1
|
Xu X, Wang X, Li Y, Chen R, Wen H, Wang Y, Ma G. Research progress of ankyrin repeat domain 1 protein: an updated review. Cell Mol Biol Lett 2024; 29:131. [PMID: 39420247 PMCID: PMC11488291 DOI: 10.1186/s11658-024-00647-w] [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: 08/06/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Ankyrin repeat domain 1 (Ankrd1) is an acute response protein that belongs to the muscle ankyrin repeat protein (MARP) family. Accumulating evidence has revealed that Ankrd1 plays a crucial role in a wide range of biological processes and diseases. This review consolidates current knowledge on Ankrd1's functions in myocardium and skeletal muscle development, neurogenesis, cancer, bone formation, angiogenesis, wound healing, fibrosis, apoptosis, inflammation, and infection. The comprehensive profile of Ankrd1 in cardiovascular diseases, myopathy, and its potential as a candidate prognostic and diagnostic biomarker are also discussed. In the future, more studies of Ankrd1 are warranted to clarify its role in diseases and assess its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xusan Xu
- Maternal and Child Research Institute, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China
| | - Xiaoxia Wang
- Department of Neurology, Longjiang Hospital, Foshan, 528300, China
| | - Yu Li
- Department of Pediatrics, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China
| | - Riling Chen
- Department of Pediatrics, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China
| | - Houlang Wen
- Medical Genetics Laboratory, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China.
| | - Yajun Wang
- Respiratory Research Institute, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China.
| | - Guoda Ma
- Maternal and Child Research Institute, Shunde Women and Children Hospital, Guangdong Medical University, Foshan, 528300, China.
| |
Collapse
|
2
|
Wang X, Sun Z, Fu J, Fang Z, Zhang W, He JC, Lee K. LRG1 loss effectively restrains glomerular TGF-β signaling to attenuate diabetic kidney disease. Mol Ther 2024; 32:3177-3193. [PMID: 38910328 PMCID: PMC11403230 DOI: 10.1016/j.ymthe.2024.06.027] [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: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
Transforming growth factor (TGF)-β signaling is a well-established pathogenic mediator of diabetic kidney disease (DKD). However, owing to its pleiotropic actions, its systemic blockade is not therapeutically optimal. The expression of TGF-β signaling regulators can substantially influence TGF-β's effects in a cell- or context-specific manner. Among these, leucine-rich α2-glycoprotein 1 (LRG1) is significantly increased in glomerular endothelial cells (GECs) in DKD. As LRG1 is a secreted molecule that can exert autocrine and paracrine effects, we examined the effects of LRG1 loss in kidney cells in diabetic OVE26 mice by single-cell transcriptomic analysis. Gene expression analysis confirmed a predominant expression of Lrg1 in GECs, which further increased in diabetic kidneys. Loss of Lrg1 led to the reversal of angiogenic and TGF-β-induced gene expression in GECs, which were associated with DKD attenuation. Notably, Lrg1 loss also mitigated the increased TGF-β-mediated gene expression in both podocytes and mesangial cells in diabetic mice, indicating that GEC-derived LRG1 potentiates TGF-β signaling in glomerular cells in an autocrine and paracrine manner. Indeed, a significant reduction in phospho-Smad proteins was observed in the glomerular cells of OVE26 mice with LRG1 loss. These results indicate that specific antagonisms of LRG1 may be an effective approach to curb the hyperactive glomerular TGF-β signaling to attenuate DKD.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeguo Sun
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jia Fu
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhengying Fang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Weijia Zhang
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Renal Section, James J. Peters Veterans Affair Medical Center, Bronx, NY 10468, USA.
| | - Kyung Lee
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
3
|
Han S, Guo J, Kong C, Li J, Lin F, Zhu J, Wang T, Chen Q, Liu Y, Hu H, Qiu T, Cheng F, Zhou J. ANKRD1 aggravates renal ischaemia‒reperfusion injury via promoting TRIM25-mediated ubiquitination of ACSL3. Clin Transl Med 2024; 14:e70024. [PMID: 39285846 PMCID: PMC11406046 DOI: 10.1002/ctm2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Renal ischaemia‒reperfusion injury (IRI) is the primary cause of acute kidney injury (AKI). To date, effective therapies for delaying renal IRI and postponing patient survival remain absent. Ankyrin repeat domain 1 (ANKRD1) has been implicated in some pathophysiologic processes, but its role in renal IRI has not been explored. METHODS The mouse model of IRI-AKI and in vitro model were utilised to investigate the role of ANKRD1. Immunoprecipitation-mass spectrometry was performed to identify potential ANKRD1-interacting proteins. Protein‒protein interactions and protein ubiquitination were examined using immunoprecipitation and proximity ligation assay and immunoblotting, respectively. Cell viability, damage and lipid peroxidation were evaluated using biochemical and cellular techniques. RESULTS First, we unveiled that ANKRD1 were significantly elevated in renal IRI models. Global knockdown of ANKRD1 in all cell types of mouse kidney by recombinant adeno-associated virus (rAAV9)-mitigated ischaemia/reperfusion-induced renal damage and failure. Silencing ANKRD1 enhanced cell viability and alleviated cell damage in human renal proximal tubule cells exposed to hypoxia reoxygenation or hydrogen peroxide, while ANKRD1 overexpression had the opposite effect. Second, we discovered that ANKRD1's detrimental function during renal IRI involves promoting lipid peroxidation and ferroptosis by directly binding to and decreasing levels of acyl-coenzyme A synthetase long-chain family member 3 (ACSL3), a key protein in lipid metabolism. Furthermore, attenuating ACSL3 in vivo through pharmaceutical approach and in vitro via RNA interference mitigated the anti-ferroptotic effect of ANKRD1 knockdown. Finally, we showed ANKRD1 facilitated post-translational degradation of ACSL3 by modulating E3 ligase tripartite motif containing 25 (TRIM25) to catalyse K63-linked ubiquitination of ACSL3, thereby amplifying lipid peroxidation and ferroptosis, exacerbating renal injury. CONCLUSIONS Our study revealed a previously unknown function of ANKRD1 in renal IRI. By driving ACSL3 ubiquitination and degradation, ANKRD1 aggravates ferroptosis and ultimately exacerbates IRI-AKI, underlining ANKRD1's potential as a therapeutic target for kidney IRI. KEY POINTS/HIGHLIGHTS Ankyrin repeat domain 1 (ANKRD1) is rapidly activated in renal ischaemia‒reperfusion injury (IRI) models in vivo and in vitro. ANKRD1 knockdown mitigates kidney damage and preserves renal function. Ferroptosis contributes to the deteriorating function of ANKRD1 in renal IRI. ANKRD1 promotes acyl-coenzyme A synthetase long-chain family member 3 (ACSL3) degradation via the ubiquitin‒proteasome pathway. The E3 ligase tripartite motif containing 25 (TRIM25) is responsible for ANKRD1-mediated ubiquitination of ACSL3.
Collapse
Affiliation(s)
- Shangting Han
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jiayu Guo
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Chenyang Kong
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
- Department of NephrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jun Li
- Key Laboratory of Medical ElectrophysiologyMinistry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical UniversityLuzhouChina
| | - Fangyou Lin
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jiefu Zhu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Tianyu Wang
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Qi Chen
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yiting Liu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Haochong Hu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Tao Qiu
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fan Cheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Jiangqiao Zhou
- Department of Organ TransplantationRenmin Hospital of Wuhan UniversityWuhanChina
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
4
|
Fu J, Yi Z, Cai M, Yuan W, Zhang W, Lee K, He JC. Global transcriptomic changes in glomerular endothelial cells in mice with podocyte depletion and glomerulosclerosis. Cell Death Dis 2021; 12:687. [PMID: 34244474 PMCID: PMC8270962 DOI: 10.1038/s41419-021-03951-x] [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: 12/10/2020] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023]
Abstract
Podocytes are a key component of the glomerular filtration barrier, and its dysfunction and eventual loss drive glomerular disease progression. Recent research has demonstrated the importance of podocyte cross-talk with other glomerular cells, such as glomerular endothelial cells (GECs), in both glomerular homeostasis and in disease settings. However, how GECs are affected globally by podocyte injury and loss in disease settings remains unclear. Therefore, to characterize the molecular changes occurring in GECs in response to the podocyte loss, we performed the transcriptomic profiling of isolated GECs after diphtheria toxin (DT)-mediated podocyte depletion in transgenic mice with podocyte-specific human DT receptor and endothelial-specific enhanced yellow fluorescent protein (EYFP) expression. DT administration led to nearly 40% of podocyte loss with the development of glomerulosclerosis. Differential gene expression analysis of isolated GECs in the diseased mice showed significant changes in pathways related to cell adhesion and actin cytoskeleton, proliferation, and angiogenesis, as well as apoptosis and cell death. However, quantification of EYFP + GECs indicated that there was a reduction in GECs in the diseased mice, suggesting that despite the ongoing proliferation, the concomitant injury and the activation of cell death program results in their overall net loss. The upstream regulator analysis strongly indicated the involvement of p53, TGF-β1, and TNF-α as key mediators of the molecular changes occurring in GECs in the diseased mice. Our findings demonstrate significant molecular changes in GECs as a secondary consequence of podocyte loss and provide a valuable resource for further in-depth analysis of potential glomerular cross-talk mediators.
Collapse
Affiliation(s)
- Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Minchao Cai
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Renal Program, James J. Peters Veterans Affairs Medical Center at Bronx, New York, NY, USA.
| |
Collapse
|
5
|
Lake JA, Papah MB, Abasht B. Increased Expression of Lipid Metabolism Genes in Early Stages of Wooden Breast Links Myopathy of Broilers to Metabolic Syndrome in Humans. Genes (Basel) 2019; 10:E746. [PMID: 31557856 PMCID: PMC6826700 DOI: 10.3390/genes10100746] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022] Open
Abstract
Wooden breast is a muscle disorder affecting modern commercial broiler chickens that causes a palpably firm pectoralis major muscle and severe reduction in meat quality. Most studies have focused on advanced stages of wooden breast apparent at market age, resulting in limited insights into the etiology and early pathogenesis of the myopathy. Therefore, the objective of this study was to identify early molecular signals in the wooden breast transcriptional cascade by performing gene expression analysis on the pectoralis major muscle of two-week-old birds that may later exhibit the wooden breast phenotype by market age at 7 weeks. Biopsy samples of the left pectoralis major muscle were collected from 101 birds at 14 days of age. Birds were subsequently raised to 7 weeks of age to allow sample selection based on the wooden breast phenotype at market age. RNA-sequencing was performed on 5 unaffected and 8 affected female chicken samples, selected based on wooden breast scores (0 to 4) assigned at necropsy where affected birds had scores of 2 or 3 (mildly or moderately affected) while unaffected birds had scores of 0 (no apparent gross lesions). Differential expression analysis identified 60 genes found to be significant at an FDR-adjusted p-value of 0.05. Of these, 26 were previously demonstrated to exhibit altered expression or genetic polymorphisms related to glucose tolerance or diabetes mellitus in mammals. Additionally, 9 genes have functions directly related to lipid metabolism and 11 genes are associated with adiposity traits such as intramuscular fat and body mass index. This study suggests that wooden breast disease is first and foremost a metabolic disorder characterized primarily by ectopic lipid accumulation in the pectoralis major.
Collapse
Affiliation(s)
- Juniper A Lake
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE 19711, USA.
| | - Michael B Papah
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA.
| | - Behnam Abasht
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
6
|
Bin L, Li X, Richers B, Streib JE, Hu JW, Taylor P, Leung DYM. Ankyrin repeat domain 1 regulates innate immune responses against herpes simplex virus 1: A potential role in eczema herpeticum. J Allergy Clin Immunol 2018; 141:2085-2093.e1. [PMID: 29371118 DOI: 10.1016/j.jaci.2018.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/16/2017] [Accepted: 01/08/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is a common inflammatory skin disease. A subset of patients with AD are susceptible to disseminated herpes simplex virus (HSV) infection, a complication termed eczema herpeticum (ADEH+). The immune mechanisms causing ADEH+ remain elusive. Using RNA sequencing, we recently found that ankyrin repeat domain 1 (ANKRD1) was significantly induced in human PBMCs upon HSV-1 stimulation, and its induction in patients with ADEH+ was significantly reduced compared with that seen in AD patients without a history of eczema herpeticum (ADEH-). OBJECTIVE We sought to validate ANKRD1 gene expression in nonatopic (NA) subjects, patients with ADEH-, and patients with ADEH+ and to delineate the biological function of ANKRD1 and the signaling pathway or pathways involved. METHODS Purification of human PBMCs, monocytes, B cells, dendritic cells, T cells, and natural killer cells; RNA extraction and quantitative RT-PCR; small interfering RNA technique; co-immunoprecipitation; and Western blot assays were used. RESULTS ANKRD1 expression was significantly reduced in PBMCs from patients with ADEH+ after HSV-1 stimulation compared with PBMCs from patients with ADEH-. We found that the induction of ANKRD1 by HSV-1 and multiple pattern recognition receptor agonists are mediated by inflammatory cytokines. Silencing ANKRD1 gene expression in antigen-presenting cells led to increased viral load and reduced IFNB1 and IL29 production. Using co-immunoprecipitation methods, we demonstrated that ANKRD1 formed protein complexes with interferon regulatory factor (IRF) 3 and IRF7, which are important transcription factors regulating signaling transduction of pattern recognition receptors. Overexpression of ANKRD1 enhanced the IRF3-mediated signaling pathways. CONCLUSION ANKRD1 is involved in IRF3-mediated antiviral innate immune signaling pathways. Its reduced expression in patients with ADEH+ might contribute to the pathogenesis of ADEH+.
Collapse
Affiliation(s)
- Lianghua Bin
- Department of Pediatrics, National Jewish Health, Denver, Colo; First Affiliated Hospital, Biomedical Translational Research Institute, the International Immunology Center and the Key Laboratory of Antibody Engineering of Guangdong Province, Jinan University, Guangzhou, China
| | - Xiaozhao Li
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | - Joanne E Streib
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | - Patricia Taylor
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colo; University of Colorado School of Medicine, Aurora, Colo.
| |
Collapse
|
7
|
Zhang N, Xie XJ, Wang JA. Multifunctional protein: cardiac ankyrin repeat protein. J Zhejiang Univ Sci B 2017; 17:333-41. [PMID: 27143260 DOI: 10.1631/jzus.b1500247] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cardiac ankyrin repeat protein (CARP) not only serves as an important component of muscle sarcomere in the cytoplasm, but also acts as a transcription co-factor in the nucleus. Previous studies have demonstrated that CARP is up-regulated in some cardiovascular disorders and muscle diseases; however, its role in these diseases remains controversial now. In this review, we will discuss the continued progress in the research related to CARP, including its discovery, structure, and the role it plays in cardiac development and heart diseases.
Collapse
Affiliation(s)
- Na Zhang
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiao-Jie Xie
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jian-An Wang
- Cardiovascular Key Lab of Zhejiang Province, Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
8
|
Kai T, Tsukamoto Y, Hijiya N, Tokunaga A, Nakada C, Uchida T, Daa T, Iha H, Takahashi M, Nomura T, Sato F, Mimata H, Ikawa M, Seto M, Matsuura K, Moriyama M. Kidney-specific knockout ofSav1in the mouse promotes hyperproliferation of renal tubular epithelium through suppression of the Hippo pathway. J Pathol 2016; 239:97-108. [DOI: 10.1002/path.4706] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/18/2016] [Accepted: 02/12/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Tomoki Kai
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
- Department of Urology, Faculty of Medicine; Oita University; Yufu Japan
| | - Yoshiyuki Tsukamoto
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
| | - Naoki Hijiya
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
| | - Akinori Tokunaga
- Section of Physiology, Department of Integrative Aging Neuroscience; National Center for Geriatrics and Gerontology; Obu Japan
| | - Chisato Nakada
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
| | - Tomohisa Uchida
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
| | - Tsutomu Daa
- Department of Diagnostic Pathology, Faculty of Medicine; Oita University; Yufu Japan
| | - Hidekatsu Iha
- Department of Microbiology, Faculty of Medicine; Oita University; Yufu Japan
| | - Mika Takahashi
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
- Department of Urology, Faculty of Medicine; Oita University; Yufu Japan
| | - Takeo Nomura
- Department of Urology, Faculty of Medicine; Oita University; Yufu Japan
| | - Fuminori Sato
- Department of Urology, Faculty of Medicine; Oita University; Yufu Japan
| | - Hiromitsu Mimata
- Department of Urology, Faculty of Medicine; Oita University; Yufu Japan
| | - Masahito Ikawa
- Animal Resource Center for Infectious Diseases; Research Institute for Microbial Diseases; Suita Japan
| | - Masao Seto
- Division of Molecular Medicine; Aichi Cancer Institute; Nagoya Japan
| | - Keiko Matsuura
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
- Department of Biology, Faculty of Medicine; Oita University; Yufu Japan
| | - Masatsugu Moriyama
- Department of Molecular Pathology, Faculty of Medicine; Oita University; Yufu Japan
| |
Collapse
|
9
|
Liu XH, Bauman WA, Cardozo C. ANKRD1 modulates inflammatory responses in C2C12 myoblasts through feedback inhibition of NF-κB signaling activity. Biochem Biophys Res Commun 2015; 464:208-13. [DOI: 10.1016/j.bbrc.2015.06.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/17/2015] [Indexed: 02/08/2023]
|
10
|
Samaras SE, Almodóvar-García K, Wu N, Yu F, Davidson JM. Global deletion of Ankrd1 results in a wound-healing phenotype associated with dermal fibroblast dysfunction. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:96-109. [PMID: 25452119 DOI: 10.1016/j.ajpath.2014.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/27/2022]
Abstract
The expression of ankyrin repeat domain protein 1 (Ankrd1), a transcriptional cofactor and sarcomeric component, is strongly elevated by wounding and tissue injury. We developed a conditional Ankrd1(fl/fl) mouse, performed global deletion with Sox2-cre, and assessed the role of this protein in cutaneous wound healing. Although global deletion of Ankrd1 did not affect mouse viability or development, Ankrd1(-/-) mice had at least two significant wound-healing phenotypes: extensive necrosis of ischemic skin flaps, which was reversed by adenoviral expression of ANKRD1, and delayed excisional wound closure, which was characterized by decreased contraction and reduced granulation tissue thickness. Skin fibroblasts isolated from Ankrd1(-/-) mice did not spread or migrate on collagen- or fibronectin-coated surfaces as efficiently as fibroblasts isolated from Ankrd1(fl/fl) mice. More important, Ankrd1(-/-) fibroblasts failed to contract three-dimensional floating collagen gels. Reconstitution of ANKRD1 by adenoviral infection stimulated both collagen gel contraction and actin fiber organization. These in vitro data were consistent with in vivo wound closure studies, and suggest that ANKRD1 is important for the proper interaction of fibroblasts with a compliant collagenous matrix both in vitro and in vivo.
Collapse
Affiliation(s)
- Susan E Samaras
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Karinna Almodóvar-García
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Nanjun Wu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Fang Yu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jeffrey M Davidson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee; Research Service, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee.
| |
Collapse
|
11
|
Bethunaickan R, Berthier CC, Zhang W, Kretzler M, Davidson A. Comparative transcriptional profiling of 3 murine models of SLE nephritis reveals both unique and shared regulatory networks. PLoS One 2013; 8:e77489. [PMID: 24167575 PMCID: PMC3805607 DOI: 10.1371/journal.pone.0077489] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 08/31/2013] [Indexed: 11/30/2022] Open
Abstract
Objective To define shared and unique features of SLE nephritis in mouse models of proliferative and glomerulosclerotic renal disease. Methods Perfused kidneys from NZB/W F1, NZW/BXSB and NZM2410 mice were harvested before and after nephritis onset. Affymetrix based gene expression profiles of kidney RNA were analyzed using Genomatix Pathway Systems and Ingenuity Pathway Analysis software. Gene expression patterns were confirmed using real-time PCR. Results 955, 1168 and 755 genes were regulated in the kidneys of nephritic NZB/W F1, NZM2410 and NZW/BXSB mice respectively. 263 genes were regulated concordantly in all three strains reflecting immune cell infiltration, endothelial cell activation, complement activation, cytokine signaling, tissue remodeling and hypoxia. STAT3 was the top associated transcription factor, having a binding site in the gene promoter of 60/263 regulated genes. The two strains with proliferative nephritis shared a macrophage/DC infiltration and activation signature. NZB/W and NZM2410 mice shared a mitochondrial dysfunction signature. Dominant T cell and plasma cell signatures in NZB/W mice reflected lymphoid aggregates; this was the only strain with regulatory T cell infiltrates. NZW/BXSB mice manifested tubular regeneration and NZM2410 mice had the most metabolic stress and manifested loss of nephrin, indicating podocyte loss. Conclusions These findings identify shared inflammatory mechanisms of SLE nephritis that can be therapeutically targeted. Nevertheless, the heterogeneity of effector mechanisms suggests that individualized therapy might need to be based on biopsy findings. Some common mechanisms are shared with non-immune–mediated renal diseases, suggesting that strategies to prevent tissue hypoxia and remodeling may be useful in SLE nephritis.
Collapse
Affiliation(s)
- Ramalingam Bethunaickan
- Center for Autoimmunity and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, New York, United States of America
| | - Celine C. Berthier
- Department of Internal Medicine, Nephrology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Weijia Zhang
- Department of Medicine, Mount Sinai Medical Center, New York, New York, United States of America
| | - Matthias Kretzler
- Department of Internal Medicine, Nephrology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Anne Davidson
- Center for Autoimmunity and Musculoskeletal Diseases, Feinstein Institute for Medical Research, Manhasset, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
12
|
Wu Y, Ruggiero CL, Bauman WA, Cardozo C. Ankrd1 is a transcriptional repressor for the androgen receptor that is downregulated by testosterone. Biochem Biophys Res Commun 2013; 437:355-60. [PMID: 23811403 DOI: 10.1016/j.bbrc.2013.06.079] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 10/26/2022]
Abstract
The ankryn repeat domain proteins, Ankrd1 and Ankrd2, are expressed at the highest levels in skeletal muscle and heart where they are localized to the I band of the sarcomere through binding to titin and myopaladin. Ankrd1 and Ankrd2 migrate from the sarcomere to the nucleus when muscle is stressed, and act as coregulators for a growing number of transcription factors. Expression of Ankrd1 is altered by castration suggesting a link to androgen action. This investigation explored the effects of testosterone on Ankrd1 and Ankrd2 expression and determined whether Ankrd1 or Ankrd2 binds to or regulates the transcriptional activity of the androgen receptor (AR). Incubation of rat L6 myoblasts expressing the human AR (L6.AR) with testosterone reduced mRNA levels for Ankrd1 by approximately 50% and increased those for Ankrd2 by 20-fold. In reporter gene assays conducted with CHO cells co-transfected with an ARE-Luc reporter gene, Ankrd1 blocked the ability of testosterone to increase reporter gene activity while Ankrd2 had no effect. The effect of Ankrd1 and Ankrd2 on repression of the MAFbx promoter by testosterone was also tested in C2C12 cells using an MAFbx-Luc reporter gene (pMAF400-Luc); Ankrd1 blocked repression of pMAF400-Luc by testosterone while Ankrd2 did not. Co-immunoprecipitation studies revealed that Ankrd1 bound to the AR whereas Ankrd2 did not. The effect of Ankrd1 or Ankrd2 on changes in gene expression induced by testosterone in L6.AR cells was also evaluated. Incubation of L6.AR cells with testosterone modestly reduced myogenin mRNA levels but did not significantly alter those for mdm2, MEF2d, TnnI1, TnnI2, or p21. When cells were transfected with Ankrd1, testosterone markedly reduced mRNA levels for MEF2d, myogenin, p21 and TnnI1, increased those for TnnI2, but did not alter those for mdm2. When cells were transfected with Ankrd2, testosterone increased MEF2d and myogenin mRNA levels, having the opposite effect to cells transfected with Ankrd1; Ankrd2 did not change the effects of testosterone on TnnI1, TnnI2, p21, or mdm2 mRNA levels. In conclusion, testosterone regulates the expression of Ankrd1 and Ankrd2; Ankrd1 binds to and directly regulates the transcriptional activity of the AR whereas Ankrd2 does not; expression levels of both Ankrd1 and Ankrd2 modulate effects of testosterone on gene expression in cultured myoblasts.
Collapse
Affiliation(s)
- Yong Wu
- National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peter Medical Center, Bronx, NY, United States
| | | | | | | |
Collapse
|
13
|
Samaras SE, Chen B, Koch SR, Sawyer DB, Lim CC, Davidson JM. 26S proteasome regulation of Ankrd1/CARP in adult rat ventricular myocytes and human microvascular endothelial cells. Biochem Biophys Res Commun 2012; 425:830-5. [PMID: 22892129 DOI: 10.1016/j.bbrc.2012.07.162] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 07/30/2012] [Indexed: 01/04/2023]
Abstract
Ankyrin repeat domain 1 protein (Ankrd1), also known as cardiac ankyrin repeat protein (CARP), increases dramatically after tissue injury, and its overexpression improves aspects of wound healing. Reports that Ankrd1/CARP protein stability may affect cardiovascular organization, together with our findings that the protein is crucial to stability of the cardiomyocyte sarcomere and increased in wound healing, led us to compare the contribution of Ankrd1/CARP stability to its abundance. We found that the 26S proteasome is the dominant regulator of Ankrd1/CARP degradation, and that Ankrd1/CARP half-life is significantly longer in cardiomyocytes (h) than endothelial cells (min). In addition, higher endothelial cell density decreased the abundance of the protein without affecting steady state mRNA levels. Taken together, our data and that of others indicate that Ankrd1/CARP is highly regulated at multiple levels of its expression. The striking difference in protein half-life between a muscle and a non-muscle cell type suggests that post-translational proteolysis is correlated with the predominantly structural versus regulatory role of the protein in the two cell types.
Collapse
Affiliation(s)
- Susan E Samaras
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232-2561, USA
| | | | | | | | | | | |
Collapse
|
14
|
Matsuura K, Nakada C, Mashio M, Narimatsu T, Yoshimoto T, Tanigawa M, Tsukamoto Y, Hijiya N, Takeuchi I, Nomura T, Sato F, Mimata H, Seto M, Moriyama M. Downregulation of SAV1 plays a role in pathogenesis of high-grade clear cell renal cell carcinoma. BMC Cancer 2011; 11:523. [PMID: 22185343 PMCID: PMC3292516 DOI: 10.1186/1471-2407-11-523] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 12/20/2011] [Indexed: 11/10/2022] Open
Abstract
Background Clinical outcome of patients with high-grade ccRCC (clear cell renal cell carcinoma) remains still poor despite recent advances in treatment strategies. Molecular mechanism of pathogenesis in developing high-grade ccRCC must be clarified. In the present study, we found that SAV1 was significantly downregulated with copy number loss in high-grade ccRCCs. Therefore, we investigated the SAV1 function on cell proliferation and apoptosis in vitro. Furthermore, we attempted to clarify the downstream signaling which is regulated by SAV1. Methods We performed array CGH and gene expression analysis of 8 RCC cell lines (786-O, 769-P, KMRC-1, KMRC-2, KMRC-3, KMRC-20, TUHR4TKB, and Caki-2), and expression level of mRNA was confirmed by quantitative RT-PCR (qRT-PCR) analysis. We next re-expressed SAV1 in 786-O cells, and analyzed its colony-forming activity. Then, we transfected siRNAs of SAV1 into the kidney epithelial cell line HK2 and renal proximal tubule epithelial cells (RPTECs), and analyzed their proliferation and apoptosis. Furthermore, the activity of YAP1, which is a downstream molecule of SAV1, was evaluated by western blot analysis, reporter assay and immunohistochemical analysis. Results We found that SAV1, a component of the Hippo pathway, is frequently downregulated in high-grade ccRCC. SAV1 is located on chromosome 14q22.1, where copy number loss had been observed in 7 of 12 high-grade ccRCCs in our previous study, suggesting that gene copy number loss is responsible for the downregulation of SAV1. Colony-forming activity by 786-O cells, which show homozygous loss of SAV1, was significantly reduced when SAV1 was re-introduced exogenously. Knockdown of SAV1 promoted proliferation of HK2 and RPTEC. Although the phosphorylation level of YAP1 was low in 786-O cells, it was elevated in SAV1-transduced 786-O cells. Furthermore, the transcriptional activity of the YAP1 and TEAD3 complex was inhibited in SAV1-transduced 786-O cells. Immunohistochemistry frequently demonstrated nuclear localization of YAP1 in ccRCC cases with SAV1 downregulation, and it was preferentially detected in high-grade ccRCC. Conclusions Taken together, downregulation of SAV1 and the consequent YAP1 activation are involved in the pathogenesis of high-grade ccRCC. It is an attractive hypothesis that Hippo signaling could be candidates for new therapeutic target.
Collapse
Affiliation(s)
- Keiko Matsuura
- Department of Molecular Pathology, Oita University, Oita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Mohamed JS, Boriek AM. Loss of desmin triggers mechanosensitivity and up-regulation of Ankrd1 expression through Akt-NF-κB signaling pathway in smooth muscle cells. FASEB J 2011; 26:757-65. [PMID: 22085644 DOI: 10.1096/fj.10-160291] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Muscle cells, including human airway smooth muscle cells (HASMCs) express ankyrin repeat protein 1 (Ankrd1), a member of ankyrin repeat protein family. Ankrd1 efficiently interacts with the type III intermediate filament desmin. Our earlier study showed that desmin is an intracellular load-bearing protein that influences airway compliance, lung recoil, and airway contractile responsiveness. These results suggest that Ankrd1 and desmin may play important roles on ASMC homeostasis. Here we show that small interfering (si)RNA-mediated knockdown of the desmin gene in HASMCs, recombinant HASMCs (reHASMCs), up-regulates Ankrd1 expression. Moreover, loss of desmin in HASMCs increases the phosphorylation of Akt, inhibitor of κB kinase (IKK)-α, and inhibitor of κB (IκB)-α proteins, leading to NF-κB activation. Treatment of reHASMCs with Akt, IKKα, IκBα, or NF-κB inhibitor inhibits the loss of desmin-induced Ankrd1 up-regulation, suggesting Akt/NF-κB-mediated Ankrd1 regulation. Transfection of reHASMCs with siRNA specific for p50 or p65 corroborates the NF-κB-mediated Ankrd1 regulation. Luciferase reporter assays show that NF-κB directly binds on Ankrd1 promoter and up-regulates Ankrd1 levels. Overall, our data provide a new link between desmin and Ankrd1 regulation, which may be important for ASMC homeostasis.
Collapse
Affiliation(s)
- Junaith S Mohamed
- Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
16
|
Laure L, Suel L, Roudaut C, Bourg N, Ouali A, Bartoli M, Richard I, Danièle N. Cardiac ankyrin repeat protein is a marker of skeletal muscle pathological remodelling. FEBS J 2009; 276:669-84. [PMID: 19143834 DOI: 10.1111/j.1742-4658.2008.06814.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In an attempt to identify potential therapeutic targets for the correction of muscle wasting, the gene expression of several pivotal proteins involved in protein metabolism was investigated in experimental atrophy induced by transient or definitive denervation, as well as in four animal models of muscular dystrophies (deficient for calpain 3, dysferlin, alpha-sarcoglycan and dystrophin, respectively). The results showed that: (a) the components of the ubiquitin-proteasome pathway are upregulated during the very early phases of atrophy but do not greatly increase in the muscular dystrophy models; (b) forkhead box protein O1 mRNA expression is augmented in the muscles of a limb girdle muscular dystrophy 2A murine model; and (c) the expression of cardiac ankyrin repeat protein (CARP), a regulator of transcription factors, appears to be persistently upregulated in every condition, suggesting that CARP could be a hub protein participating in common pathological molecular pathway(s). Interestingly, the mRNA level of a cell cycle inhibitor known to be upregulated by CARP in other tissues, p21(WAF1/CIP1), is consistently increased whenever CARP is upregulated. CARP overexpression in muscle fibres fails to affect their calibre, indicating that CARP per se cannot initiate atrophy. However, a switch towards fast-twitch fibres is observed, suggesting that CARP plays a role in skeletal muscle plasticity. The observation that p21(WAF1/CIP1) is upregulated, put in perspective with the effects of CARP on the fibre type, fits well with the idea that the mechanisms at stake might be required to oppose muscle remodelling in skeletal muscle.
Collapse
|