiation and proliferation, and acts as a negative regulator of intramuscular and subcutaneous adipocyte apoptosis. These findings provide a theoretical basis for the subsequent elucidation of FHL2 in adipocytes.
Collapse
Affiliation(s)
- An Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
- College of Animal & Veterinary Science, Southwest Minzu University, Chengdu, China
| | - Youli Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- College of Animal & Veterinary Science, Southwest Minzu University, Chengdu, China
| | - Yong Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
- College of Animal & Veterinary Science, Southwest Minzu University, Chengdu, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
- College of Animal & Veterinary Science, Southwest Minzu University, Chengdu, China
| | - Yanyan Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
- College of Animal & Veterinary Science, Southwest Minzu University, Chengdu, China
| | - Wei Liu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- College of Animal & Veterinary Science, Southwest Minzu University, Chengdu, China
| | - Jiangjiang Zhu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Yaqiu Lin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China.
| |
Collapse
|
3
|
Wang Y, Kuang Z, Xing X, Qiu Y, Zhang J, Shao D, Huang J, Dai C, He W. Proximal tubular FHL2, a novel downstream target of hypoxia inducible factor 1, is a protector against ischemic acute kidney injury. Cell Mol Life Sci 2024; 81:244. [PMID: 38814462 PMCID: PMC11139843 DOI: 10.1007/s00018-024-05289-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Four-and-a-half LIM domains protein 2 (FHL2) is an adaptor protein that may interact with hypoxia inducible factor 1α (HIF-1α) or β-catenin, two pivotal protective signaling in acute kidney injury (AKI). However, little is known about the regulation and function of FHL2 during AKI. We found that FHL2 was induced in renal tubular cells in patients with acute tubular necrosis and mice model of ischemia-reperfusion injury (IRI). In cultured renal proximal tubular cells (PTCs), hypoxia induced FHL2 expression and promoted the binding of HIF-1 to FHL2 promoter. Compared with control littermates, mice with PTC-specific deletion of FHL2 gene displayed worse renal function, more severe morphologic lesion, more tubular cell death and less cell proliferation, accompanying by downregulation of AQP1 and Na, K-ATPase after IRI. Consistently, loss of FHL2 in PTCs restricted activation of HIF-1 and β-catenin signaling simultaneously, leading to attenuation of glycolysis, upregulation of apoptosis-related proteins and downregulation of proliferation-related proteins during IRI. In vitro, knockdown of FHL2 suppressed hypoxia-induced activation of HIF-1α and β-catenin signaling pathways. Overexpression of FHL2 induced physical interactions between FHL2 and HIF-1α, β-catenin, GSK-3β or p300, and the combination of these interactions favored the stabilization and nuclear translocation of HIF-1α and β-catenin, enhancing their mediated gene transcription. Collectively, these findings identify FHL2 as a direct downstream target gene of HIF-1 signaling and demonstrate that FHL2 could play a critical role in protecting against ischemic AKI by promoting the activation of HIF-1 and β-catenin signaling through the interactions with its multiple protein partners.
Collapse
Affiliation(s)
- Yan Wang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China
| | - Ziwei Kuang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China
| | - Xueqi Xing
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China
| | - Yumei Qiu
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China
| | - Jie Zhang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China
| | - Dandan Shao
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China
| | - Jiaxin Huang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China
| | - Chunsun Dai
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China.
| | - Weichun He
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, Jiangsu, 210003, China.
| |
Collapse
|
4
|
Gisch DL, Brennan M, Lake BB, Basta J, Keller MS, Melo Ferreira R, Akilesh S, Ghag R, Lu C, Cheng YH, Collins KS, Parikh SV, Rovin BH, Robbins L, Stout L, Conklin KY, Diep D, Zhang B, Knoten A, Barwinska D, Asghari M, Sabo AR, Ferkowicz MJ, Sutton TA, Kelly KJ, De Boer IH, Rosas SE, Kiryluk K, Hodgin JB, Alakwaa F, Winfree S, Jefferson N, Türkmen A, Gaut JP, Gehlenborg N, Phillips CL, El-Achkar TM, Dagher PC, Hato T, Zhang K, Himmelfarb J, Kretzler M, Mollah S, Jain S, Rauchman M, Eadon MT. The chromatin landscape of healthy and injured cell types in the human kidney. Nat Commun 2024; 15:433. [PMID: 38199997 PMCID: PMC10781985 DOI: 10.1038/s41467-023-44467-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
There is a need to define regions of gene activation or repression that control human kidney cells in states of health, injury, and repair to understand the molecular pathogenesis of kidney disease and design therapeutic strategies. Comprehensive integration of gene expression with epigenetic features that define regulatory elements remains a significant challenge. We measure dual single nucleus RNA expression and chromatin accessibility, DNA methylation, and H3K27ac, H3K4me1, H3K4me3, and H3K27me3 histone modifications to decipher the chromatin landscape and gene regulation of the kidney in reference and adaptive injury states. We establish a spatially-anchored epigenomic atlas to define the kidney's active, silent, and regulatory accessible chromatin regions across the genome. Using this atlas, we note distinct control of adaptive injury in different epithelial cell types. A proximal tubule cell transcription factor network of ELF3, KLF6, and KLF10 regulates the transition between health and injury, while in thick ascending limb cells this transition is regulated by NR2F1. Further, combined perturbation of ELF3, KLF6, and KLF10 distinguishes two adaptive proximal tubular cell subtypes, one of which manifested a repair trajectory after knockout. This atlas will serve as a foundation to facilitate targeted cell-specific therapeutics by reprogramming gene regulatory networks.
Collapse
Affiliation(s)
- Debora L Gisch
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | - Blue B Lake
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- San Diego Institute of Science, Altos Labs, San Diego, CA, USA
| | - Jeannine Basta
- Washington University in Saint Louis, St. Louis, MO, 63103, USA
| | | | | | | | - Reetika Ghag
- Washington University in Saint Louis, St. Louis, MO, 63103, USA
| | - Charles Lu
- Washington University in Saint Louis, St. Louis, MO, 63103, USA
| | - Ying-Hua Cheng
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | - Samir V Parikh
- Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Brad H Rovin
- Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Lynn Robbins
- St. Louis Veteran Affairs Medical Center, St. Louis, MO, 63106, USA
| | - Lisa Stout
- Washington University in Saint Louis, St. Louis, MO, 63103, USA
| | - Kimberly Y Conklin
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Dinh Diep
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Bo Zhang
- Washington University in Saint Louis, St. Louis, MO, 63103, USA
| | - Amanda Knoten
- Washington University in Saint Louis, St. Louis, MO, 63103, USA
| | - Daria Barwinska
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mahla Asghari
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Angela R Sabo
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | - Timothy A Sutton
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | | | - Sylvia E Rosas
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA
| | | | | | | | - Seth Winfree
- University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nichole Jefferson
- Kidney Precision Medicine Project Community Engagement Committee, Dallas, TX, USA
| | - Aydın Türkmen
- Istanbul School of Medicine, Division of Nephrology, Istanbul, Turkey
| | - Joseph P Gaut
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | - Pierre C Dagher
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Takashi Hato
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kun Zhang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Shamim Mollah
- Washington University in Saint Louis, St. Louis, MO, 63103, USA
| | - Sanjay Jain
- Washington University in Saint Louis, St. Louis, MO, 63103, USA.
| | - Michael Rauchman
- Washington University in Saint Louis, St. Louis, MO, 63103, USA.
| | - Michael T Eadon
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
5
|
Abstract
LIM domain protein 2, also known as LIM protein FHL2, is a member of the LIM-only family. Due to its LIM domain protein characteristics, FHL2 is capable of interacting with various proteins and plays a crucial role in regulating gene expression, cell growth, and signal transduction in muscle and cardiac tissue. In recent years, mounting evidence has indicated that the FHLs protein family is closely associated with the development and occurrence of human tumors. On the one hand, FHL2 acts as a tumor suppressor by down-regulating in tumor tissue and effectively inhibiting tumor development by limiting cell proliferation. On the other hand, FHL2 serves as an oncoprotein by up-regulating in tumor tissue and binding to multiple transcription factors to suppress cell apoptosis, stimulate cell proliferation and migration, and promote tumor progression. Therefore, FHL2 is considered a double-edged sword in tumors with independent and complex functions. This article reviews the role of FHL2 in tumor occurrence and development, discusses FHL2 interaction with other proteins and transcription factors, and its involvement in multiple cell signaling pathways. Finally, the clinical significance of FHL2 as a potential target in tumor therapy is examined.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Changsheng West Road 28, Hengyang, 421001, China
| | - Qun Zeng
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Changsheng West Road 28, Hengyang, 421001, China
| | - Meihua She
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Changsheng West Road 28, Hengyang, 421001, China.
| |
Collapse
|
6
|
Zhang J, She M, Dai Y, Nie X, Tang M, Zeng Q. Lmpt regulates the function of Drosophila muscle by acting as a repressor of Wnt signaling. Gene 2023; 876:147514. [PMID: 37245676 DOI: 10.1016/j.gene.2023.147514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/27/2023] [Accepted: 05/23/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND LIM domain is considered to be important in mediating protein-protein interactions, and members of the LIM protein family can co-regulate tissue-specific gene expression by interacting with different transcription factors. However, its exact function in vivo remains unclear. Our study demonstrates that the LIM protein family member Lmpt may act as a cofactor that interacts with other transcription factors to regulate cellular functions. METHODS In this study, we generated Lmpt knockdown Drosophila (Lmpt-KD) using the UAS-Gal4 system. We assessed the lifespan and motility of Lmpt-KD Drosophila and analyzed the expression of muscle-related and metabolism-related genes using qRT-PCR. Additionally, we utilized Western blot and Top-Flash luciferase reporter assay to evaluate the level of the Wnt signaling pathway. RESULTS Our study revealed that knockdown of the Lmpt gene in Drosophila resulted in a shortened lifespan and reduced motility. We also observed a significant increase in oxidative free radicals in the fly gut. Furthermore, qRT-PCR analysis indicated that knockdown of Lmpt led to decreased expression of muscle-related and metabolism-related genes in Drosophila, suggesting that Lmpt plays a crucial role in maintaining muscle and metabolic functions. Finally, we found that reduction of Lmpt significantly upregulated the expression of Wnt signaling pathway proteins. CONCLUSION Our results demonstrate that Lmpt is essential for motility and survival in Drosophila and acts as a repressor in Wnt signaling.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, 421001 Hengyang, China
| | - Meihua She
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, 421001 Hengyang, China
| | - Yongkang Dai
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, 421001 Hengyang, China
| | - Xiao Nie
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, 421001 Hengyang, China
| | - Min Tang
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, 421001 Hengyang, China
| | - Qun Zeng
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, 421001 Hengyang, China.
| |
Collapse
|
7
|
Gisch DL, Brennan M, Lake BB, Basta J, Keller M, Ferreira RM, Akilesh S, Ghag R, Lu C, Cheng YH, Collins KS, Parikh SV, Rovin BH, Robbins L, Conklin KY, Diep D, Zhang B, Knoten A, Barwinska D, Asghari M, Sabo AR, Ferkowicz MJ, Sutton TA, Kelly KJ, Boer IHD, Rosas SE, Kiryluk K, Hodgin JB, Alakwaa F, Jefferson N, Gaut JP, Gehlenborg N, Phillips CL, El-Achkar TM, Dagher PC, Hato T, Zhang K, Himmelfarb J, Kretzler M, Mollah S, Jain S, Rauchman M, Eadon MT. The chromatin landscape of healthy and injured cell types in the human kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.543965. [PMID: 37333123 PMCID: PMC10274789 DOI: 10.1101/2023.06.07.543965] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
There is a need to define regions of gene activation or repression that control human kidney cells in states of health, injury, and repair to understand the molecular pathogenesis of kidney disease and design therapeutic strategies. However, comprehensive integration of gene expression with epigenetic features that define regulatory elements remains a significant challenge. We measured dual single nucleus RNA expression and chromatin accessibility, DNA methylation, and H3K27ac, H3K4me1, H3K4me3, and H3K27me3 histone modifications to decipher the chromatin landscape and gene regulation of the kidney in reference and adaptive injury states. We established a comprehensive and spatially-anchored epigenomic atlas to define the kidney's active, silent, and regulatory accessible chromatin regions across the genome. Using this atlas, we noted distinct control of adaptive injury in different epithelial cell types. A proximal tubule cell transcription factor network of ELF3 , KLF6 , and KLF10 regulated the transition between health and injury, while in thick ascending limb cells this transition was regulated by NR2F1 . Further, combined perturbation of ELF3 , KLF6 , and KLF10 distinguished two adaptive proximal tubular cell subtypes, one of which manifested a repair trajectory after knockout. This atlas will serve as a foundation to facilitate targeted cell-specific therapeutics by reprogramming gene regulatory networks.
Collapse
|
8
|
Habibe JJ, Boulund U, Clemente-Olivo MP, de Vries CJM, Eringa EC, Nieuwdorp M, Ferwerda B, Zwinderman K, van den Born BJH, Galenkamp H, van Raalte DH. FHL2 Genetic Polymorphisms and Pro-Diabetogenic Lipid Profile in the Multiethnic HELIUS Cohort. Int J Mol Sci 2023; 24:4332. [PMID: 36901761 PMCID: PMC10001862 DOI: 10.3390/ijms24054332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2D) is a prevalent disease often accompanied by the occurrence of dyslipidemia. Four and a half LIM domains 2 (FHL2) is a scaffolding protein, whose involvement in metabolic disease has recently been demonstrated. The association of human FHL2 with T2D and dyslipidemia in a multiethnic setting is unknown. Therefore, we used the large multiethnic Amsterdam-based Healthy Life in an Urban Setting (HELIUS) cohort to investigate FHL2 genetic loci and their potential role in T2D and dyslipidemia. Baseline data of 10,056 participants from the HELIUS study were available for analysis. The HELIUS study contained individuals of European Dutch, South Asian Surinamese, African Surinamese, Ghanaian, Turkish, and Moroccan descent living in Amsterdam and were randomly sampled from the municipality register. Nineteen FHL2 polymorphisms were genotyped, and associations with lipid panels and T2D status were investigated. We observed that seven FHL2 polymorphisms associated nominally with a pro-diabetogenic lipid profile including triglyceride (TG), high-density and low-density lipoprotein-cholesterol (HDL-C and LDL-C), and total cholesterol (TC) concentrations, but not with blood glucose concentrations or T2D status in the complete HELIUS cohort upon correcting for age, gender, BMI, and ancestry. Upon stratifying for ethnicity, we observed that only two of the nominally significant associations passed multiple testing adjustments, namely, the association of rs4640402 with increased TG and rs880427 with decreased HDL-C concentrations in the Ghanaian population. Our results highlight the effect of ethnicity on pro-diabetogenic selected lipid biomarkers within the HELIUS cohort, as well as the need for more large multiethnic cohort studies.
Collapse
Affiliation(s)
- Jayron J. Habibe
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Ulrika Boulund
- Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Maria P. Clemente-Olivo
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Carlie J. M. de Vries
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Etto C. Eringa
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Medical Center, 1081 HV Amsterdam, The Netherlands
- Department of Physiology, Cardiovascular Institute Maastricht, 6229 ER Maastricht, The Netherlands
| | - Max Nieuwdorp
- Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Bart Ferwerda
- Department of Clinical Epidemiology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Koos Zwinderman
- Department of Clinical Epidemiology, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Bert-Jan H. van den Born
- Department of Internal Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Henrike Galenkamp
- Department of Public and Occupational Health, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Daniel H. van Raalte
- Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Internal Medicine, Diabetes Center, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
9
|
Di Camillo B, Puricelli L, Iori E, Toffolo GM, Tessari P, Arrigoni G. Modeling SILAC Data to Assess Protein Turnover in a Cellular Model of Diabetic Nephropathy. Int J Mol Sci 2023; 24:ijms24032811. [PMID: 36769128 PMCID: PMC9917874 DOI: 10.3390/ijms24032811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Protein turnover rate is finely regulated through intracellular mechanisms and signals that are still incompletely understood but that are essential for the correct function of cellular processes. Indeed, a dysfunctional proteostasis often impacts the cell's ability to remove unfolded, misfolded, degraded, non-functional, or damaged proteins. Thus, altered cellular mechanisms controlling protein turnover impinge on the pathophysiology of many diseases, making the study of protein synthesis and degradation rates an important step for a more comprehensive understanding of these pathologies. In this manuscript, we describe the application of a dynamic-SILAC approach to study the turnover rate and the abundance of proteins in a cellular model of diabetic nephropathy. We estimated protein half-lives and relative abundance for thousands of proteins, several of which are characterized by either an altered turnover rate or altered abundance between diabetic nephropathic subjects and diabetic controls. Many of these proteins were previously shown to be related to diabetic complications and represent therefore, possible biomarkers or therapeutic targets. Beside the aspects strictly related to the pathological condition, our data also represent a consistent compendium of protein half-lives in human fibroblasts and a rich source of important information related to basic cell biology.
Collapse
Affiliation(s)
- Barbara Di Camillo
- Department of Information Engineering, University of Padova, 35131 Padova, Italy
- Correspondence: (B.D.C.); (G.A.)
| | - Lucia Puricelli
- Department of Medicine, University of Padova, 35128 Padova, Italy
- Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, 35128 Padova, Italy
| | - Elisabetta Iori
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Gianna Maria Toffolo
- Department of Information Engineering, University of Padova, 35131 Padova, Italy
| | - Paolo Tessari
- Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Giorgio Arrigoni
- Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, 35128 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Correspondence: (B.D.C.); (G.A.)
| |
Collapse
|
10
|
miR-377 Inhibits Proliferation and Differentiation of Bovine Skeletal Muscle Satellite Cells by Targeting FHL2. Genes (Basel) 2022; 13:genes13060947. [PMID: 35741709 PMCID: PMC9223022 DOI: 10.3390/genes13060947] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
Non-coding RNAs, especially microRNAs (miRNAs), play an important role in skeletal muscle growth and development. miR-377 regulates many basic biological processes and plays a key role in tumor cell proliferation, migration and apoptosis. Nevertheless, the function of miR-377 during skeletal muscle development and how it regulates skeletal muscle satellite cells (SMSCs) remains unclear. In the present study, we proposed to elucidate the regulatory mechanism of miR-377 in the proliferation and differentiation of bovine primary SMSCs. Our results showed that miR-377 can significantly inhibit the proliferation of SMSCs. In addition, we found that miR-377 can reduce myotube formation and restrain skeletal myogenic differentiation. Moreover, the results obtained from the biosynthesis and dual luciferase experiments showed that FHL2 was the target gene of miR-377. We further probed the function of FHL2 in muscle development and found that FHL2 silencing significantly suppressed the proliferation and differentiation of SMSCS, which is contrary to the role of miR-377. Furthermore, FHL2 interacts with Dishevelled-2 (Dvl2) to enable Wnt/β-catenin signaling pathway, consequently regulating skeletal muscle development. miR-377 negatively regulates the Wnt/β-catenin signaling pathway by targeting FHL2-mediated Dvl2. Overall, these findings demonstrated that miR-377 regulates the bovine SMSCs proliferation and differentiation by targeting FHL2 and attenuating the Wnt/β-catenin signaling pathway.
Collapse
|
11
|
Zhou D, Wang Y, Gui Y, Fu H, Zhou S, Wang Y, Bastacky SI, Stolz DB, Liu Y. Non-canonical Wnt/calcium signaling is protective against podocyte injury and glomerulosclerosis. Kidney Int 2022; 102:96-107. [PMID: 35341792 DOI: 10.1016/j.kint.2022.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
Activation of canonical Wnt signaling has been implicated in podocyte injury and proteinuria. As Wnts are secreted proteins, whether Wnts derived from podocytes are obligatory for promoting proteinuria remains unknown. To address this, we generated conditional knockout mice where Wntless, a cargo receptor protein required for Wnt secretion, was specifically deleted in glomerular podocytes. Mice with podocyte-specific ablation of Wintless (Podo-Wntless-/-) were phenotypically normal. However, after inducing kidney damage with Adriamycin for six days, Podo-Wntless-/- mice developed more severe podocyte injury and albuminuria than their control littermates. Surprisingly, ablation of Wntless resulted in upregulation of β-catenin, accompanied by reduction of nephrin, podocin, podocalyxin, and Wilms tumor 1 proteins. In chronic injury induced by Adriamycin, increased albuminuria, aggravated podocyte lesions and extracellular matrix deposition were evident in Podo-Wntlessl-/- mice, compared to wild type mice. Mechanistically, specific ablation of Wintless in podocytes caused down-regulation of the nuclear factor of activated T cell 1 (NFAT1) and Nemo-like kinase (NLK), key downstream mediators of non-canonical Wnt/calcium signaling. In vitro, knockdown of either NFAT1 or NLK induced β-catenin activation while overexpression of NLK significantly repressed β-catenin induction and largely preserved nephrin in glomerular podocytes. Thus, our results indicate that podocyte-derived Wnts play an important role in protecting podocytes from injury by repressing β-catenin via activating non-canonical Wnt/calcium signaling.
Collapse
Affiliation(s)
- Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Yuanyuan Wang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Yuan Gui
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Haiyan Fu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shanshan Zhou
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Sheldon I Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Donna B Stolz
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Habibe JJ, Clemente-Olivo MP, de Vries CJ. How (Epi)Genetic Regulation of the LIM-Domain Protein FHL2 Impacts Multifactorial Disease. Cells 2021; 10:2611. [PMID: 34685595 PMCID: PMC8534169 DOI: 10.3390/cells10102611] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/13/2023] Open
Abstract
Susceptibility to complex pathological conditions such as obesity, type 2 diabetes and cardiovascular disease is highly variable among individuals and arises from specific changes in gene expression in combination with external factors. The regulation of gene expression is determined by genetic variation (SNPs) and epigenetic marks that are influenced by environmental factors. Aging is a major risk factor for many multifactorial diseases and is increasingly associated with changes in DNA methylation, leading to differences in gene expression. Four and a half LIM domains 2 (FHL2) is a key regulator of intracellular signal transduction pathways and the FHL2 gene is consistently found as one of the top hyper-methylated genes upon aging. Remarkably, FHL2 expression increases with methylation. This was demonstrated in relevant metabolic tissues: white adipose tissue, pancreatic β-cells, and skeletal muscle. In this review, we provide an overview of the current knowledge on regulation of FHL2 by genetic variation and epigenetic DNA modification, and the potential consequences for age-related complex multifactorial diseases.
Collapse
Affiliation(s)
- Jayron J. Habibe
- Department of Medical Biochemistry, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, 1105 AZ Amsterdam, The Netherlands; (J.J.H.); (M.P.C.-O.)
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands
| | - Maria P. Clemente-Olivo
- Department of Medical Biochemistry, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, 1105 AZ Amsterdam, The Netherlands; (J.J.H.); (M.P.C.-O.)
| | - Carlie J. de Vries
- Department of Medical Biochemistry, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, 1105 AZ Amsterdam, The Netherlands; (J.J.H.); (M.P.C.-O.)
| |
Collapse
|
13
|
Chen Q, Gao C, Wang M, Fei X, Zhao N. TRIM18-Regulated STAT3 Signaling Pathway via PTP1B Promotes Renal Epithelial-Mesenchymal Transition, Inflammation, and Fibrosis in Diabetic Kidney Disease. Front Physiol 2021; 12:709506. [PMID: 34434118 PMCID: PMC8381599 DOI: 10.3389/fphys.2021.709506] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/15/2021] [Indexed: 01/15/2023] Open
Abstract
Diabetic kidney disease (DKD) has become a key cause of end-stage renal disease worldwide. Inflammation and fibrosis have been shown to play important roles in the pathogenesis of DKD. MID1, also known as TRIM18, is an E3 ubiquitin ligase of the tripartite motif (TRIM) subfamily of RING-containing proteins and increased in renal tubule in patients with DKD. However, the function and molecular mechanism of TRIM18 in DKD remain unexplored. Herein we report that TRIM18 expression levels were increased in patients with DKD. An animal study confirms that TRIM18 is involved in kidney injury and fibrosis in diabetic mice. TRIM18 knockdown inhibits high glucose (HG)-induced epithelial–mesenchymal transition (EMT), inflammation, and fibrosis of HK-2 cells. This is accompanied by decreased levels of tumor necrosis factor alpha, interleukin-6, hydroxyproline (Hyp), connective tissue growth factor, and α-smooth muscle actin. Additionally, TRIM18 knockdown inhibits HG-induced increase in the phosphorylated-/total signal transducer and activator of transcription (STAT3). Treatment with niclosamide (STAT3 inhibitor) or protein tyrosine phosphatase-1B (PTP1B) overexpression blocked the TRIM18 induced EMT, inflammation and fibrosis. Co-immunoprecipitation and Western blot assays showed that TRIM18 promoted the ubiquitination of PTP1B. These findings highlight the importance of the TRIM18/PTP1B/STAT3 signaling pathway in DKD and can help in the development of new therapeutics for DKD treatment.
Collapse
Affiliation(s)
- Qi Chen
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chan Gao
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Wang
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Fei
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Zhao
- Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
14
|
Miranda MZ, Lichner Z, Szászi K, Kapus A. MRTF: Basic Biology and Role in Kidney Disease. Int J Mol Sci 2021; 22:ijms22116040. [PMID: 34204945 PMCID: PMC8199744 DOI: 10.3390/ijms22116040] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/21/2021] [Accepted: 05/30/2021] [Indexed: 12/23/2022] Open
Abstract
A lesser known but crucially important downstream effect of Rho family GTPases is the regulation of gene expression. This major role is mediated via the cytoskeleton, the organization of which dictates the nucleocytoplasmic shuttling of a set of transcription factors. Central among these is myocardin-related transcription factor (MRTF), which upon actin polymerization translocates to the nucleus and binds to its cognate partner, serum response factor (SRF). The MRTF/SRF complex then drives a large cohort of genes involved in cytoskeleton remodeling, contractility, extracellular matrix organization and many other processes. Accordingly, MRTF, activated by a variety of mechanical and chemical stimuli, affects a plethora of functions with physiological and pathological relevance. These include cell motility, development, metabolism and thus metastasis formation, inflammatory responses and—predominantly-organ fibrosis. The aim of this review is twofold: to provide an up-to-date summary about the basic biology and regulation of this versatile transcriptional coactivator; and to highlight its principal involvement in the pathobiology of kidney disease. Acting through both direct transcriptional and epigenetic mechanisms, MRTF plays a key (yet not fully appreciated) role in the induction of a profibrotic epithelial phenotype (PEP) as well as in fibroblast-myofibroblast transition, prime pathomechanisms in chronic kidney disease and renal fibrosis.
Collapse
Affiliation(s)
- Maria Zena Miranda
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
| | - Zsuzsanna Lichner
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| |
Collapse
|
15
|
Lu HC, Dai WN, He LY. Epigenetic Histone Modifications in the Pathogenesis of Diabetic Kidney Disease. Diabetes Metab Syndr Obes 2021; 14:329-344. [PMID: 33519221 PMCID: PMC7837569 DOI: 10.2147/dmso.s288500] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD), as the main complication of diabetes mellitus, is the primary cause of the end-stage renal disease (ESRD) and the most common chronic kidney disease. Overall, 30-40% of patients with type 1 and type 2 diabetes eventually develop DKD. Although some diabetes patients have intensified glycemic control, they still develop diabetic kidney disease. Current treatment methods can alleviate but do not markedly halt disease development, resulting in renal failure and severe complications, even contributing to elevated morbidity and mortality rates. DKD is a disease with interactions of genes and the environment. Emerging evidence indicates that DKD-associated key genes are also regulated by the epigenetic mechanism. Recently, increasing researches involving cells and experimental animals demonstrated that histone post-translational modifications can mediate gene expression, which correlated with diabetic kidney disease. Novel therapeutic strategies for epigenetic events could be beneficial for the early detection and treatment of DKD to prevent it from developing into end-stage renal disease (ESRD). In this review, we discuss prior findings in the field of histone modifications in DKD, especially histone acetylation and histone methylation. We then focus on recent developments in histone acetylation and methylation involved in the pathogenesis of DKD.
Collapse
Affiliation(s)
- Heng-Cheng Lu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, People’s Republic of China
| | - Wen-Ni Dai
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, People’s Republic of China
| | - Li-Yu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, People’s Republic of China
- Correspondence: Li-Yu He Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, Hunan, People’s Republic of ChinaTel +8673185292064Fax +8673185295843 Email
| |
Collapse
|
16
|
WNT-β-catenin signalling - a versatile player in kidney injury and repair. Nat Rev Nephrol 2020; 17:172-184. [PMID: 32989282 DOI: 10.1038/s41581-020-00343-w] [Citation(s) in RCA: 235] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
Abstract
The WNT-β-catenin system is an evolutionary conserved signalling pathway that is of particular importance for morphogenesis and cell organization during embryogenesis. The system is usually suppressed in adulthood; however, it can be re-activated in organ injury and regeneration. WNT-deficient mice display severe kidney defects at birth. Transient WNT-β-catenin activation stimulates tissue regeneration after acute kidney injury, whereas sustained (uncontrolled) WNT-β-catenin signalling promotes kidney fibrosis in chronic kidney disease (CKD), podocyte injury and proteinuria, persistent tissue damage during acute kidney injury and cystic kidney diseases. Additionally, WNT-β-catenin signalling is involved in CKD-associated vascular calcification and mineral bone disease. The WNT-β-catenin pathway is tightly regulated, for example, by proteins of the Dickkopf (DKK) family. In particular, DKK3 is released by 'stressed' tubular epithelial cells; DKK3 drives kidney fibrosis and is associated with short-term risk of CKD progression and acute kidney injury. Thus, targeting the WNT-β-catenin pathway might represent a promising therapeutic strategy in kidney injury and associated complications.
Collapse
|
17
|
Xu F, Ye Z, Tao S, Liu W, Su J, Fang X, Wang X. Ligustilide alleviates podocyte injury via suppressing the SIRT1/NF-κB signaling pathways in rats with diabetic nephropathy. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1154. [PMID: 33241003 PMCID: PMC7576076 DOI: 10.21037/atm-20-5811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Diabetic nephropathy (DN) is one of the common chronic microvascular complications of diabetes, and podocyte injury and dysfunction are strictly related to the pathogenesis of DN. Studies have shown that ligustilide (LIG) has anti-inflammatory, antioxidant, and anti-apoptotic activities. This study was designed to investigate the therapeutic effect of LIG in DN rats and their mechanisms. Methods DN rat models (n=10) were induced by streptozotocin (STZ) combined with a high-fat diet. Rats in the LIG group were intragastrically administered with LIG daily for eight weeks, and animals in the positive control group were treated with Losartan potassium. The body weight and blood glucose were checked weekly during the treatment. The pathological changes of kidney tissue were observed with hematoxylin and eosin (HE) staining. Blood lipid profiles and renal function-related markers, including total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), blood urea nitrogen (BUN), and serum creatinine (Scr) were monitored using a biochemical analyzer. The protein expression of nephrin was determined by immunohistochemistry and Western blotting. Finally, Western blot was used to determine the protein expression of Sirtuin 1 (SIRT1) and nuclear factor-kappa B (NF-κB). Results Compared with the healthy control group, rats in the DN group have slower weight gain, increased blood sugar level, renal lesions, and impaired renal function, along with decreased nephrin expression, abnormally activated NF-κB, and inhibited SIRT1 protein expression. All the above conditions were improved after intervention with either losartan potassium or LIG. Conclusions LIG attenuates podocyte injury by regulating the SIRT1/NF-κB signaling pathway and thereby exerts its protective effect on renal function in DN rats.
Collapse
Affiliation(s)
- Feng Xu
- Department of Endocrinology and Neurosurgery, the First People's Hospital of Nantong, Nantong, China
| | - Zi Ye
- Department of Endocrinology and Neurosurgery, the First People's Hospital of Nantong, Nantong, China
| | - Shuo Tao
- Department of Nephrology, the First People's Hospital of Nantong, Nantong, China
| | - Wangshu Liu
- Department of Endocrinology, the First People's Hospital of Nantong, Nantong, China
| | - Jianbing Su
- Department of Endocrinology, the First People's Hospital of Nantong, Nantong, China
| | - Xingxing Fang
- Department of Endocrinology, the First People's Hospital of Nantong, Nantong, China
| | - Xueqin Wang
- Department of Endocrinology, the First People's Hospital of Nantong, Nantong, China
| |
Collapse
|
18
|
Duan Y, Qiu Y, Huang X, Dai C, Yang J, He W. Deletion of FHL2 in fibroblasts attenuates fibroblasts activation and kidney fibrosis via restraining TGF-β1-induced Wnt/β-catenin signaling. J Mol Med (Berl) 2020; 98:291-307. [PMID: 31927599 DOI: 10.1007/s00109-019-01870-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/25/2022]
Abstract
Four-and-a-half LIM domains protein 2 (FHL2) has been proposed involving in β-catenin activity. We previously reported that FHL2 mediates TGF-β1-induced tubular epithelial-to-mesenchymal transition through activating Wnt/β-catenin signaling. However, the potential role and mechanism for FHL2 in TGF-β1-induced fibroblast activation and kidney fibrosis remains unknown. Here, we initially observed higher levels of FHL2 expression in fibrotic kidneys from both patients and mice, especially in α-smooth muscle actin (α-SMA)-positive cells in the interstitium. In cultured interstitial fibroblasts, FHL2 expression was induced by TGF-β1. Knockdown of FHL2 remarkably suppressed TGF-β1-induced α-SMA, type I collagen, and fibronectin expression, while overexpression of FHL2 was sufficient to activate fibroblasts. In mice, fibroblast-specific deletion of FHL2 diminished renal induction of α-SMA, type I collagen, and fibronectin and interstitial extracellular matrix deposition at 2 weeks after ureteral obstruction. We next investigated Wnt/β-catenin activity and found that β-catenin was activated in most FHL2-positive cells in renal interstitium from mice with obstructive nephropathy. In vitro, TGF-β1 induced a physical interaction between FHL2 and β-catenin, especially in the nucleus. Downregulation of FHL2 inhibited TGF-β1-induced active β-catenin upregulation, β-catenin nuclear translocation, and β-catenin-mediated transcription, whereas overexpression of FHL2 was able to activate Wnt/β-catenin signaling. FHL2 overexpression-induced β-catenin-mediated gene transcription could be hindered by ICG-001, but FHL2 overexpression-induced upregulation of active β-catenin could not be. Collectively, this study reveals that the signal regulatory effect of FHL2 on β-catenin plays an important role in TGF-β1-induced fibroblast activation and kidney fibrosis.
Collapse
Affiliation(s)
- Ying Duan
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210003, Jiangsu, China.,Department of Blood Purification Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Yumei Qiu
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210003, Jiangsu, China
| | - Xiaowen Huang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210003, Jiangsu, China
| | - Chunsun Dai
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210003, Jiangsu, China
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210003, Jiangsu, China
| | - Weichun He
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210003, Jiangsu, China.
| |
Collapse
|
19
|
Tang YL, Dong XY, Zeng ZG, Feng Z. Gene expression-based analysis identified NTNG1 and HGF as biomarkers for diabetic kidney disease. Medicine (Baltimore) 2020; 99:e18596. [PMID: 31895808 PMCID: PMC6946191 DOI: 10.1097/md.0000000000018596] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease. Because the molecular mechanisms of DKD are not fully understood, exploration of hub genes and the mechanisms underlying this disease are essential for elucidating the pathogenesis and progression of DKD. Accordingly, in this study, we performed an analysis of gene expression in DKD. The differentially expressed genes (DEGs) included 39 upregulated genes and 113 downregulated genes in the GSE30528 dataset and 127 upregulated genes and 18 downregulated genes in the GSE30529 dataset. Additionally, functional analyses were performed to determine the roles of DEGs using glomeruli samples from patients with DKD and healthy controls from the GSE30528 dataset and using tubule samples from patients with DKD and healthy controls from the GSE30529 dataset. These DEGs were enriched in pathways such as the Wnt signaling pathway, metabolic pathways, and the mammalian target of rapamycin signaling pathway in the GSE30528 dataset and the longevity regulating pathway and Ras signaling pathway in the GSE30529 dataset. Moreover, a protein-protein interaction network was constructed using the identified DEGs, and hub gene analysis was performed. Furthermore, correlation analyses between key genes and pathological characteristics of DKD indicated that CCR4, NTNG1, HGF and ISL1 are related to DKD, and NTNG1 and HGF may server as diagnostic biomarkers in DKD using the receiver-operator characteristic (ROC) curve. Collectively, our findings established 2 reliable biomarkers for DKD.
Collapse
Affiliation(s)
| | | | - Zhen-Guo Zeng
- Department of Critical Care Medicine, First Affiliated Hospital of Nanchang University, Nanchang, PR China
| | - Zhen Feng
- Department of Rehabilitation Medicine
| |
Collapse
|
20
|
Fu H, Liu S, Bastacky SI, Wang X, Tian XJ, Zhou D. Diabetic kidney diseases revisited: A new perspective for a new era. Mol Metab 2019; 30:250-263. [PMID: 31767176 PMCID: PMC6838932 DOI: 10.1016/j.molmet.2019.10.005] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/08/2019] [Accepted: 10/13/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Globally, diabetic kidney disease (DKD) is the leading cause of end-stage renal disease. As the most common microvascular complication of diabetes, DKD is a thorny, clinical problem in terms of its diagnosis and management. Intensive glucose control in DKD could slow down but not significantly halt disease progression. Revisiting the tremendous advances that have occurred in the field would enhance recognition of DKD pathogenesis as well as improve our understanding of translational science in DKD in this new era. SCOPE OF REVIEW In this review, we summarize advances in the understanding of the local microenvironmental changes in diabetic kidneys and discuss the involvement of genetic and epigenetic factors in the pathogenesis of DKD. We also review DKD prevalence changes and analyze the challenges in optimizing the diagnostic approaches and management strategies for DKD in the clinic. As we enter the era of 'big data', we also explore the possibility of linking systems biology with translational medicine in DKD in the current healthcare system. MAJOR CONCLUSION Newer understanding of the structural changes of diabetic kidneys and mechanisms of DKD pathogenesis, as well as emergent research technologies will shed light on new methods of dealing with the existing clinical challenges of DKD.
Collapse
Affiliation(s)
- Haiyan Fu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Silvia Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sheldon I Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaojie Wang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiao-Jun Tian
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Dong Zhou
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Shi J, Xu W, Zheng R, Miao H, Hu Q. Neuregulin 4 attenuate tubulointerstitial fibrosis and advanced glycosylation end products accumulation in diabetic nephropathy rats via regulating TNF-R1 signaling. Am J Transl Res 2019; 11:5501-5513. [PMID: 31632525 PMCID: PMC6789242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 07/26/2019] [Indexed: 06/10/2023]
Abstract
Neuregulin 4 (Nrg4) play important roles in the pathogenesis of obesity-associated disorders, including type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD). This study aims to investigate roles of Nrg4 in the pathophysiologic mechanism underlying the progression of tubulointerstitial fibrosis (TIF) in diabetic nephropathy (DN). In present study, Nrg4 is under-expression in serum and renal tissue of diabetic nephropathy rats. In present study, Nrg4 attenuate renal function injury, tubulointerstitial fibrosis, inflammation and suppress the expression levels of advanced glycosylation end products (AGEs) in vivo and vitro. Furthermore, the results reveal that Nrg4 ameliorates fibrosis and attenuate the expression of AGEs and inflammation via TNF-R1 signaling instead of TNF-R2 signaling in HK-2 cells. In conclusion, these results revealed that Nrg4 may effectively ameliorates TIF and attenuate the expression of AGEs in DN through TNF-R1 signaling instead of TNF-R2 signaling. We have provided evidence indicating that Nrg4 possesses therapeutic effect on TIF in DN.
Collapse
Affiliation(s)
- Jinbao Shi
- Department of Traditional Chinese Medicine, Min Dong Hospital of Ningde CityNingde, Fujian Province, China
| | - Weilong Xu
- Department of Endocrinology, Affiliated Hospital of Nanjing University of Chinese MedicineNanjing, Jiangsu Province, China
| | - Ruiping Zheng
- Department of Traditional Chinese Medicine, Min Dong Hospital of Ningde CityNingde, Fujian Province, China
| | - Huizi Miao
- Department of Traditional Chinese Medicine, Min Dong Hospital of Ningde CityNingde, Fujian Province, China
| | - Qiaosheng Hu
- Department of Endocrinology, Lianshui County People’s HospitalLianshui, Jiangsu Province, China
| |
Collapse
|
22
|
Xie S, Ge F, Yao Y, Zhang W, Wang S, Zhang M, Zhong R, Fang L, Qu D. The aqueous extract of Lycopus lucidus Turcz exerts protective effects on podocytes injury of diabetic nephropathy via inhibiting TGF-β1 signal pathway. Am J Transl Res 2019; 11:5689-5702. [PMID: 31632540 PMCID: PMC6789289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 07/26/2019] [Indexed: 06/10/2023]
Abstract
Diabetic nephropathy (DN) is known as a major microvascular complication leading cause of end-stage renal disease, it generally followed by the process of podocyte fragmentation and detachment. Transforming growth factor β1 (TGF-β1) signaling pathway plays a pivotal role in the initiation and progression of DN. In present study, we aim to investigate the effect of lycopus extracts on podocytes injury and TGF-β signaling. In present study, lycopus extracts treatment abolished the gain in blood glucose and body weight in a dose dependent manner and possessed protective effect on the renal damage, which was indicated by the decreased concentration of Scr, BUN and urine creatinine of serum. Histopathological examination also demonstrated lycopus extracts exert protective effect on renal damage. Western blotting and immunohistochemical results revealed lycopus extracts treatment upregulated the expression of nephrin and down-regulated the expression levels of TGF-β1 and Smad4. Moreover, lycopus extracts treatment suppressed TGF-β1-induced phosphorylation of Smad2/3, ERK1/2 and p38 both in vivo and vitro. In conclusion, lycopus extracts is a novel agent that ameliorate podocytes injury by inhibiting TGF-β signaling pathway and possess potential therapeutic effect on renal damage of DN rats.
Collapse
Affiliation(s)
- Shengfang Xie
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Department of Nephrology, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Fengfeng Ge
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Department of Nephrology, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Yuanzhang Yao
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Department of Nephrology, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Wei Zhang
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Shuopeng Wang
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Department of Nephrology, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Min Zhang
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Department of Nephrology, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Rongling Zhong
- Animal Experiment Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Animal Experiment Center, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Liming Fang
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Department of Nephrology, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| | - Ding Qu
- Research Center for Multicomponent Traditional Medicine and Microecology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese MedicineNanjing 210028, Jiangsu, China
- Research Center for Multicomponent Traditional Medicine and Microecology, Jiangsu Province Academy of Traditional Chinese MedicineNanjing 210028, Jiangsu, China
| |
Collapse
|
23
|
Ling C, Rönn T. Epigenetics in Human Obesity and Type 2 Diabetes. Cell Metab 2019; 29:1028-1044. [PMID: 30982733 PMCID: PMC6509280 DOI: 10.1016/j.cmet.2019.03.009] [Citation(s) in RCA: 509] [Impact Index Per Article: 84.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/05/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Epigenetic mechanisms control gene activity and the development of an organism. The epigenome includes DNA methylation, histone modifications, and RNA-mediated processes, and disruption of this balance may cause several pathologies and contribute to obesity and type 2 diabetes (T2D). This Review summarizes epigenetic signatures obtained from human tissues of relevance for metabolism-i.e., adipose tissue, skeletal muscle, pancreatic islets, liver, and blood-in relation to obesity and T2D. Although this research field is still young, these comprehensive data support not only a role for epigenetics in disease development, but also epigenetic alterations as a response to disease. Genetic predisposition, as well as aging, contribute to epigenetic variability, and several environmental factors, including exercise and diet, further interact with the human epigenome. The reversible nature of epigenetic modifications holds promise for future therapeutic strategies in obesity and T2D.
Collapse
Affiliation(s)
- Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden.
| | - Tina Rönn
- Epigenetics and Diabetes Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| |
Collapse
|
24
|
FHL2 mediates podocyte Rac1 activation and foot process effacement in hypertensive nephropathy. Sci Rep 2019; 9:6693. [PMID: 31040292 PMCID: PMC6491468 DOI: 10.1038/s41598-019-42328-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 03/06/2019] [Indexed: 01/04/2023] Open
Abstract
RAAS inhibition has been the standard treatment for CKD for years because it can reduce proteinuria and hence retard renal function decline, but the proteinuria reduction effect is still insufficient in many patients. Podocyte foot process and slit diaphragm are the final barrier to prevent serum proteins leak into urine, and podocyte foot process effacement is the common pathway of all proteinruic diseases. Cell structure are regulated by three evolutionarily conserved Rho GTPases, notably, Rac1 activation is sufficient and necessary for podocyte foot process effacement, however, Rac1 inhibition is not an option for kidney disease treatment because of its systemic side effects. Four-and-a-half LIM domains protein 2 (FHL2) is highly expressed in podocytes and has been implicated in regulating diverse biological functions. Here, we used micro-dissected human kidney samples, in vitro podocyte culture experiments, and a hypertension animal model to determine the possible role of FHL2 in hypertensive nephropathy. FHL2 was abundantly upregulated in hypertensive human glomeruli and animal kidney samples. Genetic deletion of the FHL2 did not alter normal renal structure or function but mitigated hypertension-induced podocyte foot process effacement and albuminuria. Mechanistically, angiotensin II-induced podocyte cytoskeleton reorganization via FAK-Rac1 axis, FHL2 binds with FAK and is an important mediator of Ang II induced Rac1 activation, thus, FHL2 inhibition can selectively block FAK-Rac1 axis in podocyte and prevent proteinuria. These results provide important insights into the mechanisms of podocyte foot process effacement and points out a promising strategy to treat kidney disease.
Collapse
|
25
|
Huang X, Xue H, Ma J, Zhang Y, Zhang J, Liu Y, Qin X, Sun C. Salidroside ameliorates Adriamycin nephropathy in mice by inhibiting β-catenin activity. J Cell Mol Med 2019; 23:4443-4453. [PMID: 30993911 PMCID: PMC6533469 DOI: 10.1111/jcmm.14340] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/21/2019] [Accepted: 04/01/2019] [Indexed: 01/08/2023] Open
Abstract
Salidroside is a major phenylethanoid glycoside in Rhodiola rosea L., a traditional Chinese medicine, with multiple biological activities. It has been shown that salidroside possesses protective effects for alleviating diabetic renal dysfunction, contrast‐induced‐nephropathy and other kidney diseases. However, the involved molecular mechanism was still not understood well. Herein, we examined the protective effects of salidroside in mice with Adriamycin (ADR)‐induced nephropathy and the underlying molecular mechanism. The results showed that salidroside treatment ameliorates proteinuria; improves expressions of nephrin and podocin; and reduces kidney fibrosis and glomerulosclerosis induced by ADR. Mechanistically, ADR induces a robust accumulation of β‐catenin in the nucleus and stimulates its downstream target gene expression. The application of salidroside largely abolishes the nuclear translocation of β‐catenin and thus inhibits its activity. Furthermore, the activation of β‐catenin almost completely counteracts the protective roles of salidroside in ADR‐injured podocytes. Taken together, our data indicate that salidroside ameliorates proteinuria, renal fibrosis and podocyte injury in ADR nephropathy, which may rely on inhibition of β‐catenin signalling pathway.
Collapse
Affiliation(s)
- Xinzhong Huang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Xue
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jinyu Ma
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China
| | | | - Jing Zhang
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China
| | - Yue Liu
- Department of Nephrology, Traditional Chinese Medicine Hospital of Tongzhou District, Nantong, China
| | - Xiaogang Qin
- Department of Nephrology, Traditional Chinese Medicine Hospital of Tongzhou District, Nantong, China
| | - Cheng Sun
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China
| |
Collapse
|
26
|
Li Y, Zhou H, Li Y, Han L, Song M, Chen F, Shang G, Wang D, Wang Z, Zhang W, Zhong M. PTPN2 improved renal injury and fibrosis by suppressing STAT-induced inflammation in early diabetic nephropathy. J Cell Mol Med 2019; 23:4179-4195. [PMID: 30955247 PMCID: PMC6533506 DOI: 10.1111/jcmm.14304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 02/20/2019] [Accepted: 03/14/2019] [Indexed: 12/11/2022] Open
Abstract
Diabetic nephropathy (DN) is a chronic inflammatory disease triggered by disordered metabolism. Recent studies suggested that protein tyrosine phosphatase non‐receptor type 2 (PTPN2) could ameliorate metabolic disorders and suppress inflammatory responses. This study investigated PTPN2's role in modulating DN and the possible cellular mechanisms involved. In a mouse model combining hyperglycaemia and hypercholesterolaemia (streptozotocin diabetic, ApoE‐/‐ mice), mice showed severe insulin resistance, renal dysfunction, micro‐inflammation, subsequent extracellular matrix expansion and decreased expression of PTPN2. We found that mice treated with PTPN2 displayed reduced serum creatinine, serum BUN and proteinuria. PTPN2 gene therapy markedly attenuated metabolic disorders and hyperglycaemia. In addition, PTPN2 gene transfer significantly suppressed renal activation of signal transducers and activators of transcription (STAT), STAT‐dependent pro‐inflammatory and pro‐fibrotic genes expression, and influx of lymphocytes in DN, indicating anti‐inflammatory effects of PTPN2 by inhibiting the activation of STAT signalling pathway in vivo. Furthermore, PTPN2 overexpression inhibited the high‐glucose induced phosphorylation of STAT, target genes expression and proliferation in mouse mesangial and tubuloepithelial cells, suggesting that the roles of PTPN2 on STAT activation was independent of glycaemic changes. Our results demonstrated that PTPN2 gene therapy could exert protective effects on DN via ameliorating metabolic disorders and inhibiting renal STAT‐dependent micro‐inflammation, suggesting its potential role for treatment of human DN.
Collapse
Affiliation(s)
- Ya Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Huimin Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yulin Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lu Han
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming Song
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fangfang Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Guokai Shang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Di Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhihao Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.,Department of Geriatric Medicine, Qilu Hospital of Shandong University, Key Laboratory of Cardiovascular Proteomics of Shandong Province, Ji'nan, China
| | - Wei Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming Zhong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
27
|
Guo Q, Zhong W, Duan A, Sun G, Cui W, Zhuang X, Liu L. Protective or deleterious role of Wnt/beta-catenin signaling in diabetic nephropathy: An unresolved issue. Pharmacol Res 2019; 144:151-157. [PMID: 30935943 DOI: 10.1016/j.phrs.2019.03.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/26/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
Abstract
In recent years, the Wnt/β-catenin signaling has gained tremendous attention due to its ability to modulate a number of diseases including diabetic nephropathy. Studies have shown that there is decrease in the secretion of Wnt proteins including Wnt4, 5a and Wnt 6 during high glucose concentration or diabetic conditions, which leads to decreased translocation of β-catenin to nucleus. The down-regulation of Wnt/β-catenin signaling leads to detrimental effects on kidney including increased apoptosis of mesangial cells and increased deposition of fibrous tissue in mesangium. The pharmacological modulators such as spironolactone, NO donor and antioxidant are shown to produce beneficial effects in diabetic nephropathy by up regulating the expression of Wnt proteins and activation of diabetes-induced suppressed Wnt/β-catenin signaling. On the other hand, it is documented that diabetes leads to overactivation of Wnt1/β-catenin signaling, which promotes podocyte injury, induce epithelial-mesenchymal transition of podocytes along with renal injury and fibrosis. Accordingly, different interventions aimed to suppress overactivated Wnt/β-catenin signaling are reported to improve the condition and symptoms associated with diabetic nephropathy. The present review discusses the dual role of Wnt/beta-catenin signaling in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Qiaoyan Guo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Wei Zhong
- Department of Ophthalmology, The China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Aosong Duan
- Department of Intensive Care Unit, The First Hospital of Jilin University, Changchun, 130021,China.
| | - Guanggong Sun
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Wenpeng Cui
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Xiaohua Zhuang
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Lihua Liu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
28
|
Lu W, Yu T, Liu S, Li S, Li S, Liu J, Xu Y, Xing H, Tian Z, Tang K, Rao Q, Wang J, Wang M. FHL2 interacts with iASPP and impacts the biological functions of leukemia cells. Oncotarget 2018; 8:40885-40895. [PMID: 28402264 PMCID: PMC5522200 DOI: 10.18632/oncotarget.16617] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/09/2017] [Indexed: 01/29/2023] Open
Abstract
iASPP is an inhibitory member of apoptosis-stimulating proteins of p53 (ASPP) family, which inhibits p53-dependent apoptosis. iASPP was highly expressed in acute leukemia, inhibited leukemia cells apoptosis and promoted leukemogenesis. In order to clarify its mechanism, a yeast two-hybrid screen was performed and FHL2 was identified for the first time as one of the binding proteins of iASPP. FHL2 was highly expressed in K562 and Kasumi-1 cells. FHL2 and iASPP interacted with each other and co-localized in both nucleus and cytoplasm. Either FHL2 or iASPP silenced could reduce cell proliferation, induce cell cycle arrest at G0/G1 phase, and increase cell apoptosis. Western blot analysis showed that the level of p21 and p27 increased, CDK4, E2F1, Cyclin E and anti-apoptotic proteins Bcl-2 and Bcl-xL reduced. Interestingly, when FHL2 was knocked down, the protein expression level of iASPP also decreased. Similarly, the expression of FHL2 would reduce when iASPP was silenced. These results indicated that FHL2 might be a novel potential target for acute myelocytic leukemia treatment.
Collapse
Affiliation(s)
- Wenting Lu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Tengteng Yu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Shuang Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Saisai Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Shouyun Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Jia Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
29
|
Cai T, Sun D, Duan Y, Qiu Y, Dai C, Yang J, He W. FHL2 promotes tubular epithelial-to-mesenchymal transition through modulating β-catenin signalling. J Cell Mol Med 2017; 22:1684-1695. [PMID: 29193729 PMCID: PMC5824423 DOI: 10.1111/jcmm.13446] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022] Open
Abstract
β-Catenin signalling plays an important role in regulating tubular epithelial-to-mesenchymal transition (EMT), an indispensable programme for driving renal fibrosis. As an adapter protein, four and a half LIM domain protein 2 (FHL2) acts as a coregulator of β-catenin in several other cell types. To determine whether FHL2 affects β-catenin signalling and thus is involved in tubular EMT, we examined its expression and function in the process of TGF-β1-induced EMT. FHL2 mRNA and protein were induced by TGF-β1 in rat tubular epithelial cells (NRK-52E), an effect that intracellular Smad signalling was required. Ectopic expression of FHL2 inhibited E-cadherin and enhanced α-smooth muscle actin (α-SMA) and fibronectin expression, whereas knockdown of FHL2 partially restored E-cadherin and reduced α-SMA and fibronectin induction stimulated by TGF-β1. Overexpression of FHL2 increased β-catenin dephosphorylation (Ser37/Thr41), nuclear translocation and β-catenin-mediated transcription and up-regulated expression of β-catenin target, EMT-related genes, such as Snail, Twist, vimentin, plasminogen activator inhibitor-1 and matrix metalloproteinase-7. Conversely, knockdown of FHL2 increased β-catenin phosphorylation (Ser33/37/Thr41), decreased its nuclear translocation and inhibited β-catenin-mediated transcription and target genes expression. TGF-β1 induced a FHL2/β-catenin interaction in NRK-52E cells, especially in the nuclei. In a mouse model of obstructive nephropathy, FHL2 mRNA and protein were induced in a time-dependent fashion, and the extent and pattern of renal β-catenin activation were positively correlated with FHL2 induction. Collectively, this study suggests that FHL2, via modulating β-catenin signalling, may implicate in regulation of TGF-β1-mediated tubular EMT and could be a potential therapeutic target for fibrotic kidney disease.
Collapse
Affiliation(s)
- Ting Cai
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danqin Sun
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Duan
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yumei Qiu
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunsun Dai
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weichun He
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
30
|
Gasparics Á, Sebe A. MRTFs- master regulators of EMT. Dev Dyn 2017; 247:396-404. [PMID: 28681541 DOI: 10.1002/dvdy.24544] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/19/2022] Open
Abstract
Recent evidence implicates the myocardin-related transcription factors (MRTFs) as key mediators of the phenotypic plasticity leading to the conversion of various cell types into myofibroblasts. This review highlights the function of MRTFs during development, fibrosis and cancer, and the role of MRTFs during epithelial-mesenchymal transitions (EMTs) underlying these processes. EMT is a sequentially orchestrated process where cells undergo a rearrangement of their cell contacts and activate a fibrogenic and myogenic expression program. MRTFs interact with and regulate the major signaling pathways and the expression of key markers and transcription factors involved in EMT. These functions indicate a central role for MRTFs in controlling the process of EMT. Developmental Dynamics 247:396-404, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ákos Gasparics
- Semmelweis University, Department of Pathophysiology, Budapest, Hungary.,Semmelweis University, 1st Department of Obstetrics and Gynecology, Budapest, Hungary
| | - Attila Sebe
- Semmelweis University, Department of Pathophysiology, Budapest, Hungary.,Paul Ehrlich Institute, Division of Medical Biotechnology, Langen, Germany
| |
Collapse
|
31
|
Li SY, Park J, Qiu C, Han SH, Palmer MB, Arany Z, Susztak K. Increasing the level of peroxisome proliferator-activated receptor γ coactivator-1α in podocytes results in collapsing glomerulopathy. JCI Insight 2017; 2:92930. [PMID: 28724797 DOI: 10.1172/jci.insight.92930] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/06/2017] [Indexed: 01/07/2023] Open
Abstract
Inherited and acquired mitochondrial defects have been associated with podocyte dysfunction and chronic kidney disease (CKD). Peroxisome proliferator-activated receptor γ coactivator-1α (PGC1α) is one of the main transcriptional regulators of mitochondrial biogenesis and function. We hypothesized that increasing PGC1α expression in podocytes could protect from CKD. We found that PGC1α and mitochondrial transcript levels are lower in podocytes of patients and mouse models with diabetic kidney disease (DKD). To increase PGC1α expression, podocyte-specific inducible PGC1α-transgenic mice were generated by crossing nephrin-rtTA mice with tetO-Ppargc1a animals. Transgene induction resulted in albuminuria and glomerulosclerosis in a dose-dependent manner. Expression of PGC1α in podocytes increased mitochondrial biogenesis and maximal respiratory capacity. PGC1α also shifted podocytes towards fatty acid usage from their baseline glucose preference. RNA sequencing analysis indicated that PGC1α induced podocyte proliferation. Histological lesions of mice with podocyte-specific PGC1α expression resembled collapsing focal segmental glomerular sclerosis. In conclusion, decreased podocyte PGC1α expression and mitochondrial content is a consistent feature of DKD, but excessive PGC1α alters mitochondrial properties and induces podocyte proliferation and dedifferentiation, indicating that there is likely a narrow therapeutic window for PGC1α levels in podocytes.
Collapse
Affiliation(s)
- Szu-Yuan Li
- Renal-Electrolyte and Hypertension Division of Department of Medicine, and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.,Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital and School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jihwan Park
- Renal-Electrolyte and Hypertension Division of Department of Medicine, and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Chengxiang Qiu
- Renal-Electrolyte and Hypertension Division of Department of Medicine, and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Seung Hyeok Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | | | - Zoltan Arany
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Katalin Susztak
- Renal-Electrolyte and Hypertension Division of Department of Medicine, and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
32
|
Saul MC, Seward CH, Troy JM, Zhang H, Sloofman LG, Lu X, Weisner PA, Caetano-Anolles D, Sun H, Zhao SD, Chandrasekaran S, Sinha S, Stubbs L. Transcriptional regulatory dynamics drive coordinated metabolic and neural response to social challenge in mice. Genome Res 2017; 27:959-972. [PMID: 28356321 PMCID: PMC5453329 DOI: 10.1101/gr.214221.116] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 03/24/2017] [Indexed: 12/22/2022]
Abstract
Agonistic encounters are powerful effectors of future behavior, and the ability to learn from this type of social challenge is an essential adaptive trait. We recently identified a conserved transcriptional program defining the response to social challenge across animal species, highly enriched in transcription factor (TF), energy metabolism, and developmental signaling genes. To understand the trajectory of this program and to uncover the most important regulatory influences controlling this response, we integrated gene expression data with the chromatin landscape in the hypothalamus, frontal cortex, and amygdala of socially challenged mice over time. The expression data revealed a complex spatiotemporal patterning of events starting with neural signaling molecules in the frontal cortex and ending in the modulation of developmental factors in the amygdala and hypothalamus, underpinned by a systems-wide shift in expression of energy metabolism-related genes. The transcriptional signals were correlated with significant shifts in chromatin accessibility and a network of challenge-associated TFs. Among these, the conserved metabolic and developmental regulator ESRRA was highlighted for an especially early and important regulatory role. Cell-type deconvolution analysis attributed the differential metabolic and developmental signals in this social context primarily to oligodendrocytes and neurons, respectively, and we show that ESRRA is expressed in both cell types. Localizing ESRRA binding sites in cortical chromatin, we show that this nuclear receptor binds both differentially expressed energy-related and neurodevelopmental TF genes. These data link metabolic and neurodevelopmental signaling to social challenge, and identify key regulatory drivers of this process with unprecedented tissue and temporal resolution.
Collapse
Affiliation(s)
- Michael C Saul
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Christopher H Seward
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Joseph M Troy
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Illinois Informatics Institute, Urbana, Illinois 61801, USA
| | - Huimin Zhang
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Laura G Sloofman
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Xiaochen Lu
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Patricia A Weisner
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Derek Caetano-Anolles
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Hao Sun
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Sihai Dave Zhao
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Sriram Chandrasekaran
- Harvard Society of Fellows, Harvard University, Cambridge, Massachusetts 02138, USA
- Faculty of Arts and Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Saurabh Sinha
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Computer Science
- Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Lisa Stubbs
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
33
|
LIM-Only Protein FHL2 Is a Negative Regulator of Transforming Growth Factor β1 Expression. Mol Cell Biol 2017; 37:MCB.00636-16. [PMID: 28223370 DOI: 10.1128/mcb.00636-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/15/2017] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor β1 (TGF-β1) is a master cytokine in many biological processes, including tissue homeostasis, epithelial-to-mesenchymal transition, and wound repair. Here, we report that four and a half LIM-only protein 2 (FHL2) is a critical regulator of TGF-β1 expression. Devoid of a DNA-binding domain, FHL2 is a transcriptional cofactor that plays the role of coactivator or corepressor, depending on the cell and promoter contexts. We detected association of FHL2 with the TGF-β1 promoter, which showed higher activity in Fhl2-/- cells than in wild-type (WT) cells in a reporter assay. Overexpression of FHL2 abrogates the activation of the TGF-β1 promoter, whereas the upregulation of TGF-β1 gene transcription correlates with reduced occupancy of FHL2 on the promoter. Moreover, ablation of FHL2 facilitates recruitment of RNA polymerase II on the TGF-β1 promoter, suggesting that FHL2 may be involved in chromatin remodeling in the control of TGF-β1 gene transcription. Enhanced expression of TGF-β1 mRNA and cytokine was evidenced in the livers of Fhl2-/- mice. We tested the in vivo impact of Fhl2 loss on hepatic fibrogenesis that involves TGF-β1 activation. Fhl2-/- mice developed more severe fibrosis than their WT counterparts. These results demonstrate the repressive function of FHL2 on TGF-β1 expression and contribute to the understanding of the TGF-β-mediated fibrogenic response.
Collapse
|
34
|
Wang S, Lu Y, Sun X, Wu D, Fu B, Chen Y, Deng H, Chen X. Identification of common and differential mechanisms of glomerulus and tubule senescence in 24-month-old rats by quantitative LC-MS/MS. Proteomics 2016; 16:2706-2717. [PMID: 27452873 DOI: 10.1002/pmic.201600121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/05/2016] [Accepted: 07/20/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Shiyu Wang
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
- Department of Nephrology; The Second Hospital of Jilin University; Changchun Jilin P.R. China
| | - Yang Lu
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Xuefeng Sun
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Di Wu
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Bo Fu
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics; School of Life Sciences; Tsinghua University; Beijing P.R. China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics; School of Life Sciences; Tsinghua University; Beijing P.R. China
| | - Xiangmei Chen
- Department of Nephrology; Chinese PLA General Hospital; Chinese PLA Institute of Nephrology, Beijing Key Laboratory of Kidney Disease, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases; Beijing P.R. China
| |
Collapse
|
35
|
Garlapow ME, Huang W, Yarboro MT, Peterson KR, Mackay TFC. Quantitative Genetics of Food Intake in Drosophila melanogaster. PLoS One 2015; 10:e0138129. [PMID: 26375667 PMCID: PMC4574202 DOI: 10.1371/journal.pone.0138129] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/25/2015] [Indexed: 12/16/2022] Open
Abstract
Food intake is an essential animal activity, regulated by neural circuits that motivate food localization, evaluate nutritional content and acceptance or rejection responses through the gustatory system, and regulate neuroendocrine feedback loops that maintain energy homeostasis. Excess food consumption in people is associated with obesity and metabolic and cardiovascular disorders. However, little is known about the genetic basis of natural variation in food consumption. To gain insights in evolutionarily conserved genetic principles that regulate food intake, we took advantage of a model system, Drosophila melanogaster, in which food intake, environmental conditions and genetic background can be controlled precisely. We quantified variation in food intake among 182 inbred, sequenced lines of the Drosophila melanogaster Genetic Reference Panel (DGRP). We found significant genetic variation in the mean and within-line environmental variance of food consumption and observed sexual dimorphism and genetic variation in sexual dimorphism for both food intake traits (mean and variance). We performed genome wide association (GWA) analyses for mean food intake and environmental variance of food intake (using the coefficient of environmental variation, CVE, as the metric for environmental variance) and identified molecular polymorphisms associated with both traits. Validation experiments using RNAi-knockdown confirmed 24 of 31 (77%) candidate genes affecting food intake and/or variance of food intake, and a test cross between selected DGRP lines confirmed a SNP affecting mean food intake identified in the GWA analysis. The majority of the validated candidate genes were novel with respect to feeding behavior, and many had mammalian orthologs implicated in metabolic diseases.
Collapse
Affiliation(s)
- Megan E. Garlapow
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695–7614, United States of America
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, United States of America
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695, United States of America
| | - Wen Huang
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695–7614, United States of America
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, United States of America
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695, United States of America
| | - Michael T. Yarboro
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, United States of America
| | - Kara R. Peterson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, United States of America
| | - Trudy F. C. Mackay
- Program in Genetics, North Carolina State University, Raleigh, NC, 27695–7614, United States of America
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, United States of America
- W. M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, 27695, United States of America
- * E-mail:
| |
Collapse
|