1
|
Busselman BW, Ratnayake I, Terasaki MR, Thakkar VP, Ilyas A, Otterpohl KL, Zimmerman JL, Chandrasekar I. Actin cytoskeleton and associated myosin motors within the renal epithelium. Am J Physiol Renal Physiol 2024; 327:F553-F565. [PMID: 39052845 PMCID: PMC11483076 DOI: 10.1152/ajprenal.00078.2024] [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: 03/07/2024] [Revised: 07/09/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
This review highlights the complexity of renal epithelial cell membrane architectures and organelles through careful review of ultrastructural and physiological studies published over the past several decades. We also showcase the vital roles played by the actin cytoskeleton and actin-associated myosin motor proteins in regulating cell type-specific physiological functions within the cells of the renal epithelium. The purpose of this review is to provide a fresh conceptual framework to explain the structure-function relationships that exist between the actin cytoskeleton, organelle structure, and cargo transport within the mammalian kidney. With recent advances in technologies to visualize the actin cytoskeleton and associated proteins within intact kidneys, it has become increasingly imperative to reimagine the functional roles of these proteins in situ to provide a rationale for their unique, cell type-specific functions that are necessary to establish and maintain complex physiological processes.
Collapse
Affiliation(s)
- Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | | | - Mark R Terasaki
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| | - Vedant P Thakkar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Arooba Ilyas
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, United States
| | - Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Jenna L Zimmerman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, United States
- Department of Cell Biology, University of Connecticut, Farmington, Connecticut, United States
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States
| |
Collapse
|
2
|
Shen K, Chen T, Xiao M. MYH9-related inherited thrombocytopenia: the genetic spectrum, underlying mechanisms, clinical phenotypes, diagnosis, and management approaches. Res Pract Thromb Haemost 2024; 8:102552. [PMID: 39309229 PMCID: PMC11415342 DOI: 10.1016/j.rpth.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 09/25/2024] Open
Abstract
Inherited thrombocytopenias have been considered exceedingly rare for a long time, but recent advances have facilitated diagnosis and greatly enabled the discovery of new causative genes. MYH9-related disease (MYH9-RD) represents one of the most frequent forms of inherited thrombocytopenia, usually presenting with nonspecific clinical manifestations, which renders it difficult to establish an accurate diagnosis. MYH9-RD is an autosomal dominant-inherited thrombocytopenia caused by deleterious variants in the MYH9 gene encoding the heavy chain of nonmuscle myosin IIA. Patients with MYH9-RD usually present with thrombocytopenia and platelet macrocytosis at birth or in infancy, and most of them may develop one or more extrahematologic manifestations of progressive nephritis, sensorial hearing loss, presenile cataracts, and elevated liver enzymatic levels during childhood and adult life. Here, we have reviewed recent advances in the study of MYH9-RD, which aims to provide an updated and comprehensive summary of the current knowledge and improve our understanding of the genetic spectrum, underlying mechanisms, clinical phenotypes, diagnosis, and management approaches of this rare disease. Importantly, our goal is to enable physicians to better understand this rare disease and highlight the critical role of genetic etiologic analysis in ensuring accurate diagnosis, clinical management, and genetic counseling while avoiding ineffective and potentially harmful therapies for MYH9-RD patients.
Collapse
Affiliation(s)
- Kefeng Shen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Chen
- Department of Ophthalmology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Ye Z, Xu J, Zhang X, Zhang Y, Ivanova D, Lu W, Zhang J, Li F, Chen X, Wang Y, Wang M, Xie B. Identification and Validation of Tumor Microenvironment-Associated Signature in Clear-Cell Renal Cell Carcinoma through Integration of DNA Methylation and Gene Expression. Int J Mol Sci 2024; 25:6792. [PMID: 38928496 PMCID: PMC11203551 DOI: 10.3390/ijms25126792] [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: 05/17/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
The tumor microenvironment (TME) is crucial in tumor development, metastasis, and response to immunotherapy. DNA methylation can regulate the TME without altering the DNA sequence. However, research on the methylation-driven TME in clear-cell renal cell carcinoma (ccRCC) is still lacking. In this study, integrated DNA methylation and RNA-seq data were used to explore methylation-driven genes (MDGs). Immune scores were calculated using the ESTIMATE, which was employed to identify TME-related genes. A new signature connected with methylation-regulated TME using univariate, multivariate Cox regression and LASSO regression analyses was developed. This signature consists of four TME-MDGs, including AJAP1, HOXB9, MYH14, and SLC6A19, which exhibit high methylation and low expression in tumors. Validation was performed using qRT-PCR which confirmed their downregulation in ccRCC clinical samples. Additionally, the signature demonstrated stable predictive performance in different subtypes of ccRCC. Risk scores are positively correlated with TMN stages, immune cell infiltration, tumor mutation burden, and adverse outcomes of immunotherapy. Interestingly, the expression of four TME-MDGs are highly correlated with the sensitivity of first-line drugs in ccRCC treatment, especially pazopanib. Molecular docking indicates a high affinity binding between the proteins and pazopanib. In summary, our study elucidates the comprehensive role of methylation-driven TME in ccRCC, aiding in identifying patients sensitive to immunotherapy and targeted therapy, and providing new therapeutic targets for ccRCC treatment.
Collapse
Affiliation(s)
- Zijian Ye
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jialiang Xu
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Xin Zhang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
| | - Yifan Zhang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Deyana Ivanova
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Weiyu Lu
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
| | - Jianning Zhang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Meijiao Wang
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing 400016, China (J.X.); (X.Z.); (Y.Z.); (W.L.); (J.Z.)
- Joint International Research Laboratory of Reproduction, Development of the Ministry of Education of China, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China; (F.L.); (X.C.); (Y.W.)
| | - Biao Xie
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
4
|
Krajewska M, Możajew M, Filipek S, Koprowski P. Interaction of ROMK2 channel with lipid kinases DGKE and AGK: Potential channel activation by localized anionic lipid synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159443. [PMID: 38056763 DOI: 10.1016/j.bbalip.2023.159443] [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: 08/28/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
In this study, we utilized enzyme-catalyzed proximity labeling with the engineered promiscuous biotin ligase Turbo-ID to identify the proxisome of the ROMK2 channel. This channel resides in various cellular membrane compartments of the cell including the plasma membrane, endoplasmic reticulum and mitochondria. Within mitochondria, ROMK2 has been suggested as a pore-forming subunit of mitochondrial ATP-regulated potassium channel (mitoKATP). We found that ROMK2 proxisome in addition to previously known protein partners included two lipid kinases: acylglycerol kinase (AGK) and diacylglycerol kinase ε (DGKE), which are localized in mitochondria and the endoplasmic reticulum, respectively. Through co-immunoprecipitation, we confirmed that these two kinases are present in complexes with ROMK2 channels. Additionally, we found that the products of AGK and DGKE, lysophosphatidic acid (LPA) and phosphatidic acid (PA), stimulated the activity of ROMK2 channels in artificial lipid bilayers. Our molecular docking studies revealed the presence of acidic lipid binding sites in the ROMK2 channel, similar to those previously identified in Kir2 channels. Based on these findings, we propose a model wherein localized lipid synthesis, mediated by channel-bound lipid kinases, contributes to the regulation of ROMK2 activity within distinct intracellular compartments, such as mitochondria and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Milena Krajewska
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Mariusz Możajew
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology PAS, Warsaw, Poland; Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Sławomir Filipek
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Piotr Koprowski
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology PAS, Warsaw, Poland.
| |
Collapse
|
5
|
Abstract
Non-muscle myosin 2 (NM2) motors are the major contractile machines in most cell types. Unsurprisingly, these ubiquitously expressed actin-based motors power a plethora of subcellular, cellular and multicellular processes. In this Cell Science at a Glance article and the accompanying poster, we review the biochemical properties and mechanisms of regulation of this myosin. We highlight the central role of NM2 in multiple fundamental cellular processes, which include cell migration, cytokinesis, epithelial barrier function and tissue morphogenesis. In addition, we highlight recent studies using advanced imaging technologies that have revealed aspects of NM2 assembly hitherto inaccessible. This article will hopefully appeal to both cytoskeletal enthusiasts and investigators from outside the cytoskeleton field who have interests in one of the many basic cellular processes requiring actomyosin force production.
Collapse
Affiliation(s)
- Melissa A. Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60525, USA
| | - John A. Hammer
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jordan R. Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60525, USA
| |
Collapse
|
6
|
Wu CT, Lidsky PV, Xiao Y, Cheng R, Lee IT, Nakayama T, Jiang S, He W, Demeter J, Knight MG, Turn RE, Rojas-Hernandez LS, Ye C, Chiem K, Shon J, Martinez-Sobrido L, Bertozzi CR, Nolan GP, Nayak JV, Milla C, Andino R, Jackson PK. SARS-CoV-2 replication in airway epithelia requires motile cilia and microvillar reprogramming. Cell 2023; 186:112-130.e20. [PMID: 36580912 PMCID: PMC9715480 DOI: 10.1016/j.cell.2022.11.030] [Citation(s) in RCA: 74] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 09/15/2022] [Accepted: 11/23/2022] [Indexed: 12/04/2022]
Abstract
How SARS-CoV-2 penetrates the airway barrier of mucus and periciliary mucins to infect nasal epithelium remains unclear. Using primary nasal epithelial organoid cultures, we found that the virus attaches to motile cilia via the ACE2 receptor. SARS-CoV-2 traverses the mucus layer, using motile cilia as tracks to access the cell body. Depleting cilia blocks infection for SARS-CoV-2 and other respiratory viruses. SARS-CoV-2 progeny attach to airway microvilli 24 h post-infection and trigger formation of apically extended and highly branched microvilli that organize viral egress from the microvilli back into the mucus layer, supporting a model of virus dispersion throughout airway tissue via mucociliary transport. Phosphoproteomics and kinase inhibition reveal that microvillar remodeling is regulated by p21-activated kinases (PAK). Importantly, Omicron variants bind with higher affinity to motile cilia and show accelerated viral entry. Our work suggests that motile cilia, microvilli, and mucociliary-dependent mucus flow are critical for efficient virus replication in nasal epithelia.
Collapse
Affiliation(s)
- Chien-Ting Wu
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Center for Clinical Sciences Research, 269 Campus Drive, Stanford, CA, USA
| | - Peter V Lidsky
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16th Street, Room S572E, Box 2280, San Francisco, CA, USA
| | - Yinghong Xiao
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16th Street, Room S572E, Box 2280, San Francisco, CA, USA
| | - Ran Cheng
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Center for Clinical Sciences Research, 269 Campus Drive, Stanford, CA, USA; Department of Biology, Stanford University, Stanford, CA, USA
| | - Ivan T Lee
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tsuguhisa Nakayama
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Sizun Jiang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Wei He
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Center for Clinical Sciences Research, 269 Campus Drive, Stanford, CA, USA
| | - Janos Demeter
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Center for Clinical Sciences Research, 269 Campus Drive, Stanford, CA, USA
| | - Miguel G Knight
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16th Street, Room S572E, Box 2280, San Francisco, CA, USA
| | - Rachel E Turn
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Center for Clinical Sciences Research, 269 Campus Drive, Stanford, CA, USA
| | - Laura S Rojas-Hernandez
- Department of Pediatric Pulmonary Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Chengjin Ye
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Kevin Chiem
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Judy Shon
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Luis Martinez-Sobrido
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jayakar V Nayak
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Department of Otolaryngology, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Carlos Milla
- Department of Pediatric Pulmonary Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, 600 16th Street, Room S572E, Box 2280, San Francisco, CA, USA.
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Center for Clinical Sciences Research, 269 Campus Drive, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
7
|
Hu Y, Yang C, Amorim T, Maqbool M, Lin J, Li C, Fang C, Xue L, Kwart A, Fang H, Yin M, Janocha AJ, Tsuchimoto D, Nakabeppu Y, Jiang X, Mejia-Garcia A, Anwer F, Khouri J, Qi X, Zheng QY, Yu JS, Yan S, LaFramboise T, Anderson KC, Herlitz LC, Munshi NC, Lin J, Zhao J. Cisplatin-Mediated Upregulation of APE2 Binding to MYH9 Provokes Mitochondrial Fragmentation and Acute Kidney Injury. Cancer Res 2021; 81:713-723. [PMID: 33288657 PMCID: PMC7869671 DOI: 10.1158/0008-5472.can-20-1010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/29/2020] [Accepted: 12/02/2020] [Indexed: 11/16/2022]
Abstract
Cisplatin chemotherapy is standard care for many cancers but is toxic to the kidneys. How this toxicity occurs is uncertain. In this study, we identified apurinic/apyrimidinic endonuclease 2 (APE2) as a critical molecule upregulated in the proximal tubule cells (PTC) following cisplatin-induced nuclear DNA and mitochondrial DNA damage in cisplatin-treated C57B6J mice. The APE2 transgenic mouse phenotype recapitulated the pathophysiological features of C-AKI (acute kidney injury, AKI) in the absence of cisplatin treatment. APE2 pulldown-MS analysis revealed that APE2 binds myosin heavy-Chain 9 (MYH9) protein in mitochondria after cisplatin treatment. Human MYH9-related disorder is caused by mutations in MYH9 that eventually lead to nephritis, macrothrombocytopenia, and deafness, a constellation of symptoms similar to the toxicity profile of cisplatin. Moreover, cisplatin-induced C-AKI was attenuated in APE2-knockout mice. Taken together, these findings suggest that cisplatin promotes AKI development by upregulating APE2, which leads to subsequent MYH9 dysfunction in PTC mitochondria due to an unrelated role of APE2 in DNA damage repair. This postulated mechanism and the availability of an engineered transgenic mouse model based on the mechanism of C-AKI provides an opportunity to identify novel targets for prophylactic treatment of this serious disease. SIGNIFICANCE: These results reveal and highlight an unexpected role of APE2 via its interaction with MYH9 and suggest that APE2 has the potential to prevent acute kidney injury in patients with cisplatin-treated cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/3/713/F1.large.jpg.
Collapse
Affiliation(s)
- Yi Hu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Chun Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Clinical Laboratory, the 4th Hospital of Harbin Medical University, Harbin, China
| | - Tania Amorim
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mohsin Maqbool
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jenny Lin
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Chen Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei, China
| | - Chuanfeng Fang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Clinical Laboratory, the 4th Hospital of Harbin Medical University, Harbin, China
| | - Li Xue
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Department of Clinical Laboratory, the 4th Hospital of Harbin Medical University, Harbin, China
| | - Ariel Kwart
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Division of Hand Surgery, Department of Orthopedic Surgery, NYU Langone Orthopedic Hospital, New York, New York
| | - Hua Fang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mei Yin
- Image Core, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Allison J Janocha
- The Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Daisuke Tsuchimoto
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Japan
| | - Yusaku Nakabeppu
- Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Japan
| | - Xiaofeng Jiang
- Department of Clinical Laboratory, the 4th Hospital of Harbin Medical University, Harbin, China
| | - Alex Mejia-Garcia
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Faiz Anwer
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Xin Qi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Qing Y Zheng
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University, Cleveland, Ohio
| | - Jennifer S Yu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Shan Yan
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina
| | - Thomas LaFramboise
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Leal C Herlitz
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Nikhil C Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- VA Boston Healthcare System, Boston, Massachusetts
| | - Jianhong Lin
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio.
| | - Jianjun Zhao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
8
|
Otterpohl KL, Busselman BW, Ratnayake I, Hart RG, Hart KR, Evans CM, Phillips CL, Beach JR, Ahrenkiel P, Molitoris BA, Surendran K, Chandrasekar I. Conditional Myh9 and Myh10 inactivation in adult mouse renal epithelium results in progressive kidney disease. JCI Insight 2020; 5:138530. [PMID: 33001861 PMCID: PMC7710296 DOI: 10.1172/jci.insight.138530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/23/2020] [Indexed: 01/07/2023] Open
Abstract
Actin-associated nonmuscle myosin II (NM2) motor proteins play critical roles in a myriad of cellular functions, including endocytosis and organelle transport pathways. Cell type–specific expression and unique subcellular localization of the NM2 proteins, encoded by the Myh9 and Myh10 genes, in the mouse kidney tubules led us to hypothesize that these proteins have specialized functional roles within the renal epithelium. Inducible conditional knockout (cKO) of Myh9 and Myh10 in the renal tubules of adult mice resulted in progressive kidney disease. Prior to overt renal tubular injury, we observed intracellular accumulation of the glycosylphosphatidylinositol-anchored protein uromodulin (UMOD) and gradual loss of Na+ K+ 2Cl– cotransporter from the apical membrane of the thick ascending limb epithelia. The UMOD accumulation coincided with expansion of endoplasmic reticulum (ER) tubules and activation of ER stress and unfolded protein response pathways in Myh9&10-cKO kidneys. We conclude that NM2 proteins are required for localization and transport of UMOD and loss of function results in accumulation of UMOD and ER stress–mediated progressive renal tubulointerstitial disease. These observations establish cell type–specific role(s) for NM2 proteins in regulation of specialized renal epithelial transport pathways and reveal the possibility that human kidney disease associated with MYH9 mutations could be of renal epithelial origin. Adult mouse renal epithelium specific knockout of Myh9 and Myh10 genes result in kidney disease.
Collapse
Affiliation(s)
- Karla L Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Brook W Busselman
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA.,Basic Biomedical Sciences Graduate Program, University of South Dakota, Vermillion, South Dakota, USA
| | - Ishara Ratnayake
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota, USA
| | - Ryan G Hart
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Kimberly R Hart
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Claire M Evans
- Histology and Imaging Core, Sanford Research, Sioux Falls, South Dakota, USA
| | - Carrie L Phillips
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jordan R Beach
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA
| | - Phil Ahrenkiel
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota, USA
| | - Bruce A Molitoris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kameswaran Surendran
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA.,Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, USA
| |
Collapse
|
9
|
Rudman-Melnick V, Adam M, Potter A, Chokshi SM, Ma Q, Drake KA, Schuh MP, Kofron JM, Devarajan P, Potter SS. Single-Cell Profiling of AKI in a Murine Model Reveals Novel Transcriptional Signatures, Profibrotic Phenotype, and Epithelial-to-Stromal Crosstalk. J Am Soc Nephrol 2020; 31:2793-2814. [PMID: 33115917 DOI: 10.1681/asn.2020010052] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Current management of AKI, a potentially fatal disorder that can also initiate or exacerbate CKD, is merely supportive. Therefore, deeper understanding of the molecular pathways perturbed in AKI is needed to identify targets with potential to lead to improved treatment. METHODS We performed single-cell RNA sequencing (scRNA-seq) with the clinically relevant unilateral ischemia-reperfusion murine model of AKI at days 1, 2, 4, 7, 11, and 14 after AKI onset. Using real-time quantitative PCR, immunofluorescence, Western blotting, and both chromogenic and single-molecule in situ hybridizations, we validated AKI signatures in multiple experiments. RESULTS Our findings show the time course of changing gene expression patterns for multiple AKI stages and all renal cell types. We observed elevated expression of crucial injury response factors-including kidney injury molecule-1 (Kim1), lipocalin 2 (Lcn2), and keratin 8 (Krt8)-and of several novel genes (Ahnak, Sh3bgrl3, and Col18a1) not previously examined in kidney pathologies. AKI induced proximal tubule dedifferentiation, with a pronounced nephrogenic signature represented by Sox4 and Cd24a. Moreover, AKI caused the formation of "mixed-identity cells" (expressing markers of different renal cell types) that are normally seen only during early kidney development. The injured tubules acquired a proinflammatory and profibrotic phenotype; moreover, AKI dramatically modified ligand-receptor crosstalk, with potential pathologic epithelial-to-stromal interactions. Advancing age in AKI onset was associated with maladaptive response and kidney fibrosis. CONCLUSIONS The scRNA-seq, comprehensive, cell-specific profiles provide a valuable resource for examining molecular pathways that are perturbed in AKI. The results fully define AKI-associated dedifferentiation programs, potential pathologic ligand-receptor crosstalk, novel genes, and the improved injury response in younger mice, and highlight potential targets of kidney injury.
Collapse
Affiliation(s)
- Valeria Rudman-Melnick
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Andrew Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Saagar M Chokshi
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Meredith P Schuh
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Medical Center, Cincinnati, Ohio
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio
| |
Collapse
|
10
|
Trendowski MR, El-Charif O, Ratain MJ, Monahan P, Mu Z, Wheeler HE, Dinh PC, Feldman DR, Ardeshir-Rouhani-Fard S, Hamilton RJ, Vaughn DJ, Fung C, Kollmannsberger C, Mushiroda T, Kubo M, Hannigan R, Strathmann F, Einhorn LH, Fossa SD, Travis LB, Dolan ME. Clinical and Genome-Wide Analysis of Serum Platinum Levels after Cisplatin-Based Chemotherapy. Clin Cancer Res 2019; 25:5913-5924. [PMID: 31296530 PMCID: PMC6774840 DOI: 10.1158/1078-0432.ccr-19-0113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/17/2019] [Accepted: 07/09/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Serum platinum is measurable for years after completion of cisplatin-based chemotherapy (CBC). We report the largest investigation of serum platinum levels to date of 1,010 testicular cancer survivors (TCS) assessed 1-35 years after CBC and evaluate genetic contributions to these levels. EXPERIMENTAL DESIGN Eligible TCS given 300 or 400 (±15) mg/m2 cisplatin underwent extensive audiometric testing, clinical examination, completed questionnaires, and had crude serum platinum levels measured. Associations between serum platinum and various risk factors and toxicities were assessed after fitting a biexponential model adjusted for follow-up time and cumulative cisplatin dose. A genome-wide association study (GWAS) was performed using the serum platinum residuals of the dose and time-adjusted model. RESULTS Serum platinum levels exceeded the reference range for approximately 31 years, with a strong inverse relationship with creatinine clearance at follow-up (age-adjusted P = 2.13 × 10-3). We observed a significant, positive association between residual platinum values and luteinizing hormone (age-adjusted P = 6.58 × 10-3). Patients with high residual platinum levels experienced greater Raynaud phenomenon than those with medium or low levels (age-adjusted ORhigh/low = 1.46; P = 0.04), as well as a higher likelihood of developing tinnitus (age-adjusted ORhigh/low = 1.68, P = 0.07). GWAS identified one single-nucleotide polymorphism (SNP) meeting genome-wide significance, rs1377817 (P = 4.6 × 10-8, a SNP intronic to MYH14). CONCLUSIONS This study indicates that residual platinum values are correlated with several cisplatin-related toxicities. One genetic variant is associated with these levels.
Collapse
Affiliation(s)
| | - Omar El-Charif
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Mark J Ratain
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Patrick Monahan
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Zepeng Mu
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Heather E Wheeler
- Departments of Biology and Computer Science, Loyola University Chicago, Chicago, Illinois
| | - Paul C Dinh
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Darren R Feldman
- Department of Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | - Robert J Hamilton
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David J Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | | | | | - Michiaki Kubo
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | - Robyn Hannigan
- School for the Environment, University of Massachusetts Boston, Boston, Massachusetts
| | | | - Lawrence H Einhorn
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana
| | - Sophie D Fossa
- Department of Oncology, Oslo University Hospital, Radiumhospital, Oslo, Norway
| | - Lois B Travis
- Department of Medical Oncology, Indiana University, Indianapolis, Indiana.
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois.
| |
Collapse
|
11
|
Fernandez-Prado R, Carriazo-Julio SM, Torra R, Ortiz A, Perez-Gomez MV. MYH9-related disease: it does exist, may be more frequent than you think and requires specific therapy. Clin Kidney J 2019; 12:488-493. [PMID: 31384439 PMCID: PMC6671427 DOI: 10.1093/ckj/sfz103] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
In this issue of ckj, Tabibzadeh et al. report one of the largest series of patients with MYH9 mutations and kidney disease. The cardinal manifestation of MYH9-related disease is thrombocytopenia with giant platelets. The population frequency of pathogenic MYH9 mutations may be at least 1 in 20 000. The literature abounds in misdiagnosed cases treated for idiopathic thrombocytopenic purpura with immune suppressants and even splenectomy. Additional manifestations include neurosensorial deafness and proteinuric and hematuric progressive kidney disease (at some point, it was called Alport syndrome with macrothrombocytopenia), leucocyte inclusions, cataracts and liver enzyme abnormalities, resulting in different names for different manifestation combinations (MATINS, May-Hegglin anomaly, Fechtner, Epstein and Sebastian syndromes, and deafness AD 17). The penetrance and severity of kidney disease are very variable, which may obscure the autosomal dominant inheritance. A correct diagnosis will both preclude unnecessary and potentially dangerous therapeutic interventions and allow genetic counselling and adequate treatment. Morphological erythrocyte, granulocyte and platelet abnormalities may allow the future development of high-throughput screening techniques adapted to clinical peripheral blood flow cytometers.
Collapse
Affiliation(s)
- Raul Fernandez-Prado
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- REDinREN, Instituto de Investigación Carlos III, Madrid, Spain
| | - Sol Maria Carriazo-Julio
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- REDinREN, Instituto de Investigación Carlos III, Madrid, Spain
| | - Roser Torra
- REDinREN, Instituto de Investigación Carlos III, Madrid, Spain
- Nephrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- REDinREN, Instituto de Investigación Carlos III, Madrid, Spain
| | - María Vanessa Perez-Gomez
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz UAM, Madrid, Spain
- REDinREN, Instituto de Investigación Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Crajoinas RO, Polidoro JZ, Girardi ACC. The potential role of myosin motor proteins in mediating the subcellular distribution of NHE3 in the renal proximal tubule. Am J Physiol Renal Physiol 2019; 316:F986-F992. [PMID: 30864843 DOI: 10.1152/ajprenal.00577.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Isoform 3 of the Na+/H+ exchanger (NHE3) is responsible for the majority of the reabsorption of NaCl, NaHCO3, and, consequently, water in the renal proximal tubule. As such, this transporter plays an essential role in acid-base balance and extracellular fluid volume homeostasis and determining systemic arterial blood pressure levels. NHE3 activity is modulated by a number of mechanisms, including the redistribution of the transporter between the body of the microvilli (where NHE3 is active) and the base of the microvilli (where NHE3 is less active). Although the physiological, pathophysiological, and pharmacological importance of the subcellular distribution of NHE3 has been well established, the exact mechanism whereby NHE3 is translocated along microvilli microdomains of the proximal tubule apical membrane is unknown. Nonmuscle myosin IIA and unconventional myosin VI move cargoes in anterograde and retrograde directions, respectively, and are known to redistribute along with NHE3 in the proximal tubule in response to a variety of natriuretic and antinatriuretic stimuli, including stimulation or inhibition of the renin-angiotensin system, high dietary Na+ intake, and high blood pressure. Therefore, this review aims to discuss the current evidence that suggests a potential role of myosin IIA and myosin VI in mediating the subcellular distribution of NHE3 along the kidney proximal tubule microvilli and their possible contribution in modifying NHE3-mediated Na+ reabsorption under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Renato O Crajoinas
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| | - Juliano Z Polidoro
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| | - Adriana C C Girardi
- Heart Institute (InCor), University of São Paulo Medical School , São Paulo , Brazil
| |
Collapse
|
13
|
Otterpohl K, Busselman B, Hart R, Shan K, Lin B, Surendran K, Chandrasekar I. The apical membrane localization and trafficking of uromodulin (UMOD) is critically dependent on nonmuscle myosin II isoforms Myh9 and Myh10 in mouse thick ascending limb epithelial cells. FASEB J 2019. [DOI: 10.1096/fasebj.2019.33.1_supplement.862.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
| | | | - Ryan Hart
- Enabling Technologies GroupSanford ResearchSioux FallsSD
| | - Kevin Shan
- Enabling Technologies GroupSanford ResearchSioux FallsSD
| | - Brendon Lin
- Enabling Technologies GroupSanford ResearchSioux FallsSD
| | | | | |
Collapse
|
14
|
Otterpohl KL, Hart RG, Evans C, Surendran K, Chandrasekar I. Nonmuscle myosin 2 proteins encoded by Myh9, Myh10, and Myh14 are uniquely distributed in the tubular segments of murine kidney. Physiol Rep 2018; 5. [PMID: 29208685 PMCID: PMC5727274 DOI: 10.14814/phy2.13513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 11/24/2022] Open
Abstract
The diverse epithelial cell types of the kidneys are segregated into nephron segments and the collecting ducts in order to endow each tubular segment with unique functions. The rich diversity of the epithelial cell types is highlighted by the unique membrane channels and receptors expressed within each nephron segment. Our previous work identified a critical role for Myh9 and Myh10 in mammalian endocytosis. Here, we examined the expression patterns of Nonmuscle myosin 2 (NM2) heavy chains encoded by Myh9, Myh10, and Myh14 in mouse kidneys as these genes may confer unique nephron segment‐specific membrane transport properties. Interestingly, we found that each segment of the renal tubules predominately expressed only two of the three NM2 isoforms, with isoform‐specific subcellular localization, and different levels of expression within a nephron segment. Additionally, we identify Myh14 to be restricted to the intercalated cells and Myh10 to be restricted to the principal cells within the collecting ducts and connecting segments. We speculate that the distinct expression pattern of the NM2 proteins likely reflects the diversity of the intracellular trafficking machinery present within the different renal tubular epithelial segments.
Collapse
Affiliation(s)
- Karla L Otterpohl
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA
| | - Ryan G Hart
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA
| | - Claire Evans
- Molecular Pathology Core, Sanford Research, Sioux Falls, South Dakota, USA
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group - Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, South Dakota, USA
| | - Indra Chandrasekar
- Enabling Technologies Group - Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, USD Sanford School of Medicine, Sioux Falls, South Dakota, USA
| |
Collapse
|