1
|
Cantu Gutierrez ME, Hill MC, Largoza GE, Gillespie WB, Martin JF, Wythe JD. Mapping the transcriptional and epigenetic landscape of organotypic endothelial diversity in the developing and adult mouse. NATURE CARDIOVASCULAR RESEARCH 2025; 4:473-495. [PMID: 40097733 PMCID: PMC12023908 DOI: 10.1038/s44161-025-00618-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/30/2025] [Indexed: 03/19/2025]
Abstract
The vascular endothelium features unique molecular and functional properties across different vessel types, such as between arteries, veins and capillaries, as well as between different organs, such as the leaky sinusoidal endothelium of the liver versus the impermeable vessels of the brain. However, the transcriptional networks governing endothelial organ specialization remain unclear. Here we profile the accessible chromatin and transcriptional landscapes of the endothelium from the mouse liver, lung, heart, kidney, brain and retina, across developmental time, to identify potential transcriptional regulators of endothelial heterogeneity. We then determine which of these putative regulators are conserved in human brain endothelial cells, and using single-cell transcriptomic profiling, we define which regulatory networks are active during brain maturation. Finally, we show that the putative transcriptional regulators identified by these three approaches molecularly and functionally reprogram naive endothelial cells. Thus, this resource can be used to identify potential transcriptional regulators controlling the establishment and maintenance of organ-specific endothelial specialization.
Collapse
Affiliation(s)
- Manuel E Cantu Gutierrez
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew C Hill
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gabrielle E Largoza
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - William B Gillespie
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Brain, Immunology, and Glia (BIG) Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - James F Martin
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Texas Heart Institute, Houston, TX, USA
| | - Joshua D Wythe
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA.
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Brain, Immunology, and Glia (BIG) Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Solovev I, Sergeeva A, Geraskina A, Shaposhnikov M, Vedunova M, Borysova O, Moskalev A. Aging and physiological barriers: mechanisms of barrier integrity changes and implications for age-related diseases. Mol Biol Rep 2024; 51:917. [PMID: 39158744 DOI: 10.1007/s11033-024-09833-7] [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: 03/28/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
The phenomenon of compartmentalization is one of the key traits of life. Biological membranes and histohematic barriers protect the internal environment of the cell and organism from endogenous and exogenous impacts. It is known that the integrity of these barriers decreases with age due to the loss of homeostasis, including age-related gene expression profile changes and the abnormal folding/assembly, crosslinking, and cleavage of barrier-forming macromolecules in addition to morphological changes in cells and tissues. The critical molecular and cellular mechanisms involved in physiological barrier integrity maintenance and aging-associated changes in their functioning are reviewed on different levels: molecular, organelle, cellular, tissue (histohematic, epithelial, and endothelial barriers), and organ one (skin). Biogerontology, which studies physiological barriers in the aspect of age, is still in its infancy; data are being accumulated, but there is no talk of the synthesis of complex theories yet. This paper mainly presents the mechanisms that will become targets of anti-aging therapy only in the future, possibly: pharmacological, cellular, and gene therapies, including potential geroprotectors, hormetins, senomorphic drugs, and senolytics.
Collapse
Affiliation(s)
- Ilya Solovev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st, Syktyvkar, 167982, Russian Federation
- Pitirim Sorokin Syktyvkar State University, 55 Oktyabrsky prosp, Syktyvkar, 167001, Russian Federation
| | - Alena Sergeeva
- Lobachevsky State University, Nizhny Novgorod, 603022, Russian Federation
| | | | - Mikhail Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st, Syktyvkar, 167982, Russian Federation
| | - Maria Vedunova
- Laboratory of genetics and epigenetics of aging, Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Moscow, 129226, Russian Federation
| | | | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 28 Kommunisticheskaya st, Syktyvkar, 167982, Russian Federation.
- Lobachevsky State University, Nizhny Novgorod, 603022, Russian Federation.
- Laboratory of genetics and epigenetics of aging, Russian Clinical Research Center for Gerontology, Pirogov Russian National Research Medical University, Moscow, 129226, Russian Federation.
| |
Collapse
|
3
|
Wang T, Li C, Wang X, Liu F. MAGI2 ameliorates podocyte apoptosis of diabetic kidney disease through communication with TGF-β-Smad3/nephrin pathway. FASEB J 2023; 37:e23305. [PMID: 37950637 DOI: 10.1096/fj.202301058r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/27/2023] [Indexed: 11/13/2023]
Abstract
Podocytes, the key component of the glomerular filtration barrier (GFB), are gradually lost during the progression of diabetic kidney disease (DKD), severely compromising kidney functionality. The molecular mechanisms regulating the survival of podocytes in DKD are incompletely understood. Here, we show that membrane-associated guanylate kinase inverted 2 (MAGI2) is specifically expressed in renal podocytes, and promotes podocyte survival in DKD. We found that MAGI2 expression was downregulated in podocytes cultured with high-glucose in vitro, and in kidneys of db/db mice as well as DKD patients. Conversely, we found enforced expression of MAGI2 via AAV transduction protected podocytes from apoptosis, with concomitant improvement of renal functions. Mechanistically, we found that MAGI2 deficiency induced by high glucose levels activates TGF-β signaling to decrease the expression of anti-apoptotic proteins. These results indicate that MAGI2 protects podocytes from cell death, and can be harnessed therapeutically to improve renal function in diabetic kidney disease.
Collapse
Affiliation(s)
- Tingli Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of medicine, University of Electronic Science and Technology of China, Chengdu, China
- West China Hospital, Sichuan University, Chengdu, China
| | - Xiaofei Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Fang Liu
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Wittenmayer N, Petkova-Tuffy A, Borgmeyer M, Lee C, Becker J, Böning A, Kügler S, Rhee J, Viotti JS, Dresbach T. S-SCAM is essential for synapse formation. Front Cell Neurosci 2023; 17:1182493. [PMID: 38045729 PMCID: PMC10690602 DOI: 10.3389/fncel.2023.1182493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 10/02/2023] [Indexed: 12/05/2023] Open
Abstract
Synapse formation is critical for the wiring of neural circuits in the developing brain. The synaptic scaffolding protein S-SCAM/MAGI-2 has important roles in the assembly of signaling complexes at post-synaptic densities. However, the role of S-SCAM in establishing the entire synapse is not known. Here, we report significant effects of RNAi-induced S-SCAM knockdown on the number of synapses in early stages of network development in vitro. In vivo knockdown during the first three postnatal weeks reduced the number of dendritic spines in the rat brain neocortex. Knockdown of S-SCAM in cultured hippocampal neurons severely reduced the clustering of both pre- and post-synaptic components. This included synaptic vesicle proteins, pre- and post-synaptic scaffolding proteins, and cell adhesion molecules, suggesting that entire synapses fail to form. Correspondingly, functional and morphological characteristics of developing neurons were affected by reducing S-SCAM protein levels; neurons displayed severely impaired synaptic transmission and reduced dendritic arborization. A next-generation sequencing approach showed normal expression of housekeeping genes but changes in expression levels in 39 synaptic signaling molecules in cultured neurons. These results indicate that S-SCAM mediates the recruitment of all key classes of synaptic molecules during synapse assembly and is critical for the development of neural circuits in the developing brain.
Collapse
Affiliation(s)
- Nina Wittenmayer
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
- Institute for Translational Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Andonia Petkova-Tuffy
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| | - Maximilian Borgmeyer
- Institute for Translational Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Chungku Lee
- Department of Molecular Neurobiology, Synaptic Physiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jürgen Becker
- Institute of Anatomy and Cell Biology, University Medical Center Göttingen, Göttingen, Germany
| | - Andreas Böning
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Synaptic Physiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Julio S. Viotti
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
- University of Bordeaux, CNRS, IINS, UMR 5297, Bordeaux, France
| | - Thomas Dresbach
- Institute of Anatomy and Embryology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Lingaas F, Tengvall K, Jansen JH, Pelander L, Hurst MH, Meuwissen T, Karlsson Å, Meadows JRS, Sundström E, Thoresen SI, Arnet EF, Guttersrud OA, Kierczak M, Hytönen MK, Lohi H, Hedhammar Å, Lindblad-Toh K, Wang C. Bayesian mixed model analysis uncovered 21 risk loci for chronic kidney disease in boxer dogs. PLoS Genet 2023; 19:e1010599. [PMID: 36693108 PMCID: PMC9897549 DOI: 10.1371/journal.pgen.1010599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/03/2023] [Accepted: 01/04/2023] [Indexed: 01/25/2023] Open
Abstract
Chronic kidney disease (CKD) affects 10% of the human population, with only a small fraction genetically defined. CKD is also common in dogs and has been diagnosed in nearly all breeds, but its genetic basis remains unclear. Here, we performed a Bayesian mixed model genome-wide association analysis for canine CKD in a boxer population of 117 canine cases and 137 controls, and identified 21 genetic regions associated with the disease. At the top markers from each CKD region, the cases carried an average of 20.2 risk alleles, significantly higher than controls (15.6 risk alleles). An ANOVA test showed that the 21 CKD regions together explained 57% of CKD phenotypic variation in the population. Based on whole genome sequencing data of 20 boxers, we identified 5,206 variants in LD with the top 50 BayesR markers. Following comparative analysis with human regulatory data, 17 putative regulatory variants were identified and tested with electrophoretic mobility shift assays. In total four variants, three intronic variants from the MAGI2 and GALNT18 genes, and one variant in an intergenic region on chr28, showed alternative binding ability for the risk and protective alleles in kidney cell lines. Many genes from the 21 CKD regions, RELN, MAGI2, FGFR2 and others, have been implicated in human kidney development or disease. The results from this study provide new information that may enlighten the etiology of CKD in both dogs and humans.
Collapse
Affiliation(s)
- Frode Lingaas
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Katarina Tengvall
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Johan Høgset Jansen
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Lena Pelander
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Theo Meuwissen
- Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Åsa Karlsson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jennifer R. S. Meadows
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Elisabeth Sundström
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Stein Istre Thoresen
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Ellen Frøysadal Arnet
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Ole Albert Guttersrud
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Marcin Kierczak
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Marjo K. Hytönen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Hannes Lohi
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Åke Hedhammar
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- * E-mail: (KL-T); (CW)
| | - Chao Wang
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- * E-mail: (KL-T); (CW)
| |
Collapse
|
6
|
Excoffon KJDA, Avila CL, Alghamri MS, Kolawole AO. The magic of MAGI-1: A scaffolding protein with multi signalosomes and functional plasticity. Biol Cell 2022; 114:185-198. [PMID: 35389514 DOI: 10.1111/boc.202200014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 11/29/2022]
Abstract
MAGI-1 is a critical cellular scaffolding protein with over 110 different cellular and microbial protein interactors. Since the discovery of MAGI-1 in 1997, MAGI-1 has been implicated in diverse cellular functions such as polarity, cell-cell communication, neurological processes, kidney function, and a host of diseases including cancer and microbial infection. Additionally, MAGI-1 has undergone nomenclature changes in response to the discovery of an additional PDZ domain, leading to lack of continuity in the literature. We address the nomenclature of MAGI-1 as well as summarize many of the critical functions of the known interactions. Given the importance of many of the interactors, such as human papillomavirus E6, the Coxsackievirus and adenovirus receptor (CAR), and PTEN, the enhancement or disruption of MAGI-based interactions has the potential to affect cellular functions that can potentially be harnessed as a therapeutic strategy for a variety of diseases.
Collapse
Affiliation(s)
| | - Christina L Avila
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Mahmoud S Alghamri
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| | - Abimbola O Kolawole
- Department of Biological Sciences, Wright State University, Dayton, Ohio, USA
| |
Collapse
|
7
|
Vo DHT, McGleave G, Overton IM. Immune Cell Networks Uncover Candidate Biomarkers of Melanoma Immunotherapy Response. J Pers Med 2022; 12:jpm12060958. [PMID: 35743743 PMCID: PMC9225330 DOI: 10.3390/jpm12060958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/30/2022] Open
Abstract
The therapeutic activation of antitumour immunity by immune checkpoint inhibitors (ICIs) is a significant advance in cancer medicine, not least due to the prospect of long-term remission. However, many patients are unresponsive to ICI therapy and may experience serious side effects; companion biomarkers are urgently needed to help inform ICI prescribing decisions. We present the IMMUNETS networks of gene coregulation in five key immune cell types and their application to interrogate control of nivolumab response in advanced melanoma cohorts. The results evidence a role for each of the IMMUNETS cell types in ICI response and in driving tumour clearance with independent cohorts from TCGA. As expected, ‘immune hot’ status, including T cell proliferation, correlates with response to first-line ICI therapy. Genes regulated in NK, dendritic, and B cells are the most prominent discriminators of nivolumab response in patients that had previously progressed on another ICI. Multivariate analysis controlling for tumour stage and age highlights CIITA and IKZF3 as candidate prognostic biomarkers. IMMUNETS provide a resource for network biology, enabling context-specific analysis of immune components in orthogonal datasets. Overall, our results illuminate the relationship between the tumour microenvironment and clinical trajectories, with potential implications for precision medicine.
Collapse
Affiliation(s)
- Duong H. T. Vo
- The Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (D.H.T.V.); (G.M.)
- Health Data Research Wales and Northern Ireland, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - Gerard McGleave
- The Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (D.H.T.V.); (G.M.)
- Health Data Research Wales and Northern Ireland, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - Ian M. Overton
- The Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (D.H.T.V.); (G.M.)
- Health Data Research Wales and Northern Ireland, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
- Correspondence:
| |
Collapse
|
8
|
Xiao M, Bohnert BN, Grahammer F, Artunc F. Rodent models to study sodium retention in experimental nephrotic syndrome. Acta Physiol (Oxf) 2022; 235:e13844. [PMID: 35569011 DOI: 10.1111/apha.13844] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
Abstract
Sodium retention and edema are hallmarks of nephrotic syndrome (NS). Different experimental rodent models have been established for simulating NS, however, not all of them feature sodium retention which requires proteinuria to exceed a certain threshold. In rats, puromycin aminonucleoside nephrosis (PAN) is a classic NS model introduced in 1955 that was adopted as doxorubicin-induced nephropathy (DIN) in 129S1/SvImJ mice. In recent years, mice with inducible podocin deletion (Nphs2Δipod ) or podocyte apoptosis (POD-ATTAC) have been developed. In these models, sodium retention is thought to be caused by activation of the epithelial sodium channel (ENaC) in the distal nephron through aberrantly filtered serine proteases or proteasuria. Strikingly, rodent NS models follow an identical chronological time course after the development of proteinuria featuring sodium retention within days and spontaneous reversal thereafter. In DIN and Nphs2Δipod mice, inhibition of ENaC by amiloride or urinary serine protease activity by aprotinin prevents sodium retention, opening up new and promising therapeutic approaches that could be translated into the treatment of nephrotic patients. However, the essential serine protease(s) responsible for ENaC activation is (are) still unknown. With the use of nephrotic rodent models, there is the possibility that this (these) will be identified in the future. This review summarizes the various rodent models used to study experimental nephrotic syndrome and the insights gained from these models with regard to the pathophysiology of sodium retention.
Collapse
Affiliation(s)
- Mengyun Xiao
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
| | - Bernhard N. Bohnert
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| | - Florian Grahammer
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Ferruh Artunc
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine University Hospital Tübingen Tübingen Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen Tübingen Germany
- German Center for Diabetes Research (DZD) at the University Tübingen Tübingen Germany
| |
Collapse
|
9
|
He Y, Liu H, Luo S, Amos CI, Lee JE, Li X, Nan H, Wei Q. Genetic variants of SDCCAG8 and MAGI2 in mitosis-related pathway genes are independent predictors of cutaneous melanoma-specific survival. Cancer Sci 2021; 112:4355-4364. [PMID: 34375487 PMCID: PMC8486203 DOI: 10.1111/cas.15102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 12/01/2022] Open
Abstract
Mitosis is a prognostic factor for cutaneous melanoma (CM), but accurate mitosis detection in CM tissues is difficult. Therefore, the 8th Edition of the American Joint Committee on Cancer staging system has removed the mitotic rate as a category criterion of the tumor T-category, based on the evidence that the mitotic rate was not an independent prognostic factor for melanoma survival. As single-nucleotide polymorphisms (SNPs) have been shown to be potential predictors for cutaneous melanoma-specific survival (CMSS), we investigated the potential prognostic value of SNPs in mitosis-related pathway genes in CMSS by analyzing their associations with outcomes of 850 CM patients from The University of Texas MD Anderson Cancer Center in a discovery dataset and validated the findings in another dataset of 409 CM patients from the Harvard University Nurses' Health Study and Health Professionals Follow-up Study. In both datasets, we identified two SNPs (SDCCAG8 rs10803138 G>A and MAGI2 rs3807694 C>T) as independent prognostic factors for CMSS, with adjusted allelic hazards ratios of 1.49 (95% confidence interval = 1.17-1.90, P = .001) and 1.45 (1.13-1.86, P = .003), respectively. Furthermore, their combined unfavorable alleles also predicted a poor survival in both discovery and validation datasets in a dose-response manner (Ptrend = .0006 and .0001, respectively). Additional functional analysis revealed that both SDCCAG8 rs10803138 A and MAGI2 rs3807694 T alleles were associated with elevated mRNA expression levels in normal tissues. Therefore, these findings suggest that SDCCAG8 rs10803138 G>A and MAGI2 rs3807694 C>T are independent prognostic biomarkers for CMSS, possibly by regulating the mRNA expression of the corresponding genes involved in mitosis.
Collapse
Affiliation(s)
- Yuanmin He
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
- Duke Cancer InstituteDuke University Medical CenterDurhamNCUSA
- Department of Population Health SciencesDuke University School of MedicineDurhamNCUSA
| | - Hongliang Liu
- Duke Cancer InstituteDuke University Medical CenterDurhamNCUSA
- Department of Population Health SciencesDuke University School of MedicineDurhamNCUSA
| | - Sheng Luo
- Department of Biostatistics and BioinformaticsDuke University School of MedicineDurhamNCUSA
| | - Christopher I. Amos
- Institute for Clinical and Translational ResearchBaylor College of MedicineHoustonTXUSA
| | - Jeffrey E. Lee
- Department of Surgical OncologyThe University of Texas M. D. Anderson Cancer CenterHoustonTXUSA
| | - Xin Li
- Department of EpidemiologyRichard M. Fairbanks School of Public HealthIndiana UniversityIndianapolisINUSA
| | - Hongmei Nan
- Department of EpidemiologyRichard M. Fairbanks School of Public HealthIndiana UniversityIndianapolisINUSA
| | - Qingyi Wei
- Duke Cancer InstituteDuke University Medical CenterDurhamNCUSA
- Department of Population Health SciencesDuke University School of MedicineDurhamNCUSA
- Department of MedicineDuke University School of MedicineDurhamNCUSA
| |
Collapse
|
10
|
Kotelevets L, Chastre E. A New Story of the Three Magi: Scaffolding Proteins and lncRNA Suppressors of Cancer. Cancers (Basel) 2021; 13:4264. [PMID: 34503076 PMCID: PMC8428372 DOI: 10.3390/cancers13174264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022] Open
Abstract
Scaffolding molecules exert a critical role in orchestrating cellular response through the spatiotemporal assembly of effector proteins as signalosomes. By increasing the efficiency and selectivity of intracellular signaling, these molecules can exert (anti/pro)oncogenic activities. As an archetype of scaffolding proteins with tumor suppressor property, the present review focuses on MAGI1, 2, and 3 (membrane-associated guanylate kinase inverted), a subgroup of the MAGUK protein family, that mediate networks involving receptors, junctional complexes, signaling molecules, and the cytoskeleton. MAGI1, 2, and 3 are comprised of 6 PDZ domains, 2 WW domains, and 1 GUK domain. These 9 protein binding modules allow selective interactions with a wide range of effectors, including the PTEN tumor suppressor, the β-catenin and YAP1 proto-oncogenes, and the regulation of the PI3K/AKT, the Wnt, and the Hippo signaling pathways. The frequent downmodulation of MAGIs in various human malignancies makes these scaffolding molecules and their ligands putative therapeutic targets. Interestingly, MAGI1 and MAGI2 genetic loci generate a series of long non-coding RNAs that act as a tumor promoter or suppressor in a tissue-dependent manner, by selectively sponging some miRNAs or by regulating epigenetic processes. Here, we discuss the different paths followed by the three MAGIs to control carcinogenesis.
Collapse
Affiliation(s)
- Larissa Kotelevets
- Sorbonne Université, INSERM, UMR_S938, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
| | - Eric Chastre
- Sorbonne Université, INSERM, UMR_S938, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
| |
Collapse
|
11
|
Zhang H, Lin L, Liu J, Pan L, Lin Z, Zhang M, Zhang J, Cao Y, Zhu J, Zhang R. Phase Separation of MAGI2-Mediated Complex Underlies Formation of Slit Diaphragm Complex in Glomerular Filtration Barrier. J Am Soc Nephrol 2021; 32:1946-1960. [PMID: 34330769 PMCID: PMC8455267 DOI: 10.1681/asn.2020111590] [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: 11/12/2020] [Accepted: 03/22/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Slit diaphragm is a specialized adhesion junction between the opposing podocytes, establishing the final filtration barrier to urinary protein loss. At the cytoplasmic insertion site of each slit diaphragm there is an electron-dense and protein-rich cellular compartment that is essential for slit diaphragm integrity and signal transduction. Mutations in genes that encode components of this membrane-less compartment have been associated with glomerular diseases. However, the molecular mechanism governing formation of compartmentalized slit diaphragm assembly remains elusive. METHODS We systematically investigated the interactions between key components at slit diaphragm, such as MAGI2, Dendrin, and CD2AP, through a combination of biochemical, biophysical, and cell biologic approaches. RESULTS We demonstrated that MAGI2, a unique MAGUK family scaffold protein at slit diaphragm, can autonomously undergo liquid-liquid phase separation. Multivalent interactions among the MAGI2-Dendrin-CD2AP complex drive the formation of the highly dense slit diaphragm condensates at physiologic conditions. The reconstituted slit diaphragm condensates can effectively recruit Nephrin. A nephrotic syndrome-associated mutation of MAGI2 interfered with formation of the slit diaphragm condensates, thus leading to impaired enrichment of Nephrin. CONCLUSIONS Key components at slit diaphragm (e.g., MAGI2 and its complex) can spontaneously undergo phase separation. The reconstituted slit diaphragm condensates can be enriched in adhesion molecules and cytoskeletal adaptor proteins. Therefore, the electron-dense slit diaphragm assembly might form via phase separation of core components of the slit diaphragm in podocytes.
Collapse
Affiliation(s)
- Haijiao Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lin Lin
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jianping Liu
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lifeng Pan
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhijie Lin
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China,School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jiong Zhang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ying Cao
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jinwei Zhu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Rongguang Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
12
|
MAGI1, a Scaffold Protein with Tumor Suppressive and Vascular Functions. Cells 2021; 10:cells10061494. [PMID: 34198584 PMCID: PMC8231924 DOI: 10.3390/cells10061494] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
MAGI1 is a cytoplasmic scaffolding protein initially identified as a component of cell-to-cell contacts stabilizing cadherin-mediated cell–cell adhesion in epithelial and endothelial cells. Clinical-pathological and experimental evidence indicates that MAGI1 expression is decreased in some inflammatory diseases, and also in several cancers, including hepatocellular carcinoma, colorectal, cervical, breast, brain, and gastric cancers and appears to act as a tumor suppressor, modulating the activity of oncogenic pathways such as the PI3K/AKT and the Wnt/β-catenin pathways. Genomic mutations and other mechanisms such as mechanical stress or inflammation have been described to regulate MAGI1 expression. Intriguingly, in breast and colorectal cancers, MAGI1 expression is induced by non-steroidal anti-inflammatory drugs (NSAIDs), suggesting a role in mediating the tumor suppressive activity of NSAIDs. More recently, MAGI1 was found to localize at mature focal adhesion and to regulate integrin-mediated adhesion and signaling in endothelial cells. Here, we review MAGI1′s role as scaffolding protein, recent developments in the understanding of MAGI1 function as tumor suppressor gene, its role in endothelial cells and its implication in cancer and vascular biology. We also discuss outstanding questions about its regulation and potential translational implications in oncology.
Collapse
|
13
|
Papakrivopoulou E, Jafree DJ, Dean CH, Long DA. The Biological Significance and Implications of Planar Cell Polarity for Nephrology. Front Physiol 2021; 12:599529. [PMID: 33716764 PMCID: PMC7952641 DOI: 10.3389/fphys.2021.599529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
The orientation of cells in two-dimensional and three-dimensional space underpins how the kidney develops and responds to disease. The process by which cells orientate themselves within the plane of a tissue is termed planar cell polarity. In this Review, we discuss how planar cell polarity and the proteins that underpin it govern kidney organogenesis and pathology. The importance of planar cell polarity and its constituent proteins in multiple facets of kidney development is emphasised, including ureteric bud branching, tubular morphogenesis and nephron maturation. An overview is given of the relevance of planar cell polarity and its proteins for inherited human renal diseases, including congenital malformations with unknown aetiology and polycystic kidney disease. Finally, recent work is described outlining the influence of planar cell polarity proteins on glomerular diseases and highlight how this fundamental pathway could yield a new treatment paradigm for nephrology.
Collapse
Affiliation(s)
- Eugenia Papakrivopoulou
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Department of Internal Medicine and Nephrology, Clinique Saint Jean, Brussels, Belgium
| | - Daniyal J Jafree
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,UCL MB/Ph.D. Programme, Faculty of Medical Science, University College London, London, United Kingdom
| | - Charlotte H Dean
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
14
|
Yamada H, Shirata N, Makino S, Miyake T, Trejo JAO, Yamamoto-Nonaka K, Kikyo M, Empitu MA, Kadariswantiningsih IN, Kimura M, Ichimura K, Yokoi H, Mukoyama M, Hotta A, Nishimori K, Yanagita M, Asanuma K. MAGI-2 orchestrates the localization of backbone proteins in the slit diaphragm of podocytes. Kidney Int 2020; 99:382-395. [PMID: 33144214 DOI: 10.1016/j.kint.2020.09.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 08/22/2020] [Accepted: 09/10/2020] [Indexed: 01/19/2023]
Abstract
Podocytes are highly specialized cells within the glomerulus that are essential for ultrafiltration. The slit diaphragm between the foot processes of podocytes functions as a final filtration barrier to prevent serum protein leakage into urine. The slit-diaphragm consists mainly of Nephrin and Neph1, and localization of these backbone proteins is essential to maintaining the integrity of the glomerular filtration barrier. However, the mechanisms that regulate the localization of these backbone proteins have remained elusive. Here, we focused on the role of membrane-associated guanylate kinase inverted 2 (MAGI-2) in order to investigate mechanisms that orchestrate localization of slit-diaphragm backbone proteins. MAGI-2 downregulation coincided with a reduced expression of slit-diaphragm backbone proteins in human kidneys glomerular disease such as focal segmental glomerulosclerosis or IgA nephropathy. Podocyte-specific deficiency of MAGI-2 in mice abrogated localization of Nephrin and Neph1 independently of other scaffold proteins. Although a deficiency of zonula occuldens-1 downregulated the endogenous Neph1 expression, MAGI-2 recovered Neph1 expression at the cellular edge in cultured podocytes. Additionally, overexpression of MAGI-2 preserved Nephrin localization to intercellular junctions. Co-immunoprecipitation and pull-down assays also revealed the importance of the PDZ domains of MAGI-2 for the interaction between MAGI-2 and slit diaphragm backbone proteins in podocytes. Thus, localization and stabilization of Nephrin and Neph1 in intercellular junctions is regulated mainly via the PDZ domains of MAGI-2 together with other slit-diaphragm scaffold proteins. Hence, these findings may elucidate a mechanism by which the backbone proteins are maintained.
Collapse
Affiliation(s)
- Hiroyuki Yamada
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan; Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naritoshi Shirata
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharmaceutical Corporation, Saitama, Japan
| | - Shinichi Makino
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan; Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takafumi Miyake
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Kanae Yamamoto-Nonaka
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mitsuhiro Kikyo
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharmaceutical Corporation, Saitama, Japan
| | - Maulana A Empitu
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | - Maiko Kimura
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideki Yokoi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Akitsu Hotta
- Department of Reprogramming Science, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Katsuhiko Nishimori
- Department of Obesity and Inflammation Research, Fukushima Medical University, Fukushima, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Katsuhiko Asanuma
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan; Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
15
|
Blaine J, Dylewski J. Regulation of the Actin Cytoskeleton in Podocytes. Cells 2020; 9:cells9071700. [PMID: 32708597 PMCID: PMC7408282 DOI: 10.3390/cells9071700] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Podocytes are an integral part of the glomerular filtration barrier, a structure that prevents filtration of large proteins and macromolecules into the urine. Podocyte function is dependent on actin cytoskeleton regulation within the foot processes, structures that link podocytes to the glomerular basement membrane. Actin cytoskeleton dynamics in podocyte foot processes are complex and regulated by multiple proteins and other factors. There are two key signal integration and structural hubs within foot processes that regulate the actin cytoskeleton: the slit diaphragm and focal adhesions. Both modulate actin filament extension as well as foot process mobility. No matter what the initial cause, the final common pathway of podocyte damage is dysregulation of the actin cytoskeleton leading to foot process retraction and proteinuria. Disruption of the actin cytoskeleton can be due to acquired causes or to genetic mutations in key actin regulatory and signaling proteins. Here, we describe the major structural and signaling components that regulate the actin cytoskeleton in podocytes as well as acquired and genetic causes of actin dysregulation.
Collapse
Affiliation(s)
- Judith Blaine
- Renal Division, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - James Dylewski
- Renal Division, University of Colorado Anschutz Medical Campus and Denver Health Medical Center, Aurora, CO 80045, USA
- Correspondence: ; Tel.: +303-724-4841
| |
Collapse
|
16
|
Valle BL, Rodriguez-Torres S, Kuhn E, Díaz-Montes T, Parrilla-Castellar E, Lawson FP, Folawiyo O, Ili-Gangas C, Brebi-Mieville P, Eshleman JR, Herman J, Shih IM, Sidransky D, Guerrero-Preston R. HIST1H2BB and MAGI2 Methylation and Somatic Mutations as Precision Medicine Biomarkers for Diagnosis and Prognosis of High-grade Serous Ovarian Cancer. Cancer Prev Res (Phila) 2020; 13:783-794. [PMID: 32581010 DOI: 10.1158/1940-6207.capr-19-0412] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 04/15/2020] [Accepted: 06/11/2020] [Indexed: 12/23/2022]
Abstract
Molecular alterations that contribute to long-term (LT) and short-term (ST) survival in ovarian high-grade serous carcinoma (HGSC) may be used as precision medicine biomarkers. DNA promoter methylation is an early event in tumorigenesis, which can be detected in blood and urine, making it a feasible companion biomarker to somatic mutations for early detection and targeted treatment workflows. We compared the methylation profile in 12 HGSC tissue samples to 30 fallopian tube epithelium samples, using the Infinium Human Methylation 450K Array. We also used 450K methylation arrays to compare methylation among HGSCs long-term survivors (more than 5 years) and short-term survivors (less than 3 years). We verified the array results using bisulfite sequencing and methylation-specific PCR (qMSP). in another cohort of HGSC patient samples (n = 35). Immunoblot and clonogenic assays after pharmacologic unmasking show that HIST1H2BB and MAGI2 promoter methylation downregulates mRNA expression levels in ovarian cancer cells. We then used qMSP in paired tissue, ascites, plasma/serum, vaginal swabs, and urine from a third cohort of patients with HGSC cancer (n = 85) to test the clinical potential of HIST1H2BB and MAGI2 in precision medicine workflows. We also performed next-generation exome sequencing of 50 frequently mutated in human cancer genes, using the Ion AmpliSeqCancer Hotspot Panel, to show that the somatic mutation profile found in tissue and plasma can be quantified in paired urine samples from patients with HGSC. Our results suggest that HIST1H2BB and MAGI2 have growth-suppressing roles and can be used as HGSC precision medicine biomarkers.
Collapse
Affiliation(s)
- Blanca L Valle
- Otolaryngology Department, Head and Neck Cancer Research Division, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Sebastian Rodriguez-Torres
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Elisabetta Kuhn
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Italy.,Departments of Pathology, Gynecology and Obstetrics, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Teresa Díaz-Montes
- The Lya Segall Ovarian Cancer Institute, Mercy Medical Center, Baltimore, Maryland
| | | | - Fahcina P Lawson
- Otolaryngology Department, Head and Neck Cancer Research Division, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Oluwasina Folawiyo
- Otolaryngology Department, Head and Neck Cancer Research Division, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Carmen Ili-Gangas
- Laboratory Integrative Biology (LIBi), Center for Excellence in Translational Medicine-Scientific and Technological Bioresources Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - Priscilla Brebi-Mieville
- Laboratory Integrative Biology (LIBi), Center for Excellence in Translational Medicine-Scientific and Technological Bioresources Nucleus (CEMT-BIOREN), Universidad de La Frontera, Temuco, Chile
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - James Herman
- Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania
| | - Ie-Ming Shih
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico; Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Italy
| | - David Sidransky
- Otolaryngology Department, Head and Neck Cancer Research Division, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Rafael Guerrero-Preston
- Otolaryngology Department, Head and Neck Cancer Research Division, The Johns Hopkins University, School of Medicine, Baltimore, Maryland. .,University of Puerto Rico School of Medicine, Department of Obstetrics and Gynecology, San Juan, Puerto Rico.,LifeGene Biomarks Inc., San Juan, Puerto Rico
| |
Collapse
|
17
|
Mei P, Freitag CE, Wei L, Zhang Y, Parwani AV, Li Z. High tumor mutation burden is associated with DNA damage repair gene mutation in breast carcinomas. Diagn Pathol 2020; 15:50. [PMID: 32393302 PMCID: PMC7212599 DOI: 10.1186/s13000-020-00971-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/05/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Immunotherapy has demonstrated encouraging clinical benefits in patients with advanced breast carcinomas and Programmed death ligand 1 (PD-L1) expression has been proposed as an immunotherapy biomarker. Challenges with current PD-L1 testing exist and tumor mutation burden (TMB) is emerging as a biomarker to predict clinical response to immunotherapy in melanoma and non-small cell lung cancer patients. However, TMB has not been well characterized in breast carcinomas. METHODS The study cohort included 62 advanced breast cancer patients (13 primary and 49 metastatic). Genetic alterations and TMB were determined by FoundationOne CDx next generation sequencing (NGS) and the association with clinicopathologic features was analyzed. RESULTS High TMB was observed in a relatively low frequency (3/62, 4.8%). TMB levels were positively associated tumor infiltrating lymphocytes and significantly higher TMB was observed in breast carcinomas with DNA damage repair gene mutation(s). There was no significant association between TMB levels and other analyzed clinicopathologic characteristics. CONCLUSIONS Our data indicate the importance of DNA damage repair proteins in maintaining DNA integrity and immune reaction and breast carcinoma patients with DDR mutation may benefit from immunotherapy.
Collapse
Affiliation(s)
- Ping Mei
- Department of Pathology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - C Eric Freitag
- Department of Pathology, Mayo Clinic, Rochester, MN, USA
| | - Lai Wei
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | | | - Anil V Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, 410 W. 10th Ave, Columbus, 43210, OH, USA
| | - Zaibo Li
- Department of Pathology, The Ohio State University Wexner Medical Center, 410 W. 10th Ave, Columbus, 43210, OH, USA.
| |
Collapse
|
18
|
Cao Z, Ji J, Wang FB, Kong C, Xu H, Xu YL, Chen X, Yu YW, Sun YH. MAGI-2 downregulation: a potential predictor of tumor progression and early recurrence in Han Chinese patients with prostate cancer. Asian J Androl 2020; 22:616-622. [PMID: 32167077 PMCID: PMC7705969 DOI: 10.4103/aja.aja_142_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Membrane-associated guanylate kinase (MAGUK) family protein MAGUK invert 2 (MAGI-2) has been demonstrated to be involved in the tumorigenic mechanism of prostate cancer. The objective of this study was to investigate the expression of MAGI-2 at mRNA and protein levels. The prognostic value of MAGI-2 in Han Chinese patients with prostate cancer was also investigated. The expression data of MAGI-2 were assessed through database retrieval, analysis of sequencing data from our group, and tissue immunohistochemistry using digital scoring system (H-score). The clinical, pathological, and follow-up data were collected. The expression of MAGI-2 in prostate tumor tissues and prostate normal tissues was evaluated and compared. MAGI-2 expression was associated with clinical parameters including tumor stage, lymph node status, Gleason score, PSA level, and biochemical recurrence of prostate cancer. The relative expression of MAGI-2 mRNA was lower in the tumor tissue in The Cancer Genome Atlas (TCGA) database and sequencing data (P < 0.001). There was no difference in MAGI-2 protein expression between tumor and normal tissues in tissue microarray (TMA) results. MAGI-2 expression was associated with pathological tumor stage (P = 0.02), Gleason score (P = 0.05), and preoperation prostate-specific antigen (PSA; P = 0.04). A positive correlation was identified between MAGI-2 and phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expressions through the analysis of TCGA and TMA data (P < 0.0001). Patients with higher MAGI-2 expression had longer biochemical recurrence-free survival in the univariate analysis (P = 0.005), which indicates an optimal prognostic value of MAGI-2 in Han Chinese patients with prostate cancer. In conclusion, MAGI-2 expression gradually decreases with tumor progression, and can be used as a predictor of tumor recurrence in Chinese patients.
Collapse
Affiliation(s)
- Zhi Cao
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Jin Ji
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Fu-Bo Wang
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Chen Kong
- Department of Traditional Chinese Medicine, New Jiangwan City Community Health Service Centre, Shanghai 200433, China
| | - Huan Xu
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Ya-Long Xu
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Xi Chen
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Yong-Wei Yu
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| | - Ying-Hao Sun
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai 200433, China
| |
Collapse
|
19
|
Meyer-Schwesinger C. An unexpected role of steroid on podocytes: from zebrafish to human nephrotic syndrome? Kidney Int 2019; 95:1015-1017. [PMID: 31010473 DOI: 10.1016/j.kint.2019.01.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/19/2019] [Accepted: 01/23/2019] [Indexed: 10/27/2022]
Abstract
In this issue of Kidney International, Jobst-Schwan et al. developed a zebrafish model of MAGI2-deficiency, which recapitulates findings of human nephrotic syndrome due to MAGI2 mutations. The authors use this model system to screen for drugs that might target and alleviate MAGI2-associated nephrotic syndrome pathways. The scientific context of this publication and the significance of its key findings are discussed in this commentary.
Collapse
Affiliation(s)
- Catherine Meyer-Schwesinger
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
20
|
Zhu B, Cao A, Li J, Young J, Wong J, Ashraf S, Bierzynska A, Menon MC, Hou S, Sawyers C, Campbell KN, Saleem MA, He JC, Hildebrandt F, D'Agati VD, Peng W, Kaufman L. Disruption of MAGI2-RapGEF2-Rap1 signaling contributes to podocyte dysfunction in congenital nephrotic syndrome caused by mutations in MAGI2. Kidney Int 2019; 96:642-655. [PMID: 31171376 PMCID: PMC7259463 DOI: 10.1016/j.kint.2019.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 03/03/2019] [Accepted: 03/14/2019] [Indexed: 11/21/2022]
Abstract
The essential role of membrane associated guanylate kinase 2 (MAGI2) in podocytes is indicated by the phenotypes of severe glomerulosclerosis of both MAGI2 knockout mice and in patients with congenital nephrotic syndrome (CNS) caused by mutations in MAGI2. Here, we show that MAGI2 forms a complex with the Rap1 guanine nucleotide exchange factor, RapGEF2, and that this complex is lost when expressing MAGI2 CNS variants. Co-expression of RapGEF2 with wild-type MAGI2, but not MAGI2 CNS variants, enhanced activation of the small GTPase Rap1, a central signaling node in podocytes. In mice, podocyte-specific RapGEF2 deletion resulted in spontaneous glomerulosclerosis, with qualitative glomerular features comparable to MAGI2 knockout mice. Knockdown of RapGEF2 or MAGI2 in human podocytes caused similar reductions in levels of Rap1 activation and Rap1-mediated downstream signaling. Furthermore, human podocytes expressing MAGI2 CNS variants show severe abnormalities of cellular morphology and dramatic loss of actin cytoskeletal organization, features completely rescued by pharmacological activation of Rap1 via a non-MAGI2 dependent upstream pathway. Finally, immunostaining of kidney sections from patients with congenital nephrotic syndrome and MAGI2 mutations showed reduced podocyte Rap1-mediated signaling. Thus, MAGI2-RapGEF2-Rap1 signaling is essential for normal podocyte function. Hence, disruption of this pathway is an important cause of the renal phenotype induced by MAGI2 CNS mutations.
Collapse
Affiliation(s)
- Bingbing Zhu
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Aili Cao
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianhua Li
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - James Young
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jenny Wong
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shazia Ashraf
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Agnieszka Bierzynska
- University of Bristol, Children's Renal Unit and Bristol Renal, Bristol, United Kingdom
| | - Madhav C Menon
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Steven Hou
- National Cancer Institute, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Charles Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kirk N Campbell
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Moin A Saleem
- University of Bristol, Children's Renal Unit and Bristol Renal, Bristol, United Kingdom
| | - John C He
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Friedhelm Hildebrandt
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Vivette D D'Agati
- Renal Pathology Laboratory, Columbia University Medical Center, New York, New York, USA
| | - Wen Peng
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Lewis Kaufman
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
21
|
Jobst-Schwan T, Hoogstraten CA, Kolvenbach CM, Schmidt JM, Kolb A, Eddy K, Schneider R, Ashraf S, Widmeier E, Majmundar AJ, Hildebrandt F. Corticosteroid treatment exacerbates nephrotic syndrome in a zebrafish model of magi2a knockout. Kidney Int 2019; 95:1079-1090. [PMID: 31010479 DOI: 10.1016/j.kint.2018.12.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/21/2018] [Accepted: 12/13/2018] [Indexed: 01/31/2023]
Abstract
Recently, recessive mutations of MAGI2 were identified as a cause of steroid-resistant nephrotic syndrome (SRNS) in humans and mice. To further delineate the pathogenesis of MAGI2 loss of function, we generated stable knockout lines for the two zebrafish orthologues magi2a and magi2b by CRISPR/Cas9. We also developed a novel assay for the direct detection of proteinuria in zebrafish independent of transgenic background. Whereas knockout of magi2b did not yield a nephrotic syndrome phenotype, magi2a-/- larvae developed ascites, periorbital edema, and proteinuria, as indicated by increased excretion of low molecular weight protein. Electron microscopy demonstrated extensive podocyte foot process effacement. As in human SRNS, we observed genotype/phenotype correlation, with edema onset occurring earlier in zebrafish with truncating alleles (5-6 days post fertilization) versus hypomorphic alleles (19-20 days post fertilization). Paradoxically, corticosteroid treatment exacerbated the phenotype, with earlier onset of edema. In contrast, treatment with cyclosporine A or tacrolimus had no significant effect. Although RhoA signaling has been implicated as a downstream mediator of MAGI2 activity, targeting of the RhoA pathway did not modify the nephrotic syndrome phenotype. In the first CRISPR/Cas9 zebrafish knockout model of SRNS, we found that corticosteroids may have a paradoxical effect in the setting of specific genetic mutations.
Collapse
Affiliation(s)
- Tilman Jobst-Schwan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Charlotte A Hoogstraten
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caroline M Kolvenbach
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Johanna Magdalena Schmidt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amy Kolb
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kaitlyn Eddy
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ronen Schneider
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shazia Ashraf
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Eugen Widmeier
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amar J Majmundar
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
22
|
Empitu MA, Kadariswantiningsih IN, Aizawa M, Asanuma K. MAGI-2 and scaffold proteins in glomerulopathy. Am J Physiol Renal Physiol 2018; 315:F1336-F1344. [PMID: 30110567 DOI: 10.1152/ajprenal.00292.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In many cells and tissues, including the glomerular filtration barrier, scaffold proteins are critical in optimizing signal transduction by enhancing structural stability and functionality of their ligands. Recently, mutations in scaffold protein membrane-associated guanylate kinase inverted 2 (MAGI-2) encoding gene were identified among the etiology of steroid-resistant nephrotic syndrome. MAGI-2 interacts with core proteins of multiple pathways, such as transforming growth factor-β signaling, planar cell polarity pathway, and Wnt/β-catenin signaling in podocyte and slit diaphragm. Through the interaction with its ligand, MAGI-2 modulates the regulation of apoptosis, cytoskeletal reorganization, and glomerular development. This review aims to summarize recent findings on the role of MAGI-2 and some other scaffold proteins, such as nephrin and synaptopodin, in the underlying mechanisms of glomerulopathy.
Collapse
Affiliation(s)
- Maulana A Empitu
- Department of Nephrology, Graduate School of Medicine, Chiba University , Chiba , Japan.,Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Airlangga , Surabaya , Indonesia
| | - Ika N Kadariswantiningsih
- Department of Nephrology, Graduate School of Medicine, Chiba University , Chiba , Japan.,Department of Medical Microbiology, Faculty of Medicine, Universitas Airlangga , Surabaya , Indonesia
| | - Masashi Aizawa
- Department of Nephrology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Katsuhiko Asanuma
- Department of Nephrology, Graduate School of Medicine, Chiba University , Chiba , Japan
| |
Collapse
|
23
|
Pemberton TJ, Verdu P, Becker NS, Willer CJ, Hewlett BS, Le Bomin S, Froment A, Rosenberg NA, Heyer E. A genome scan for genes underlying adult body size differences between Central African hunter-gatherers and farmers. Hum Genet 2018; 137:487-509. [PMID: 30008065 DOI: 10.1007/s00439-018-1902-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 07/03/2018] [Indexed: 12/16/2022]
Abstract
The evolutionary and biological bases of the Central African "pygmy" phenotype, a characteristic of rainforest hunter-gatherers defined by reduced body size compared with neighboring farmers, remain largely unknown. Here, we perform a joint investigation in Central African hunter-gatherers and farmers of adult standing height, sitting height, leg length, and body mass index (BMI), considering 358 hunter-gatherers and 169 farmers with genotypes for 153,798 SNPs. In addition to reduced standing heights, hunter-gatherers have shorter sitting heights and leg lengths and higher sitting/standing height ratios than farmers and lower BMI for males. Standing height, sitting height, and leg length are strongly correlated with inferred levels of farmer genetic ancestry, whereas BMI is only weakly correlated, perhaps reflecting greater contributions of non-genetic factors to body weight than to height. Single- and multi-marker association tests identify one region and eight genes associated with hunter-gatherer/farmer status, and 24 genes associated with the height-related traits. Many of these genes have putative functions consistent with roles in determining their associated traits and the pygmy phenotype, and they include three associated with standing height in non-Africans (PRKG1, DSCAM, MAGI2). We find evidence that European height-associated SNPs or variants in linkage disequilibrium with them contribute to standing- and sitting-height determination in Central Africans, but not to the differential status of hunter-gatherers and farmers. These findings provide new insights into the biological basis of the pygmy phenotype, and they highlight the potential of cross-population studies for exploring the genetic basis of phenotypes that vary naturally across populations.
Collapse
Affiliation(s)
- Trevor J Pemberton
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.
| | - Paul Verdu
- CNRS-MNHN-Université Paris Diderot, UMR 7206 Eco-Anthropologie et Ethnobiologie, Paris, France.
| | - Noémie S Becker
- Division of Evolutionary Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Cristen J Willer
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Barry S Hewlett
- Department of Anthropology, Washington State University, Vancouver, WA, USA
| | - Sylvie Le Bomin
- CNRS-MNHN-Université Paris Diderot, UMR 7206 Eco-Anthropologie et Ethnobiologie, Paris, France
| | | | | | - Evelyne Heyer
- CNRS-MNHN-Université Paris Diderot, UMR 7206 Eco-Anthropologie et Ethnobiologie, Paris, France.
| |
Collapse
|
24
|
Ashraf S, Kudo H, Rao J, Kikuchi A, Widmeier E, Lawson JA, Tan W, Hermle T, Warejko JK, Shril S, Airik M, Jobst-Schwan T, Lovric S, Braun DA, Gee HY, Schapiro D, Majmundar AJ, Sadowski CE, Pabst WL, Daga A, van der Ven AT, Schmidt JM, Low BC, Gupta AB, Tripathi BK, Wong J, Campbell K, Metcalfe K, Schanze D, Niihori T, Kaito H, Nozu K, Tsukaguchi H, Tanaka R, Hamahira K, Kobayashi Y, Takizawa T, Funayama R, Nakayama K, Aoki Y, Kumagai N, Iijima K, Fehrenbach H, Kari JA, El Desoky S, Jalalah S, Bogdanovic R, Stajić N, Zappel H, Rakhmetova A, Wassmer SR, Jungraithmayr T, Strehlau J, Kumar AS, Bagga A, Soliman NA, Mane SM, Kaufman L, Lowy DR, Jairajpuri MA, Lifton RP, Pei Y, Zenker M, Kure S, Hildebrandt F. Mutations in six nephrosis genes delineate a pathogenic pathway amenable to treatment. Nat Commun 2018; 9:1960. [PMID: 29773874 PMCID: PMC5958119 DOI: 10.1038/s41467-018-04193-w] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 04/07/2018] [Indexed: 02/06/2023] Open
Abstract
No efficient treatment exists for nephrotic syndrome (NS), a frequent cause of chronic kidney disease. Here we show mutations in six different genes (MAGI2, TNS2, DLC1, CDK20, ITSN1, ITSN2) as causing NS in 17 families with partially treatment-sensitive NS (pTSNS). These proteins interact and we delineate their roles in Rho-like small GTPase (RLSG) activity, and demonstrate deficiency for mutants of pTSNS patients. We find that CDK20 regulates DLC1. Knockdown of MAGI2, DLC1, or CDK20 in cultured podocytes reduces migration rate. Treatment with dexamethasone abolishes RhoA activation by knockdown of DLC1 or CDK20 indicating that steroid treatment in patients with pTSNS and mutations in these genes is mediated by this RLSG module. Furthermore, we discover ITSN1 and ITSN2 as podocytic guanine nucleotide exchange factors for Cdc42. We generate Itsn2-L knockout mice that recapitulate the mild NS phenotype. We, thus, define a functional network of RhoA regulation, thereby revealing potential therapeutic targets.
Collapse
Affiliation(s)
- Shazia Ashraf
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Hiroki Kudo
- Department of Pediatrics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Jia Rao
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Eugen Widmeier
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer A Lawson
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Weizhen Tan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias Hermle
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jillian K Warejko
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shirlee Shril
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Merlin Airik
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tilman Jobst-Schwan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Svjetlana Lovric
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heon Yung Gee
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - David Schapiro
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amar J Majmundar
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carolin E Sadowski
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Werner L Pabst
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ankana Daga
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amelie T van der Ven
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Johanna M Schmidt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Boon Chuan Low
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Anjali Bansal Gupta
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Brajendra K Tripathi
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jenny Wong
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirk Campbell
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kay Metcalfe
- Manchester Centre for Genomic Medicine, St Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Tetsuya Niihori
- Department of Medical Genetics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Hiroshi Kaito
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hiroyasu Tsukaguchi
- 2nd Department of Internal Medicine, Kansai Medical University, 2-3-1 Shin-machi, Hirakata-shi, Osaka, 573-1191, Japan
| | - Ryojiro Tanaka
- Department of Nephrology, Hyogo Prefectural Kobe Children's Hospital, 1-6-7 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Kiyoshi Hamahira
- Department of Pediatrics, Himeji Red Cross Hospital, 1-12-1 Shimoteno, Himeji, Hyogo, 670-8540, Japan
| | - Yasuko Kobayashi
- Department of Pediatrics, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
- Academic Renal Unit, School of Clinical Science, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Takumi Takizawa
- Department of Pediatrics, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Ryo Funayama
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Keiko Nakayama
- Division of Cell Proliferation, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8575, Japan
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Naonori Kumagai
- Department of Pediatrics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Henry Fehrenbach
- Department of Pediatric Nephrology, Children's Hospital, Memmingen, Germany
| | - Jameela A Kari
- Pediatric Nephrology Center of Excellence and Pediatric Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherif El Desoky
- Pediatric Nephrology Center of Excellence and Pediatric Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sawsan Jalalah
- Pathology Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Radovan Bogdanovic
- Institute for Mother and Child Health Care of Serbia "Dr Vukan Čupić", Department of Nephrology, University of Belgrade, Faculty of Medicine, Belgrade, 11000, Serbia
| | - Nataša Stajić
- Institute for Mother and Child Health Care of Serbia "Dr Vukan Čupić", Department of Nephrology, University of Belgrade, Faculty of Medicine, Belgrade, 11000, Serbia
| | - Hildegard Zappel
- Department for Paediatrics II, University of Göttingen, Göttingen, Germany
| | - Assel Rakhmetova
- Department of Nephrology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | | | | | - Juergen Strehlau
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany
| | - Aravind Selvin Kumar
- Department of Pediatric Nephrology and Medical Genetics, Institute of Child Health and Hospital for Children, TN Dr.M.G.R. Medical University, Chennai, India
| | - Arvind Bagga
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Neveen A Soliman
- Department of Pediatrics, Center of Pediatric Nephrology & Transplantation, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Shrikant M Mane
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Lewis Kaufman
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas R Lowy
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, 10065, USA
| | - York Pei
- Division of Nephrology, University Health Network, and University of Toronto, Toronto, ON, Canada
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan.
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Shirata N, Ihara KI, Yamamoto-Nonaka K, Seki T, Makino SI, Oliva Trejo JA, Miyake T, Yamada H, Campbell KN, Nakagawa T, Mori K, Yanagita M, Mundel P, Nishimori K, Asanuma K. Glomerulosclerosis Induced by Deficiency of Membrane-Associated Guanylate Kinase Inverted 2 in Kidney Podocytes. J Am Soc Nephrol 2017; 28:2654-2669. [PMID: 28539383 DOI: 10.1681/asn.2016121356] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/03/2017] [Indexed: 11/03/2022] Open
Abstract
Membrane-associated guanylate kinase inverted 2 (MAGI-2) is a component of the slit diaphragm (SD) of glomerular podocytes. Here, we investigated the podocyte-specific function of MAGI-2 using newly generated podocyte-specific MAGI-2-knockout (MAGI-2-KO) mice. Compared with podocytes from wild-type mice, podocytes from MAGI-2-KO mice exhibited SD disruption, morphologic abnormalities of foot processes, and podocyte apoptosis leading to podocyte loss. These pathologic changes manifested as massive albuminuria by 8 weeks of age and glomerulosclerosis and significantly higher plasma creatinine levels at 12 weeks of age; all MAGI-2-KO mice died by 20 weeks of age. Loss of MAGI-2 in podocytes associated with decreased expression and nuclear translocation of dendrin, which is also a component of the SD complex. Dendrin translocates from the SD to the nucleus of injured podocytes, promoting apoptosis. Our coimmunoprecipitation and in vitro reconstitution studies showed that dendrin is phosphorylated by Fyn and dephosphorylated by PTP1B, and that Fyn-induced phosphorylation prevents Nedd4-2-mediated ubiquitination of dendrin. Under physiologic conditions in vivo, phosphorylated dendrin localized at the SDs; in the absence of MAGI-2, dephosphorylated dendrin accumulated in the nucleus. Furthermore, induction of experimental GN in rats led to the downregulation of MAGI-2 expression and the nuclear accumulation of dendrin in podocytes. In summary, MAGI-2 and Fyn protect dendrin from Nedd4-2-mediated ubiquitination and from nuclear translocation, thereby maintaining the physiologic homeostasis of podocytes, and the lack of MAGI-2 in podocytes results in FSGS.
Collapse
Affiliation(s)
- Naritoshi Shirata
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharmaceutical Corporation, Toda, Japan
| | - Kan-Ichiro Ihara
- The Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kanae Yamamoto-Nonaka
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Takuto Seki
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Shin-Ichi Makino
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Juan Alejandro Oliva Trejo
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takafumi Miyake
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Yamada
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kirk Nicholas Campbell
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York; and
| | - Takahiko Nakagawa
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiyoshi Mori
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Peter Mundel
- Department of Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts
| | - Katsuhiko Nishimori
- The Laboratory of Molecular Biology, Department of Molecular and Cell Biology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Katsuhiko Asanuma
- The Laboratory for Kidney Research (TMK project), Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan; .,Division of Nephrology, Department of Internal Medicine, Faculty of Medicine, Juntendo University, Tokyo, Japan.,Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
26
|
Giannico GA, Arnold SA, Gellert LL, Hameed O. New and Emerging Diagnostic and Prognostic Immunohistochemical Biomarkers in Prostate Pathology. Adv Anat Pathol 2017; 24:35-44. [PMID: 27941540 PMCID: PMC10182893 DOI: 10.1097/pap.0000000000000136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The diagnosis of minimal prostatic adenocarcinoma can be challenging on prostate needle biopsy, and immunohistochemistry may be used to support the diagnosis of cancer. The International Society of Urologic Pathology currently recommends the use of the basal cell markers high-molecular-weight cytokeraratin and p63, and α-methylacyl-coenzyme-A racemase. However, there are caveats associated with the interpretation of these markers, particularly with benign mimickers. Another issue is that of early detection of presence and progression of disease and prediction of recurrence after clinical intervention. There remains a lack of reliable biomarkers to accurately predict low-risk cancer and avoid over treatment. As such, aggressive forms of prostate cancer may be missed and indolent disease may be subjected to unnecessary radical therapy. New biomarker discovery promises to improve early detection and prognosis and to provide targets for therapeutic interventions. In this review, we present the emerging immunohistochemical biomarkers of prostate cancer PTEN, ERG, FASN, MAGI-2, and SPINK1, and address their diagnostic and prognostic advantages and limitations.
Collapse
Affiliation(s)
- Giovanna A. Giannico
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| | - Shanna A. Arnold
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
- Department of Veterans Affairs, Nashville, TN
| | - Lan L. Gellert
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| | - Omar Hameed
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| |
Collapse
|
27
|
Bierzynska A, Soderquest K, Dean P, Colby E, Rollason R, Jones C, Inward CD, McCarthy HJ, Simpson MA, Lord GM, Williams M, Welsh GI, Koziell AB, Saleem MA. MAGI2 Mutations Cause Congenital Nephrotic Syndrome. J Am Soc Nephrol 2016; 28:1614-1621. [PMID: 27932480 DOI: 10.1681/asn.2016040387] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS), a heterogeneous disorder of the renal glomerular filtration barrier, results in impairment of glomerular permselectivity. Inheritance of genetic SRNS may be autosomal dominant or recessive, with a subset of autosomal recessive SRNS presenting as congenital nephrotic syndrome (CNS). Mutations in 53 genes are associated with human SRNS, but these mutations explain ≤30% of patients with hereditary cases and only 20% of patients with sporadic cases. The proteins encoded by these genes are expressed in podocytes, and malfunction of these proteins leads to a universal end point of podocyte injury, glomerular filtration barrier disruption, and SRNS. Here, we identified novel disease-causing mutations in membrane-associated guanylate kinase, WW, and PDZ domain-containing 2 (MAGI2) through whole-exome sequencing of a deeply phenotyped cohort of patients with congenital, childhood-onset SRNS. Although MAGI2 has been shown to interact with nephrin and regulate podocyte cytoskeleton and slit diaphragm dynamics, MAGI2 mutations have not been described in human SRNS. We detected two unique frameshift mutations and one duplication in three patients (two families); two siblings shared the same homozygous frameshift mutation, whereas one individual with sporadic SRNS exhibited compound heterozygosity. Two mutations were predicted to introduce premature stop codons, and one was predicted to result in read through of the normal translational termination codon. Immunohistochemistry in kidney sections from these patients revealed that mutations resulted in lack of or diminished podocyte MAGI2 expression. Our data support the finding that mutations in the MAGI2 gene are causal for congenital SRNS.
Collapse
Affiliation(s)
- Agnieszka Bierzynska
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Katrina Soderquest
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, and
| | - Philip Dean
- Bristol Genetics Laboratory, North Bristol National Health Service Trust, Bristol, United Kingdom; and
| | - Elizabeth Colby
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Ruth Rollason
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Caroline Jones
- Alder Hey Children's Hospital, Liverpool, United Kingdom
| | - Carol D Inward
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Hugh J McCarthy
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Michael A Simpson
- Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Graham M Lord
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, and
| | - Maggie Williams
- Bristol Genetics Laboratory, North Bristol National Health Service Trust, Bristol, United Kingdom; and
| | - Gavin I Welsh
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Ania B Koziell
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, and
| | - Moin A Saleem
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom;
| | | | | |
Collapse
|
28
|
Ni J, Bao S, Johnson RI, Zhu B, Li J, Vadaparampil J, Smith CM, Campbell KN, Grahammer F, Huber TB, He JC, D'Agati VD, Chan A, Kaufman L. MAGI-1 Interacts with Nephrin to Maintain Slit Diaphragm Structure through Enhanced Rap1 Activation in Podocytes. J Biol Chem 2016; 291:24406-24417. [PMID: 27707879 PMCID: PMC5114397 DOI: 10.1074/jbc.m116.745026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/05/2016] [Indexed: 12/15/2022] Open
Abstract
MAGI-1 is a multidomain cytosolic scaffolding protein that in the kidney is specifically located at the podocyte slit diaphragm, a specialized junction that is universally injured in proteinuric diseases. There it interacts with several essential molecules, including nephrin and neph1, which are required for slit diaphragm formation and as an intracellular signaling hub. Here, we show that diminished MAGI-1 expression in cultured podocytes reduced nephrin and neph1 membrane localization and weakened tight junction integrity. Global magi1 knock-out mice, however, demonstrated normal glomerular histology and function into adulthood. We hypothesized that a second mild but complementary genetic insult might induce glomerular disease susceptibility in these mice. To identify such a gene, we utilized the developing fly eye to test for functional complementation between MAGI and its binding partners. In this way, we identified diminished expression of fly Hibris (nephrin) or Roughest (neph1) as dramatically exacerbating the effects of MAGI depletion. Indeed, when these combinations were studied in mice, the addition of nephrin, but not neph1, heterozygosity to homozygous deletion of MAGI-1 resulted in spontaneous glomerulosclerosis. In cultured podocytes, MAGI-1 depletion reduced intercellular contact-induced Rap1 activation, a pathway critical for proper podocyte function. Similarly, magi1 knock-out mice showed diminished glomerular Rap1 activation, an effect dramatically enhanced by concomitant nephrin haploinsufficiency. Finally, combined overexpression of MAGI-1 and nephrin increased Rap1 activation, but not when substituting a mutant MAGI-1 that cannot bind nephrin. We conclude that the interaction between nephrin and MAGI-1 regulates Rap1 activation in podocytes to maintain long term slit diaphragm structure.
Collapse
Affiliation(s)
- Jie Ni
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029,; the Division of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin, China 150001
| | - Sujin Bao
- the Saint James School of Medicine, Saint Vincent and the Grenadines
| | - Ruth I Johnson
- the Biology Department, Wesleyan University, Middletown, Connecticut, 06459
| | - Bingbing Zhu
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029,; the Department of Nephrology, Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China 200062
| | - Jianhua Li
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Justin Vadaparampil
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Christopher M Smith
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Kirk N Campbell
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Florian Grahammer
- the Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Tobias B Huber
- the Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany,; the BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, 79104 Freiburg, Germany,; FRIAS, Freiburg Institute for Advanced Studies and Center for Systems Biology (ZBSA), Albert-Ludwigs-University, 79104 Freiburg, Germany
| | - John C He
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Vivette D D'Agati
- the Department of Pathology, Columbia University Medical Center, New York, New York 10032, and
| | - Andrew Chan
- the School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Lewis Kaufman
- From the Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York 10029,.
| |
Collapse
|
29
|
McNicholas BA, Eng DG, Lichtnekert J, Rabinowitz PS, Pippin JW, Shankland SJ. Reducing mTOR augments parietal epithelial cell density in a model of acute podocyte depletion and in aged kidneys. Am J Physiol Renal Physiol 2016; 311:F626-39. [PMID: 27440779 PMCID: PMC5142165 DOI: 10.1152/ajprenal.00196.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/12/2016] [Indexed: 02/06/2023] Open
Abstract
Parietal epithelial cell (PEC) response to glomerular injury may underlie a common pathway driving fibrogenesis following podocyte loss that typifies several glomerular disorders. Although the mammalian target of rapamycin (mTOR) pathway is important in cell homeostasis, little is known of the biological role or impact of reducing mTOR activity on PEC response following podocyte depletion, nor in the aging kidney. The purpose of these studies was to determine the impact on PECs of reducing mTOR activity following abrupt experimental depletion in podocyte number, as well as in a model of chronic podocyte loss and sclerosis associated with aging. Podocyte depletion was induced by an anti-podocyte antibody and rapamycin started at day 5 until death at day 14 Reducing mTOR did not lead to a greater reduction in podocyte density, despite greater glomerulosclerosis. However, mTOR inhibition lead to an increase in PEC density and PEC-derived crescent formation. Additionally, markers of epithelial-to-mesenchymal transition (platelet-derived growth factor receptor-β, α-smooth muscle actin, Notch-3) and PEC activation (CD44, collagen IV) were further increased by mTOR reduction. Aged mice treated with rapamycin for 1, 2, and 10 wk before death at 26.5 mo (≈75-yr-old human age) had increased the number of glomeruli with a crescentic appearance. mTOR inhibition at either a high or low level lead to changes in PEC phenotype, indicating PEC morphology is sensitive to changes mediated by global mTOR inhibition.
Collapse
Affiliation(s)
| | - Diana G Eng
- Division of Nephrology, University of Washington, Seattle, Washington; and
| | - Julia Lichtnekert
- Division of Nephrology, University of Washington, Seattle, Washington; and
| | | | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington; and
| | - Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington; and
| |
Collapse
|
30
|
Involvement of Tight Junction Plaque Proteins in Cancer. CURRENT PATHOBIOLOGY REPORTS 2016. [DOI: 10.1007/s40139-016-0108-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
31
|
Abstract
Podocytes are highly specialized cells of the kidney glomerulus that wrap around capillaries and that neighbor cells of the Bowman’s capsule. When it comes to glomerular filtration, podocytes play an active role in preventing plasma proteins from entering the urinary ultrafiltrate by providing a barrier comprising filtration slits between foot processes, which in aggregate represent a dynamic network of cellular extensions. Foot processes interdigitate with foot processes from adjacent podocytes and form a network of narrow and rather uniform gaps. The fenestrated endothelial cells retain blood cells but permit passage of small solutes and an overlying basement membrane less permeable to macromolecules, in particular to albumin. The cytoskeletal dynamics and structural plasticity of podocytes as well as the signaling between each of these distinct layers are essential for an efficient glomerular filtration and thus for proper renal function. The genetic or acquired impairment of podocytes may lead to foot process effacement (podocyte fusion or retraction), a morphological hallmark of proteinuric renal diseases. Here, we briefly discuss aspects of a contemporary view of podocytes in glomerular filtration, the patterns of structural changes in podocytes associated with common glomerular diseases, and the current state of basic and clinical research.
Collapse
Affiliation(s)
- Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Mehmet M Altintas
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
32
|
Lefebvre J, Clarkson M, Massa F, Bradford ST, Charlet A, Buske F, Lacas-Gervais S, Schulz H, Gimpel C, Hata Y, Schaefer F, Schedl A. Alternatively spliced isoforms of WT1 control podocyte-specific gene expression. Kidney Int 2015; 88:321-31. [DOI: 10.1038/ki.2015.140] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 03/26/2015] [Accepted: 03/26/2015] [Indexed: 01/26/2023]
|
33
|
Weins A, Wong JS, Basgen JM, Gupta R, Daehn I, Casagrande L, Lessman D, Schwartzman M, Meliambro K, Patrakka J, Shaw A, Tryggvason K, He JC, Nicholas SB, Mundel P, Campbell KN. Dendrin ablation prolongs life span by delaying kidney failure. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2143-57. [PMID: 26073036 DOI: 10.1016/j.ajpath.2015.04.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 04/01/2015] [Accepted: 04/21/2015] [Indexed: 12/16/2022]
Abstract
Podocyte loss is central to the progression of proteinuric kidney diseases leading to end-stage kidney disease (ESKD), requiring renal replacement therapy, such as dialysis. Despite modern tools and techniques, the 5-year mortality of some patients requiring dialysis remains at about 70% to 80%. Thus, there is a great unmet need for podocyte-specific treatments aimed at preventing podocyte loss and the ensuing development of ESKD. Here, we show that ablation of the podocyte death-promoting protein dendrin delays the onset of ESKD, thereby expanding the life span of mice lacking the adapter protein CD2AP. Ablation of dendrin delays onset and severity of proteinuria and podocyte loss. In addition, dendrin ablation ameliorates mesangial volume expansion and up-regulation of mesangial fibronectin expression, which is mediated by a podocyte-secreted factor. In conclusion, onset of ESKD and death can be markedly delayed by blocking the function of dendrin.
Collapse
Affiliation(s)
- Astrid Weins
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jenny S Wong
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John M Basgen
- Department of Research, Morphometry and Stereology Laboratory, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Ritu Gupta
- Department of Pathology, Albert Einstein College of Medicine, Medicine, Bronx, New York
| | - Ilse Daehn
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lisette Casagrande
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - David Lessman
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Monica Schwartzman
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kristin Meliambro
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jaakko Patrakka
- KI/AZ Integrated CardioMetabolic Center (ICMC), Department of Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Andrey Shaw
- Division of Immunobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Karl Tryggvason
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - John Cijiang He
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Susanne B Nicholas
- Division of Nephrology, University of California Los Angeles, Los Angeles, California
| | - Peter Mundel
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kirk N Campbell
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
34
|
|