1
|
Bhattacharya R, Ward T, Kalejaiye TD, Mishra A, Leeman S, Arzaghi H, Seidman JG, Seidman CE, Musah S. Engineered human iPS cell models reveal altered podocytogenesis and glomerular capillary wall in CHD-associated SMAD2 mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606108. [PMID: 39211233 PMCID: PMC11360959 DOI: 10.1101/2024.08.02.606108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Early developmental programming involves extensive cell lineage diversification through shared molecular signaling networks. Clinical observations of congenital heart disease (CHD) patients carrying SMAD2 genetic variants revealed correlations with multi-organ impairments at the developmental and functional levels. For example, many CHD patients present with glomerulosclerosis, periglomerular fibrosis, and albuminuria. Still, it remains largely unknown whether SMAD2 variants associated with CHD can directly alter kidney cell fate, tissue patterning, and organ-level function. To address this question, we engineered human iPS cells (iPSCs) and organ-on-a-chip systems to uncover the role of pathogenic SMAD2 variants in kidney podocytogenesis. Our results show that abrogation of SMAD2 causes altered patterning of the mesoderm and intermediate mesoderm (IM) cell lineages, which give rise to nearly all kidney cell types. Upon further differentiation of IM cells, the mutant podocytes failed to develop arborizations and interdigitations. A reconstituted glomerulus-on-a-chip platform exhibited significant proteinuria as clinically observed in glomerulopathies. This study implicates CHD-associated SMAD2 mutations in kidney tissue malformation and provides opportunities for therapeutic discovery in the future.
Collapse
|
2
|
Li X, Xiong Q, Yang Q, Shi J, Han Y, Dong Y, Qian J, Qian Z, Wang H, Wang T, Wu F. PTPRO inhibits LPS-induced apoptosis in alveolar epithelial cells. Biochem Biophys Res Commun 2024; 718:150083. [PMID: 38735138 DOI: 10.1016/j.bbrc.2024.150083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Acute lung injury (ALI) and its severe manifestation, acute respiratory distress syndrome (ARDS), represent critical clinical syndromes with multifactorial origins, notably stemming from sepsis within intensive care units (ICUs). Despite their high mortality rates, no selective cure is available beside ventilation support. Apoptosis plays a complex and pivotal role in the pathophysiology of acute lung injury. Excessive apoptosis of alveolar epithelial and microvascular endothelial cells can lead to disruption of lung epithelial barrier integrity, impairing the body's ability to exchange blood and gas. At the same time, apoptosis of damaged or dysfunctional cells, including endothelial and epithelial cells, can help maintain tissue integrity and accelerate recovery from organ pro-inflammatory stress. The balance between pro-survival and pro-apoptotic signals in lung injury determines patient outcomes, making the modulation of apoptosis an area of intense research in the quest for more effective therapies. Here we found that protein tyrosine phosphatase receptor type O (PTPRO), a poorly understood receptor-like protein tyrosine phosphatase, is consistently upregulated in multiple tissue types of mice under septic conditions and in the lung alveolar epithelial cells. PTPRO reduction by its selective short-interfering RNA (siRNA) leads to excessive apoptosis in lung alveolar epithelial cells without affecting cell proliferation. Consistently PTPRO overexpression by a DNA construct attenuates apoptotic signaling induced by LPS. These effects of PTPTO on cellular apoptosis are dependent on an ErbB2/PI3K/Akt/NFκB signaling pathway. Here we revealed a novel regulatory pathway of cellular apoptosis by PTPRO in lung alveolar epithelial cells during sepsis.
Collapse
Affiliation(s)
- Xuemeng Li
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Qianqian Xiong
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Qingqing Yang
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Jing Shi
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Yuhong Han
- Department of Clinical Laboratory, The Second People's Hospital of Fuyang City, Fuyang, Anhui Province, China
| | - Yishu Dong
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Jun Qian
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zhongqing Qian
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Hongtao Wang
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China
| | - Ting Wang
- Department of Internal Medicine, University of Arizona, Phoenix, AZ, USA
| | - Fengjiao Wu
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, and Department of Laboratory Medicine, Bengbu Medical University, Bengbu, Anhui Province, China.
| |
Collapse
|
3
|
Hu Z, Cano I, Lei F, Liu J, Ramos RB, Gordon H, Paschalis EI, Saint-Geniez M, Ng YSE, D'Amore PA. Deletion of the endothelial glycocalyx component endomucin leads to impaired glomerular structure and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603749. [PMID: 39071302 PMCID: PMC11275787 DOI: 10.1101/2024.07.16.603749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Endomucin (EMCN), an endothelial-specific glycocalyx component, was found to be highly expressed by the endothelium of the renal glomerulus. We reported an anti-inflammatory role of EMCN and its involvement in the regulation of vascular endothelial growth factor (VEGF) activity through modulating VEGF receptor 2 (VEGFR2) endocytosis. The goal of this study is to investigate the phenotypic and functional effects of EMCN deficiency using the first global EMCN knockout mouse model. Methods Global EMCN knockout mice were generated by crossing EMCN-floxed mice with ROSA26-Cre mice. Flow cytometry was employed to analyze infiltrating myeloid cells in the kidneys. The ultrastructure of the glomerular filtration barrier was examined by transmission electron microscopy, while urinary albumin, creatinine, and total protein levels were analyzed from freshly collected urine samples. Expression and localization of EMCN, EGFP, CD45, CD31, CD34, podocin, albumin, and α-smooth muscle actin were examined by immunohistochemistry. Mice were weighed regularly, and their systemic blood pressure was measured using a non-invasive tail-cuff system. Glomerular endothelial cells and podocytes were isolated by fluorescence-activated cell sorting for RNA-seq. Transcriptional profiles were analyzed to identify differentially expressed genes in both endothelium and podocytes, followed by gene ontology analysis of up- and down-regulated genes. Protein levels of EMCN, albumin, and podocin were quantified by Western blot. Results EMCN -/- mice were viable with no gross anatomical defects in kidneys. The EMCN -/- mice exhibited increased infiltration of CD45 + cells, with an increased proportion of Ly6G high Ly6C high myeloid cells and higher VCAM-1 expression. EMCN -/- mice displayed albuminuria with increased albumin in the Bowman's space compared to the EMCN +/+ littermates. Glomeruli in EMCN -/- mice revealed fused and effaced podocyte foot processes and disorganized endothelial fenestrations. We found no significant difference in blood pressure between EMCN knockout mice and their wild-type littermates. RNA-seq of glomerular endothelial cells revealed downregulation of cell-cell adhesion and MAPK/ERK pathways, along with glycocalyx and extracellular matrix remodeling. In podocytes, we observed reduced VEGF signaling and alterations in cytoskeletal organization. Notably, there was a significant decrease in both mRNA and protein levels of podocin, a key component of the slit diaphragm. Conclusion Our study demonstrates a critical role of the endothelial marker EMCN in supporting normal glomerular filtration barrier structure and function by maintaining glomerular endothelial tight junction and homeostasis and podocyte function through endothelial-podocyte crosstalk.
Collapse
|
4
|
Neupane S, Aryal YP, Kwak HJ, Lee SG, Kim TY, Pokharel E, Kim JY, Kim JH, Sohn WJ, An SY, An CH, Jung JK, Ha JH, Yamamoto H, Cho SW, Lee S, Lee Y, Park KK, Min BK, Park C, Kwon TY, Cho SJ, Kim JY. Developmental roles of glomerular epithelial protein-1 in mice molar morphogenesis. Cell Tissue Res 2024; 395:53-62. [PMID: 37985496 DOI: 10.1007/s00441-023-03841-y] [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: 06/28/2022] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
Glomerular epithelial protein-1 (Glepp1), a R3 subtype family of receptor-type protein tyrosine phosphatases, plays important role in the activation of Src family kinases and regulates cellular processes such as cell proliferation, differentiation, and apoptosis. In this study, we firstly examined the functional evaluation of Glepp1 in tooth development and morphogenesis. The precise expression level and developmental function of Glepp1 were examined by RT-qPCR, in situ hybridization, and loss and gain of functional study using a range of in vitro organ cultivation methods. Expression of Glepp1 was detected in the developing tooth germs in cap and bell stage of tooth development. Knocking down Glepp1 at E13 for 2 days showed the altered expression levels of tooth development-related signaling molecules, including Bmps, Dspp, Fgf4, Lef1, and Shh. Moreover, transient knock down of Glepp1 revealed alterations in cellular physiology, examined by the localization patterns of Ki67 and E-cadherin. Similarly, knocking down of Glepp1 showed disrupted enamel rod and interrod formation in 3-week renal transplanted teeth. In addition, due to attrition of odontoblastic layers, the expression signals of Dspp and the localization of NESTIN were almost not detected after knock down of Glepp1; however, their expressions were increased after Glepp1 overexpression. Thus, our results suggested that Glepp1 plays modulating roles during odontogenesis by regulating the expression levels of signaling molecules and cellular events to achieve the proper structural formation of hard tissue matrices in mice molar development.
Collapse
Affiliation(s)
- Sanjiv Neupane
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, USA
| | - Yam Prasad Aryal
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Hee-Jin Kwak
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Korea
| | - Sung-Gwon Lee
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Tae-Young Kim
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Elina Pokharel
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Ji-Youn Kim
- Department of Dental Hygiene, Gachon University, Incheon, Korea
| | - Jung-Hyeuk Kim
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Korea
| | - Wern-Joo Sohn
- Pre-Major of Cosmetics and Pharmaceutics, Daegu Haany University, Gyeongsan, Korea
| | - Seo-Young An
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Chang-Hyeon An
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jae-Kwang Jung
- Department of Oral Medicine, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jung-Hong Ha
- Department of Conservative Dentistry, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Hitoshi Yamamoto
- Department of Histology and Developmental Biology, Tokyo Dental College, Tokyo, Japan
| | - Sung-Won Cho
- Division of Anatomy and Developmental Biology, Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - Sanggyu Lee
- School of Life Science, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Youngkyun Lee
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Kwang-Kyun Park
- Professor Emeritus Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - Bong-Ki Min
- Center for Research Facilities, Yeungnam University, Gyeongsan, Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Tae-Yub Kwon
- Department of Dental Biomaterials, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Sung-Jin Cho
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Korea.
| | - Jae-Young Kim
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, Korea.
| |
Collapse
|
5
|
Cybulsky AV, Papillon J, Bryan C, Navarro‐Betancourt JR, Sabourin LA. Role of the Ste20-like kinase SLK in podocyte adhesion. Physiol Rep 2024; 12:e15897. [PMID: 38163671 PMCID: PMC10758337 DOI: 10.14814/phy2.15897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024] Open
Abstract
SLK controls the cytoskeleton, cell adhesion, and migration. Podocyte-specific deletion of SLK in mice leads to podocyte injury as mice age and exacerbates injury in experimental focal segment glomerulosclerosis (FSGS; adriamycin nephrosis). We hypothesized that adhesion proteins may be substrates of SLK. In adriamycin nephrosis, podocyte ultrastructural injury was exaggerated by SLK deletion. Analysis of a protein kinase phosphorylation site dataset showed that podocyte adhesion proteins-paxillin, vinculin, and talin-1 may be potential SLK substrates. In cultured podocytes, deletion of SLK increased adhesion to collagen. Analysis of paxillin, vinculin, and talin-1 showed that SLK deletion reduced focal adhesion complexes (FACs) containing these proteins mainly in adriamycin-induced injury; there was no change in FAC turnover (focal adhesion kinase Y397 phosphorylation). In podocytes, paxillin S250 showed basal phosphorylation that was slightly enhanced by SLK; however, SLK did not phosphorylate talin-1. In adriamycin nephrosis, SLK deletion did not alter glomerular expression/localization of talin-1 and vinculin, but increased focal adhesion kinase phosphorylation modestly. Therefore, SLK decreases podocyte adhesion, but FAC proteins in podocytes are not major substrates of SLK in health and disease.
Collapse
Affiliation(s)
- Andrey V. Cybulsky
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - Joan Papillon
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - Craig Bryan
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - José R. Navarro‐Betancourt
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - Luc A. Sabourin
- Ottawa Hospital Research Institute, Cancer TherapeuticsOttawaOntarioCanada
| |
Collapse
|
6
|
Saad M, Mehawej C, Faour WH. Analysis of G-quadruplex forming sequences in podocytes-marker genes and their potential roles in inherited glomerular diseases. Heliyon 2023; 9:e20233. [PMID: 37809648 PMCID: PMC10559976 DOI: 10.1016/j.heliyon.2023.e20233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 08/04/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
Nephrotic Syndrome is the most widespread pediatric kidney disorder. Genetic alterations in podocyte genes are thought to be responsible for the disease. G-quadruplexes are non-conventional guanine-rich DNA and RNA structures, which are commonly found in regulatory regions. This study examined the potential G-quadruplexes forming sequences in the promoters and gene bodies of podocyte-marker genes. High G-quadruplexes density was found in the vascular endothelial growth facto, cluster of differentiation-151, integrin subunit beta-4, metalloendopeptidase, Wilms tumor-1, integrin subunit beta-3, synaptopodin, and nephrin promoters. Vascular endothelial growth facto, cluster of differentiation-151 and integrin subunit beta-4 had the highest G-quadruplexes density in their gene bodies and promoters. Additionally, highly stable G-quadruplexes forming sequences were identified within all podocyte-marker genes. Furthermore, it is hypothesized that Wilms tumor-1 is capable of controlling the transcription of podocalyxin by binding to two possible G-quadruplexes forming motifs. We next analyzed the most frequently reported genetic mutations in the selected genes for their effect on DNA G-quadruplexes formation, and the thermodynamic stability of predicted RNA G-quadruplexes, using RNAfold. Importantly, the missense mutation c.121_122del in the nephrin gene reported in patients with NS type 1 affected DNA G-quadruplexes formation in this region as well as the thermodynamic stability of the corresponding RNA G-quadruplexes. Overall, we report the potential regulatory roles of G-quadruplexes in the etiology of nephrotic syndrome and their possible use as drug targets to treat kidney diseases.
Collapse
Affiliation(s)
- Mona Saad
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Cybel Mehawej
- Department of Human Genetics, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Wissam H. Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
7
|
Grubczak K, Starosz A, Makowska B, Parfienowicz Z, Krętowska M, Naumnik B, Moniuszko M. The influence of calcitriol and methylprednisolone on podocytes function in minimal change disease in vitro model. Sci Rep 2023; 13:12731. [PMID: 37543700 PMCID: PMC10404287 DOI: 10.1038/s41598-023-39893-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023] Open
Abstract
Minimal change disease (MCD), considered one of the major causes of nephrotic syndrome, is a complex pathological condition with disturbances in podocytes' foot processes. Numerous studies suggested the essential role of vitamin D3 in maintaining proper glomerulus function. However, the data on direct potential of that compound in reference to podocytes are scarce. Thus, here we assessed the influence of calcitriol (active vitamin D3) on podocyte function, apart from commonly used steroids (methylprednisolone). CIHP-1 podocyte cell line was used to implement the LPS-PAN-induced MCD in vitro model. Viability, podocyte-related slit diaphragm proteins, morphology, function as a barrier was evaluated using flow cytometry, RT-PCR, confocal microscopy, and TEER analysis. Calcitriol or methylprednisolone did not affect cell viability. Podocyte-related proteins demonstrated different responses to in vitro treatment compared to previously reported changes in total glomeruli. Podocyte morphology was partially restored in the presence of the tested compounds. In addition, TEER analysis revealed improvement of LPS-PAN-induced cells' function as a barrier when vitamin D3 or steroid was used. In conclusion, a significant potential for modulation of MCD in vitro model podocytes with calcitriol or selected steroids was reported. Further studies on vitamin D3 in context of podocyte-related phenomenon accompanying MCD are of great importance.
Collapse
Affiliation(s)
- Kamil Grubczak
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, Jerzego Waszyngtona 13, 15-269, Białystok, Poland.
| | - Aleksandra Starosz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, Jerzego Waszyngtona 13, 15-269, Białystok, Poland
| | - Barbara Makowska
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, Jerzego Waszyngtona 13, 15-269, Białystok, Poland
| | - Zuzanna Parfienowicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, Jerzego Waszyngtona 13, 15-269, Białystok, Poland
| | - Magdalena Krętowska
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, Jerzego Waszyngtona 13, 15-269, Białystok, Poland
| | - Beata Naumnik
- Ist Department of Nephrology and Transplantation with Dialysis Unit, Medical University of Bialystok, Żurawia 14, 15-540, Białystok, Poland.
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, Jerzego Waszyngtona 13, 15-269, Białystok, Poland
- Department of Allergology and Internal Medicine, Medical University of Białystok, Marii Skłodowskiej-Curie 24A, 15-276, Białystok, Poland
| |
Collapse
|
8
|
Mamilla M, Yartha SGR, Tuli R, Konipineni S, Rayaprolu DT, Borgharkar G, Kalluru PKR, Thugu TR. Role of Magnesium in Diabetic Nephropathy for Better Outcomes. Cureus 2023; 15:e43076. [PMID: 37692668 PMCID: PMC10484355 DOI: 10.7759/cureus.43076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease worldwide, resulting from uncontrolled diabetes. Oxidative stress plays a critical role in the pathophysiology of DN, leading to cellular damage and disease progression. Magnesium, an essential mineral, has emerged as a potential therapeutic agent due to its antioxidative, anti-inflammatory, and antifibrotic properties. An extensive literature search was conducted on Medline using the keywords "Diabetic nephropathy," "Magnesium," and "Chronic Kidney Disease," and the results published after 2000 were exclusively studied to build this review. This review aims to summarize the role of magnesium in DN and explore its therapeutic potential. Magnesium acts as a cofactor for antioxidant enzymes, directly scavenges reactive oxygen species, and enhances the expression of antioxidant proteins. Furthermore, magnesium exhibits anti-inflammatory effects by suppressing pro-inflammatory cytokine production and inhibiting inflammatory signaling pathways. Magnesium supplementation has been shown to reduce oxidative stress markers and improve antioxidant enzyme activities in clinical studies. Additionally, magnesium has been found to mitigate renal fibrosis, maintain tubular integrity and function, improve endothelial function, and modulate renal hemodynamics. Although limited clinical trials suggest the renoprotective effects of magnesium in DN, further research is needed to determine the optimal dosage, duration, and long-term effects of magnesium supplementation. Despite existing drawbacks and gaps in the literature, magnesium holds promise as adjunctive therapy for DN by targeting oxidative stress and preserving renal function.
Collapse
Affiliation(s)
- Mahesh Mamilla
- Internal Medicine, Sri Venkateswara Medical College, Tirupati, IND
| | | | - Richa Tuli
- Internal Medicine, School of Medicine, Xiamen University, Xiamen, CHN
| | - Sunil Konipineni
- Internal Medicine, Zaporizhzhia State Medical University, Zaporizhzhia, UKR
| | | | - Gargi Borgharkar
- Public Health, University of Alabama at Birmingham, Birmingham, USA
| | | | - Thanmai Reddy Thugu
- Internal Medicine, Sri Padmavathi Medical College for Women, Sri Venkateswara Institute of Medical Sciences, Tirupati, IND
| |
Collapse
|
9
|
Agarwal S, Sudhini YR, Polat OK, Reiser J, Altintas MM. Renal cell markers: lighthouses for managing renal diseases. Am J Physiol Renal Physiol 2021; 321:F715-F739. [PMID: 34632812 DOI: 10.1152/ajprenal.00182.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kidneys, one of the vital organs in our body, are responsible for maintaining whole body homeostasis. The complexity of renal function (e.g., filtration, reabsorption, fluid and electrolyte regulation, and urine production) demands diversity not only at the level of cell types but also in their overall distribution and structural framework within the kidney. To gain an in depth molecular-level understanding of the renal system, it is imperative to discern the components of kidney and the types of cells residing in each of the subregions. Recent developments in labeling, tracing, and imaging techniques have enabled us to mark, monitor, and identify these cells in vivo with high efficiency in a minimally invasive manner. In this review, we summarize different cell types, specific markers that are uniquely associated with those cell types, and their distribution in the kidney, which altogether make kidneys so special and different. Cellular sorting based on the presence of certain proteins on the cell surface allowed for the assignment of multiple markers for each cell type. However, different studies using different techniques have found contradictions in cell type-specific markers. Thus, the term "cell marker" might be imprecise and suboptimal, leading to uncertainty when interpreting the data. Therefore, we strongly believe that there is an unmet need to define the best cell markers for a cell type. Although the compendium of renal-selective marker proteins presented in this review is a resource that may be useful to researchers, we acknowledge that the list may not be necessarily exhaustive.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | | - Onur K Polat
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | |
Collapse
|
10
|
Xie F, Dong H, Zhang H. Regulatory Functions of Protein Tyrosine Phosphatase Receptor Type O in Immune Cells. Front Immunol 2021; 12:783370. [PMID: 34880876 PMCID: PMC8645932 DOI: 10.3389/fimmu.2021.783370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/04/2021] [Indexed: 01/01/2023] Open
Abstract
The members of the protein tyrosine phosphatase (PTP) family are key regulators in multiple signal transduction pathways and therefore they play important roles in many cellular processes, including immune response. As a member of PTP family, protein tyrosine phosphatase receptor type O (PTPRO) belongs to the R3 receptor-like protein tyrosine phosphatases. The expression of PTPRO isoforms is tissue-specific and the truncated PTPRO (PTPROt) is mainly observed in hematopoietic cells, including B cells, T cells, macrophages and other immune cells. Therefore, PTPROt may play an important role in immune cells by affecting their growth, differentiation, activation and immune responses. In this review, we will focus on the regulatory roles and underlying molecular mechanisms of PTPRO/PTPROt in immune cells, including B cells, T cells, and macrophages.
Collapse
Affiliation(s)
- Feiling Xie
- Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
| | - Hongmei Dong
- Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
| | - Hao Zhang
- Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Minister of Education Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| |
Collapse
|
11
|
Buscher K, Heitplatz B, van Marck V, Song J, Loismann S, Rixen R, Hüchtmann B, Kurian S, Ehinger E, Wolf D, Ley K, Pavenstädt H, Reuter S. Data-Driven Kidney Transplant Phenotyping as a Histology-Independent Framework for Biomarker Discovery. J Am Soc Nephrol 2021; 32:1933-1945. [PMID: 34078665 PMCID: PMC8455252 DOI: 10.1681/asn.2020121685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/15/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND In transplant medicine, clinical decision making largely relies on histology of biopsy specimens. However, histology suffers from low specificity, sensitivity, and reproducibility, leading to suboptimal stratification of patients. We developed a histology-independent immune framework of kidney graft homeostasis and rejection. METHODS We applied tailored RNA deconvolution for leukocyte enumeration and coregulated gene network analysis to published bulk human kidney transplant RNA transcriptomes as input for unsupervised, high-dimensional phenotype clustering. We used framework-based graft survival analysis to identify a biomarker that was subsequently characterized in independent transplant biopsy specimens. RESULTS We found seven immune phenotypes that confirm known rejection types and uncovered novel signatures. The molecular phenotypes allow for improved graft survival analysis compared with histology, and identify a high-risk group in nonrejecting transplants. Two fibrosis-related phenotypes with distinct immune features emerged with reduced graft survival. We identified lysyl oxidase-like 2 (LOXL2)-expressing peritubular CD68+ macrophages as a framework-derived biomarker of impaired allograft function. These cells precede graft fibrosis, as demonstrated in longitudinal biopsy specimens, and may be clinically useful as a biomarker for early fibrogenesis. CONCLUSIONS This study provides a comprehensive, data-driven atlas of human kidney transplant phenotypes and demonstrates its utility to identify novel clinical biomarkers.
Collapse
Affiliation(s)
- Konrad Buscher
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, Muenster, Germany,Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California
| | - Barbara Heitplatz
- Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Veerle van Marck
- Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Jian Song
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Sophie Loismann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Rebecca Rixen
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, Muenster, Germany
| | - Birte Hüchtmann
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, Muenster, Germany
| | - Sunil Kurian
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California
| | - Erik Ehinger
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, California
| | - Dennis Wolf
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California,Department of Cardiology and Angiology I, University Heart Center, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, California
| | - Hermann Pavenstädt
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, Muenster, Germany
| | - Stefan Reuter
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Muenster, Muenster, Germany
| |
Collapse
|
12
|
Clotet-Freixas S, McEvoy CM, Batruch I, Pastrello C, Kotlyar M, Van JAD, Arambewela M, Boshart A, Farkona S, Niu Y, Li Y, Famure O, Bozovic A, Kulasingam V, Chen P, Kim SJ, Chan E, Moshkelgosha S, Rahman SA, Das J, Martinu T, Juvet S, Jurisica I, Chruscinski A, John R, Konvalinka A. Extracellular Matrix Injury of Kidney Allografts in Antibody-Mediated Rejection: A Proteomics Study. J Am Soc Nephrol 2020; 31:2705-2724. [PMID: 32900843 DOI: 10.1681/asn.2020030286] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Antibody-mediated rejection (AMR) accounts for >50% of kidney allograft loss. Donor-specific antibodies (DSA) against HLA and non-HLA antigens in the glomeruli and the tubulointerstitium cause AMR while inflammatory cytokines such as TNFα trigger graft injury. The mechanisms governing cell-specific injury in AMR remain unclear. METHODS Unbiased proteomic analysis of laser-captured and microdissected glomeruli and tubulointerstitium was performed on 30 for-cause kidney biopsy specimens with early AMR, acute cellular rejection (ACR), or acute tubular necrosis (ATN). RESULTS A total of 107 of 2026 glomerular and 112 of 2399 tubulointerstitial proteins was significantly differentially expressed in AMR versus ACR; 112 of 2026 glomerular and 181 of 2399 tubulointerstitial proteins were significantly dysregulated in AMR versus ATN (P<0.05). Basement membrane and extracellular matrix (ECM) proteins were significantly decreased in both AMR compartments. Glomerular and tubulointerstitial laminin subunit γ-1 (LAMC1) expression decreased in AMR, as did glomerular nephrin (NPHS1) and receptor-type tyrosine-phosphatase O (PTPRO). The proteomic analysis revealed upregulated galectin-1, which is an immunomodulatory protein linked to the ECM, in AMR glomeruli. Anti-HLA class I antibodies significantly increased cathepsin-V (CTSV) expression and galectin-1 expression and secretion in human glomerular endothelial cells. CTSV had been predicted to cleave ECM proteins in the AMR glomeruli. Glutathione S-transferase ω-1, an ECM-modifying enzyme, was significantly increased in the AMR tubulointerstitium and in TNFα-treated proximal tubular epithelial cells. CONCLUSIONS Basement membranes are often remodeled in chronic AMR. Proteomic analysis performed on laser-captured and microdissected glomeruli and tubulointerstitium identified early ECM remodeling, which may represent a new therapeutic opportunity.
Collapse
Affiliation(s)
- Sergi Clotet-Freixas
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Caitriona M McEvoy
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Ihor Batruch
- Department of Laboratory Medicine and Pathobiology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Chiara Pastrello
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Max Kotlyar
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Julie Anh Dung Van
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Madhurangi Arambewela
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alex Boshart
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Sofia Farkona
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yun Niu
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yanhong Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Olusegun Famure
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Andrea Bozovic
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Vathany Kulasingam
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Peixuen Chen
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - S Joseph Kim
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Emilie Chan
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Sajad Moshkelgosha
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Respirology, Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Syed Ashiqur Rahman
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Center for Systems Immunology, Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jishnu Das
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Center for Systems Immunology, Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tereza Martinu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Respirology, Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Stephen Juvet
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Respirology, Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Andrzej Chruscinski
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Rohan John
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ana Konvalinka
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada .,Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
13
|
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: 62] [Impact Index Per Article: 15.5] [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
|
14
|
Woychyshyn B, Papillon J, Guillemette J, Navarro-Betancourt JR, Cybulsky AV. Genetic ablation of SLK exacerbates glomerular injury in adriamycin nephrosis in mice. Am J Physiol Renal Physiol 2020; 318:F1377-F1390. [PMID: 32308020 DOI: 10.1152/ajprenal.00028.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ste20-like kinase SLK is critical for embryonic development and may play an important role in wound healing, muscle homeostasis, cell migration, and tumor growth. Mice with podocyte-specific deletion of SLK show albuminuria and damage to podocytes as they age. The present study addressed the role of SLK in glomerular injury. We induced adriamycin nephrosis in 3- to 4-mo-old control and podocyte SLK knockout (KO) mice. Compared with control, SLK deletion exacerbated albuminuria and loss of podocytes, synaptopodin, and podocalyxin. Glomeruli of adriamycin-treated SLK KO mice showed diffuse increases in the matrix and sclerosis as well as collapse of the actin cytoskeleton. SLK can phosphorylate ezrin. The complex of phospho-ezrin, Na+/H+ exchanger regulatory factor 2, and podocalyxin in the apical domain of the podocyte is a key determinant of normal podocyte architecture. Deletion of SLK reduced glomerular ezrin and ezrin phosphorylation in adriamycin nephrosis. Also, deletion of SLK reduced the colocalization of ezrin and podocalyxin in the glomerulus. Cultured glomerular epithelial cells with KO of SLK showed reduced ezrin phosphorylation and podocalyxin expression as well as reduced F-actin. Thus, SLK deletion leads to podocyte injury as mice age and exacerbates injury in adriamycin nephrosis. The mechanism may at least in part involve ezrin phosphorylation as well as disruption of the cytoskeleton and podocyte apical membrane structure.
Collapse
Affiliation(s)
- Boyan Woychyshyn
- Departments of Medicine and Physiology, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| | - Joan Papillon
- Departments of Medicine and Physiology, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| | - Julie Guillemette
- Departments of Medicine and Physiology, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| | - José R Navarro-Betancourt
- Departments of Medicine and Physiology, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| | - Andrey V Cybulsky
- Departments of Medicine and Physiology, McGill University Health Centre Research Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Atchison DK, O'Connor CL, Menon R, Otto EA, Ganesh SK, Wiggins RC, Smrcka AV, Bitzer M. Hypertension induces glomerulosclerosis in phospholipase C-ε1 deficiency. Am J Physiol Renal Physiol 2020; 318:F1177-F1187. [PMID: 32223311 DOI: 10.1152/ajprenal.00541.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Loss-of-function mutations in phospholipase C-ε1 (PLCE1) have been detected in patients with nephrotic syndrome, but other family members with the same mutation were asymptomatic, suggesting additional stressor are required to cause the full phenotype. Consistent with these observations, we determined that global Plce1-deficient mice have histologically normal glomeruli and no albuminuria at baseline. Angiotensin II (ANG II) is known to induce glomerular damage in genetically susceptible individuals. Therefore, we tested whether ANG II enhances glomerular damage in Plce1-deficient mice. ANG II increased blood pressure equally in Plce1-deficient and wild-type littermates. Additionally, it led to 20-fold increased albuminuria and significantly more sclerotic glomeruli in Plce1-deficient mice compared with wild-type littermates. Furthermore, Plce1-deficient mice demonstrated diffuse mesangial expansion, podocyte loss, and focal podocyte foot process effacement. To determine whether these effects are mediated by hypertension and hyperfiltration, rather than directly through ANG II, we raised blood pressure to a similar level using DOCA + salt + uninephrectomy and norepinephrine. This caused a fivefold increase in albuminuria in Plce1-deficient mice and a significant increase in the number of sclerotic glomeruli. Consistent with previous findings in mice, we detected strong PLCE1 transcript expression in podocytes using single cell sequencing of human kidney tissue. In hemagglutinin-tagged Plce1 transgenic mice, Plce1 was detected in podocytes and also in glomerular arterioles using immunohistochemistry. Our data demonstrate that Plce1 deficiency in mice predisposes to glomerular damage secondary to hypertensive insults.
Collapse
Affiliation(s)
- Douglas K Atchison
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Christopher L O'Connor
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Edgar A Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Santhi K Ganesh
- Cardiovascular Division, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Roger C Wiggins
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Markus Bitzer
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
Subramanian A, Sidhom EH, Emani M, Vernon K, Sahakian N, Zhou Y, Kost-Alimova M, Slyper M, Waldman J, Dionne D, Nguyen LT, Weins A, Marshall JL, Rosenblatt-Rosen O, Regev A, Greka A. Single cell census of human kidney organoids shows reproducibility and diminished off-target cells after transplantation. Nat Commun 2019; 10:5462. [PMID: 31784515 DOI: 10.0.4.14/s41467-019-13382-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/05/2019] [Indexed: 05/24/2023] Open
Abstract
Human iPSC-derived kidney organoids have the potential to revolutionize discovery, but assessing their consistency and reproducibility across iPSC lines, and reducing the generation of off-target cells remain an open challenge. Here, we profile four human iPSC lines for a total of 450,118 single cells to show how organoid composition and development are comparable to human fetal and adult kidneys. Although cell classes are largely reproducible across time points, protocols, and replicates, we detect variability in cell proportions between different iPSC lines, largely due to off-target cells. To address this, we analyze organoids transplanted under the mouse kidney capsule and find diminished off-target cells. Our work shows how single cell RNA-seq (scRNA-seq) can score organoids for reproducibility, faithfulness and quality, that kidney organoids derived from different iPSC lines are comparable surrogates for human kidney, and that transplantation enhances their formation by diminishing off-target cells.
Collapse
Affiliation(s)
| | - Eriene-Heidi Sidhom
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Katherine Vernon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Yiming Zhou
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Maria Kost-Alimova
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michal Slyper
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Waldman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Lan T Nguyen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Astrid Weins
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
17
|
Subramanian A, Sidhom EH, Emani M, Vernon K, Sahakian N, Zhou Y, Kost-Alimova M, Slyper M, Waldman J, Dionne D, Nguyen LT, Weins A, Marshall JL, Rosenblatt-Rosen O, Regev A, Greka A. Single cell census of human kidney organoids shows reproducibility and diminished off-target cells after transplantation. Nat Commun 2019; 10:5462. [PMID: 31784515 PMCID: PMC6884507 DOI: 10.1038/s41467-019-13382-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Human iPSC-derived kidney organoids have the potential to revolutionize discovery, but assessing their consistency and reproducibility across iPSC lines, and reducing the generation of off-target cells remain an open challenge. Here, we profile four human iPSC lines for a total of 450,118 single cells to show how organoid composition and development are comparable to human fetal and adult kidneys. Although cell classes are largely reproducible across time points, protocols, and replicates, we detect variability in cell proportions between different iPSC lines, largely due to off-target cells. To address this, we analyze organoids transplanted under the mouse kidney capsule and find diminished off-target cells. Our work shows how single cell RNA-seq (scRNA-seq) can score organoids for reproducibility, faithfulness and quality, that kidney organoids derived from different iPSC lines are comparable surrogates for human kidney, and that transplantation enhances their formation by diminishing off-target cells.
Collapse
Affiliation(s)
| | - Eriene-Heidi Sidhom
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Katherine Vernon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Yiming Zhou
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Maria Kost-Alimova
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michal Slyper
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Waldman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Lan T Nguyen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Astrid Weins
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Ren W, Yi H, Bao Y, Liu Y, Gao X. Oestrogen inhibits PTPRO to prevent the apoptosis of renal podocytes. Exp Ther Med 2019; 17:2373-2380. [PMID: 30783489 DOI: 10.3892/etm.2019.7167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 11/07/2018] [Indexed: 12/20/2022] Open
Abstract
Podocytes are a major component of the glomerular filtration membrane, and their apoptosis is involved in a variety of nephrotic syndromes. In the current study, the effects and molecular mechanisms of oestrogen on the proliferation and apoptosis of podocytes were investigated to elucidate the role of oestrogen in the pathogenesis of childhood nephrotic syndrome. The cell proliferation of mouse renal podocytes (MPC-5) and human primary renal podocytes was promoted by 17β-oestradiol (E2) in what appear to be a time-dependent manner. Apoptosis was inhibited by E2 and promoted by the E2 antagonist, tamoxifen. The expression of protein tyrosine phosphatase receptor type O (PTPRO) decreased with the increasing dosage of E2, but increased with the increasing dosage tamoxifen in MPC-5 and human podocytes. The protein, oestrogen receptor (ER)α, was not expressed in MPC-5 and human podocytes. E2 binding to ERβ completely eliminated PTPRO expression in MPC-5. In podocytes, PTPRO was phosphorylated by E2 at the Y1007 and associated with tyrosine-protein kinase JAK2 (JAK2) activation, rather than JAK1 activation. PTPRO was involved in the binding of E2 to signal transducer and activator of transcription (STAT)3 at the Y705 and S727 sites, resulting in the phosphorylation of STAT3 in podocytes. Through PTPRO, E2 also regulated the proliferation and apoptosis of podocytes. In conclusion, oestrogen binding to ERβ, rather than ERα, promoted the proliferation of podocytes and inhibited the apoptosis of podocytes by inhibiting the expression of PTPRO. The mechanism may be associated with the activation of the JAK2/STAT3 signalling pathway. The current study may provide a novel direction for the treatment of childhood nephrotic syndrome.
Collapse
Affiliation(s)
- Wei Ren
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Huiru Yi
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Ying Bao
- Department of Nephrology, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Yingru Liu
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Xinru Gao
- Department of Medical Ultrasound Center, The Northwest Women's and Children's Hospital, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
19
|
|
20
|
Identification of Protein Tyrosine Phosphatase Receptor Type O (PTPRO) as a Synaptic Adhesion Molecule that Promotes Synapse Formation. J Neurosci 2017; 37:9828-9843. [PMID: 28871037 DOI: 10.1523/jneurosci.0729-17.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/26/2017] [Accepted: 08/22/2017] [Indexed: 01/07/2023] Open
Abstract
The proper formation of synapses-specialized unitary structures formed between two neurons-is critical to mediating information flow in the brain. Synaptic cell adhesion molecules (CAMs) are thought to participate in the initiation of the synapse formation process. However, in vivo functional analysis demonstrates that most well known synaptic CAMs regulate synaptic maturation and plasticity rather than synapse formation, suggesting that either CAMs work synergistically in the process of forming synapses or more CAMs remain to be found. By screening for unknown CAMs using a co-culture system, we revealed that protein tyrosine phosphatase receptor type O (PTPRO) is a potent CAM that induces the formation of artificial synapse clusters in co-cultures of human embryonic kidney 293 cells and hippocampal neurons cultured from newborn mice regardless of gender. PTPRO was enriched in the mouse brain and localized to postsynaptic sites at excitatory synapses. The overexpression of PTPRO in cultured hippocampal neurons increased the number of synapses and the frequency of miniature EPSCs (mEPSCs). The knock-down (KD) of PTPRO expression in cultured neurons by short hairpin RNA (shRNA) reduced the number of synapses and the frequencies of the mEPSCs. The effects of shRNA KD were rescued by expressing either full-length PTPRO or a truncated PTPRO lacking the cytoplasmic domain. Consistent with these results, the N-terminal extracellular domain of PTPRO was required for its synaptogenic activity in the co-culture assay. Our data show that PTPRO is a synaptic CAM that serves as a potent initiator of the formation of excitatory synapses.SIGNIFICANCE STATEMENT The formation of synapses is critical for the brain to execute its function and synaptic cell adhesion molecules (CAMs) play essential roles in initiating the formation of synapses. By screening for unknown CAMs using a co-culture system, we revealed that protein tyrosine phosphatase receptor type O (PTPRO) is a potent CAM that induces the formation of artificial synapse clusters. Using loss-of-function and gain-of-function approaches, we show that PTPRO promotes the formation of excitatory synapses. The N-terminal extracellular domain of PTPRO was required for its synaptogenic activity in cultured hippocampal neurons and the co-culture assay. Together, our data show that PTPRO is a synaptic CAM that serves as a potent initiator of synapse formation.
Collapse
|
21
|
Unraveling the podocyte injury in lupus nephritis: Clinical and experimental approaches. Semin Arthritis Rheum 2017; 46:632-641. [DOI: 10.1016/j.semarthrit.2016.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/08/2016] [Accepted: 10/10/2016] [Indexed: 12/15/2022]
|
22
|
Regulation of Nephrin Phosphorylation in Diabetes and Chronic Kidney Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [PMID: 28639250 DOI: 10.1007/5584_2017_62] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Diabetes is the leading cause of microalbuminuria and end-stage renal failure in industrial countries. Disruption of the filtration barrier, seen in almost all nephrotic diseases and diabetes, is the result of the loss or effacement of the podocyte foot process, notably damage of proteins within the slit diaphragm such as nephrin. For many years, nephrin has been viewed as a structural component of the slit diaphragm. It is now well recognized that nephrin contains several tyrosine residues in its cytoplasmic domain, which influences the development of glomerular injury. In this review, we propose an overview of nephrin signaling pathways in kidney injury.
Collapse
|
23
|
Three-dimensional electron microscopy reveals the evolution of glomerular barrier injury. Sci Rep 2016; 6:35068. [PMID: 27725732 PMCID: PMC5057164 DOI: 10.1038/srep35068] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/20/2016] [Indexed: 12/26/2022] Open
Abstract
Glomeruli are highly sophisticated filters and glomerular disease is the leading cause of kidney failure. Morphological change in glomerular podocytes and the underlying basement membrane are frequently observed in disease, irrespective of the underlying molecular etiology. Standard electron microscopy techniques have enabled the identification and classification of glomerular diseases based on two-dimensional information, however complex three-dimensional ultrastructural relationships between cells and their extracellular matrix cannot be easily resolved with this approach. We employed serial block face-scanning electron microscopy to investigate Alport syndrome, the commonest monogenic glomerular disease, and compared findings to other genetic mouse models of glomerular disease (Myo1e−/−, Ptpro−/−). These analyses revealed the evolution of basement membrane and cellular defects through the progression of glomerular injury. Specifically we identified sub-podocyte expansions of the basement membrane with both cellular and matrix gene defects and found a corresponding reduction in podocyte foot process number. Furthermore, we discovered novel podocyte protrusions invading into the glomerular basement membrane in disease and these occurred frequently in expanded regions of basement membrane. These findings provide new insights into mechanisms of glomerular barrier dysfunction and suggest that common cell-matrix-adhesion pathways are involved in the progression of disease regardless of the primary insult.
Collapse
|
24
|
Novel role of Vav1-Rac1 pathway in actin cytoskeleton regulation in interleukin-13-induced minimal change-like nephropathy. Clin Sci (Lond) 2016; 130:2317-2327. [PMID: 27707912 DOI: 10.1042/cs20160312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/09/2016] [Accepted: 10/04/2016] [Indexed: 11/17/2022]
Abstract
Our established interleukin-13 (IL-13) overexpression rat model of minimal change-like nephropathy provided a platform to study the molecular signalling pathways in T-helper 2 (Th2) cytokine associated minimal change nephrotic syndrome (MCNS). We hypothesized that IL-13 may act directly on podocytes, causing podocyte foot process effacement and hence proteinuria in our rat model of minimal change-like nephropathy. The present study aimed firstly to delineate the glomerular 'gene signature' associated with IL-13-mediated dysregulation of podocyte-related proteins, and subsequently to investigate the role of the differentially regulated genes (DEGs) in IL-13-mediated podocyte injury. Glomerular transcriptional profile of IL-13-overexpressed rats showed characteristic features of podocyte injury with 87% of podocyte-related genes being significantly down-regulated. Gene expression of Vav1 was shown to be highly up-regulated in the glomeruli of IL-13-overexpressed rats and pathway analysis of the DEGs suggested a possible novel role of Vav1 in podocyte cytoskeleton remodelling. Immunofluorescence examination demonstrated glomerular expression of Vav1 in rats which co-localized with synaptopodin, confirming podocyte expression. However, positive staining for the phosphorylated form of Vav1 (p-Vav1) was only seen in IL-13-overexpressed rats. Moreover, in vitro IL-13 stimulation of human podocytes resulted in phosphorylation of Vav1. This was associated with Rac1 activation and actin cytoskeleton rearrangement, which was abrogated in Vav1 knockdown podocytes. In conclusion, we have demonstrated the role of Vav1-Rac1 pathway characterized by phosphorylation of Vav1, activation of Rac1 and the subsequent actin cytoskeleton rearrangement in IL-13-induced podocyte injury, possibly explaining the podocyte foot process effacement seen in our IL-13 overexpression rat model.
Collapse
|
25
|
Tian J, Wang HP, Mao YY, Jin J, Chen JH. Reduced Glomerular Epithelial Protein 1 Expression and Podocyte Injury in Immunoglobulin a Nephropathy. J Int Med Res 2016; 35:338-45. [PMID: 17593862 DOI: 10.1177/147323000703500308] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Expression of glomerular epithelial protein 1 (GLEPP1), a receptor tyrosine phosphatase present on the apical cell surface of the glomerular podocyte, and podocyte morphology were investigated in renal specimens from 51 patients with biopsy-diagnosed immunoglobulin A nephropathy (IgAN) and 11 controls. Clinical parameters, such as daily proteinuria were obtained from the patients' records and pathological manifestations of IgAN in the specimens were graded. GLEPP1 was strongly expressed and diffusely distributed in the glomeruli of control specimens. GLEPP1 expression was reduced in IgAN, especially in patients with nephrotic proteinuria and severe pathological manifestations. Podocyte injury was evident in IgAN and was associated with lower GLEPP1 expression and higher pathological grade. GLEPP1 expression was also significantly associated with clinical parameters. The results of this study suggest that GLEPP1 expression may be a useful marker of podocyte injury in IgAN, and may be predictive of clinical and pathological severity.
Collapse
Affiliation(s)
- J Tian
- Kidney Disease Centre, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | |
Collapse
|
26
|
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
|
27
|
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: 3.9] [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]
|
28
|
Abstract
The function of the kidney, filtering blood and concentrating metabolic waste into urine, takes place in an intricate and functionally elegant structure called the renal glomerulus. Normal glomerular function retains circulating cells and valuable macromolecular components of plasma in blood, resulting in urine with just trace amounts of proteins. Endothelial cells of glomerular capillaries, the podocytes wrapped around them, and the fused extracellular matrix these cells form altogether comprise the glomerular filtration barrier, a dynamic and highly selective filter that sieves on the basis of molecular size and electrical charge. Current understanding of the structural organization and the cellular and molecular basis of renal filtration draws from studies of human glomerular diseases and animal models of glomerular dysfunction.
Collapse
Affiliation(s)
- Rizaldy P Scott
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Susan E Quaggin
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
29
|
McClelland KS, Bell K, Larney C, Harley VR, Sinclair AH, Oshlack A, Koopman P, Bowles J. Purification and Transcriptomic Analysis of Mouse Fetal Leydig Cells Reveals Candidate Genes for Specification of Gonadal Steroidogenic Cells1. Biol Reprod 2015; 92:145. [DOI: 10.1095/biolreprod.115.128918] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/02/2015] [Indexed: 01/12/2023] Open
|
30
|
Redina OE, Smolenskaya SE, Klimov LO, Markel AL. Candidate genes in quantitative trait loci associated with absolute and relative kidney weight in rats with Inherited Stress Induced Arterial Hypertension. BMC Genet 2015; 16 Suppl 1:S1. [PMID: 25707311 PMCID: PMC4331803 DOI: 10.1186/1471-2156-16-s1-s1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The kidney mass is significantly increased in hypertensive ISIAH rats with Inherited Stress Induced Arterial Hypertension as compared with normotensive WAG rats. The QTL/microarray approach was carried out to determine the positional candidate genes in the QTL for absolute and relative kidney weight. RESULTS Several known and predicted genes differentially expressed in ISIAH and WAG kidney were mapped to genetic loci associated with the absolute and relative kidney weight in 6-month old F2 hybrid (ISIAHxWAG) males. The knowledge-driven filtering of the list of candidates helped to suggest several positional candidate genes, which may be related to the structural and mass changes in hypertensive ISIAH kidney. CONCLUSIONS The further experimental validation of causative genes and detection of polymorphisms will provide opportunities to advance our understanding of the underlying nature of structural and mass changes in hypertensive ISIAH kidney.
Collapse
|
31
|
Genetic causes of proteinuria and nephrotic syndrome: impact on podocyte pathobiology. Pediatr Nephrol 2015; 30:221-33. [PMID: 24584664 PMCID: PMC4262721 DOI: 10.1007/s00467-014-2753-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 12/31/2013] [Accepted: 01/02/2014] [Indexed: 12/11/2022]
Abstract
In the past 20 years, multiple genetic mutations have been identified in patients with congenital nephrotic syndrome (CNS) and both familial and sporadic focal segmental glomerulosclerosis (FSGS). Characterization of the genetic basis of CNS and FSGS has led to the recognition of the importance of podocyte injury to the development of glomerulosclerosis. Genetic mutations induce injury due to effects on the podocyte's structure, actin cytoskeleton, calcium signaling, and lysosomal and mitochondrial function. Transgenic animal studies have contributed to our understanding of podocyte pathobiology. Podocyte endoplasmic reticulum stress response, cell polarity, and autophagy play a role in maintenance of podocyte health. Further investigations related to the effects of genetic mutations on podocytes may identify new pathways for targeting therapeutics for nephrotic syndrome.
Collapse
|
32
|
Motiwala T, Kutay H, Zanesi N, Frissora FW, Mo X, Muthusamy N, Jacob ST. PTPROt-mediated regulation of p53/Foxm1 suppresses leukemic phenotype in a CLL mouse model. Leukemia 2014; 29:1350-9. [PMID: 25482129 PMCID: PMC4456291 DOI: 10.1038/leu.2014.341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/27/2014] [Accepted: 11/11/2014] [Indexed: 12/17/2022]
Abstract
The gene encoding PTPROt is methylated and suppressed in Chronic Lymphocytc Leukemia. PTPROt exhibits in vitro tumor suppressor characteristics through the regulation of B-cell receptor signaling. Here, we generated transgenic (Tg) mice with B-cell specific expression of PTPROt. While lymphocyte development is normal in these mice, crossing them with TCL1 Tg mouse model of CLL results in a survival advantage compared to the TCL1 Tg mice. Gene expression profiling of splenic B-lymphocytes before detectable signs of CLL followed by Ingenuity Pathway Analysis revealed that the most prominently regulated functions in TCL1 Tg vs non-transgenic (NTg) and TCL1 Tg vs PTPROt/TCL1 double Tg are the same and also biologically relevant to this study. Further, enhanced expression of the chemokine Ccl3, the oncogenic transcription factor Foxm1 and its targets in TCL1 Tg mice were significantly suppressed in the double Tg mice suggesting a protective function of PTPROt against leukemogenesis. This study also showed that PTPROt mediated regulation of Foxm1 involves activation of p53, a transcriptional repressor of Foxm1, which is facilitated through suppression of B-cell receptor signaling. These results establish the in vivo tumor suppressive function of PTPROt, and identify p53/Foxm1 axis as a key downstream effect of PTPROt-mediated suppression of BCR signaling.
Collapse
Affiliation(s)
- T Motiwala
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA
| | - H Kutay
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA
| | - N Zanesi
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH, USA
| | - F W Frissora
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - X Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - N Muthusamy
- Department of Internal Medicine, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - S T Jacob
- 1] Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH, USA [2] Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
33
|
Denhez B, Lizotte F, Guimond MO, Jones N, Takano T, Geraldes P. Increased SHP-1 protein expression by high glucose levels reduces nephrin phosphorylation in podocytes. J Biol Chem 2014; 290:350-8. [PMID: 25404734 DOI: 10.1074/jbc.m114.612721] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Nephrin, a critical podocyte membrane component that is reduced in diabetic nephropathy, has been shown to activate phosphotyrosine signaling pathways in human podocytes. Nephrin signaling is important to reduce cell death induced by apoptotic stimuli. We have shown previously that high glucose level exposure and diabetes increased the expression of SHP-1, causing podocyte apoptosis. SHP-1 possesses two Src homology 2 domains that serve as docking elements to dephosphorylate tyrosine residues of target proteins. However, it remains unknown whether SHP-1 interacts with nephrin and whether its elevated expression affects the nephrin phosphorylation state in diabetes. Here we show that human podocytes exposed to high glucose levels exhibited elevated expression of SHP-1, which was associated with nephrin. Coexpression of nephrin-CD16 and SHP-1 reduced nephrin tyrosine phosphorylation in transfected human embryonic kidney 293 cells. A single tyrosine-to-phenylalanine mutation revealed that rat nephrin Tyr(1127) and Tyr(1152) are required to allow SHP-1 interaction with nephrin. Overexpression of dominant negative SHP-1 in human podocytes prevented high glucose-induced reduction of nephrin phosphorylation. In vivo, immunoblot analysis demonstrated that nephrin expression and phosphorylation were decreased in glomeruli of type 1 diabetic Akita mice (Ins2(+/C96Y)) compared with control littermate mice (Ins2(+/+)), and this was associated with elevated SHP-1 and cleaved caspase-3 expression. Furthermore, immunofluorescence analysis indicated increased colocalization of SHP-1 with nephrin in diabetic mice compared with control littermates. In conclusion, our results demonstrate that high glucose exposure increases SHP-1 interaction with nephrin, causing decreased nephrin phosphorylation, which may, in turn, contribute to diabetic nephropathy.
Collapse
Affiliation(s)
- Benoit Denhez
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Departments of Medicine, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Farah Lizotte
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Departments of Medicine, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Marie-Odile Guimond
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Departments of Medicine, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Nina Jones
- the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada, and
| | - Tomoko Takano
- the McGill University Health Center, Montreal, Québec H3H 2R9, Canada
| | - Pedro Geraldes
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke and Division of Endocrinology, Departments of Medicine, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada,
| |
Collapse
|
34
|
Jeon M, Zinn K. R3 receptor tyrosine phosphatases: conserved regulators of receptor tyrosine kinase signaling and tubular organ development. Semin Cell Dev Biol 2014; 37:119-26. [PMID: 25242281 DOI: 10.1016/j.semcdb.2014.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 09/04/2014] [Indexed: 12/25/2022]
Abstract
R3 receptor tyrosine phosphatases (RPTPs) are characterized by extracellular domains composed solely of long chains of fibronectin type III repeats, and by the presence of a single phosphatase domain. There are five proteins in mammals with this structure, two in Drosophila and one in Caenorhabditis elegans. R3 RPTPs are selective regulators of receptor tyrosine kinase (RTK) signaling, and a number of different RTKs have been shown to be direct targets for their phosphatase activities. Genetic studies in both invertebrate model systems and in mammals have shown that R3 RPTPs are essential for tubular organ development. They also have important functions during nervous system development. R3 RPTPs are likely to be tumor suppressors in a number of types of cancer.
Collapse
Affiliation(s)
- Mili Jeon
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States; Department of Molecular and Cellular Physiology and Structural Biology, Howard Hughes Medical Institute, Stanford School of Medicine, Palo Alto, CA 94305, United States
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States.
| |
Collapse
|
35
|
Kumagai T, Baldwin C, Aoudjit L, Nezvitsky L, Robins R, Jiang R, Takano T. Protein Tyrosine Phosphatase 1B Inhibition Protects against Podocyte Injury and Proteinuria. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2211-24. [DOI: 10.1016/j.ajpath.2014.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 04/12/2014] [Accepted: 05/09/2014] [Indexed: 12/18/2022]
|
36
|
Bierzynska A, Soderquest K, Koziell A. Genes and podocytes - new insights into mechanisms of podocytopathy. Front Endocrinol (Lausanne) 2014; 5:226. [PMID: 25667580 PMCID: PMC4304234 DOI: 10.3389/fendo.2014.00226] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 12/09/2014] [Indexed: 12/24/2022] Open
Abstract
After decades of primarily morphological study, positional cloning of the NPHS1 gene was the landmark event that established aberrant podocyte genetics as a pivotal cause of malfunction of the glomerular filter. This ended any uncertainty whether genetic mutation plays a significant role in hereditary nephrotic syndromes (NS) and confirmed podocytes as critical players in regulating glomerular protein filtration. Although subsequent sequencing of candidate genes chosen on the basis of podocyte biology had less success, unbiased analysis of genetically informative kindreds and syndromic disease has led to further gene discovery. However, the 45 genes currently associated with human NS explain not more than 20-30% of hereditary and only 10-20% of sporadic cases. It is becoming increasingly clear both from genetic analysis and phenotypic data - including occasional response to immunosuppressive agents and post-transplant disease recurrence in Mendelian disease - that monogenic inheritance of abnormalities in podocyte-specific genes disrupting filter function is only part of the story. Recent advances in genetic screening technology combined with increasingly robust bioinformatics are set to allow identification and characterization of novel disease causing variants and more importantly, disease modifying genes. Emerging data also support a significant but incompletely characterized immunoregulatory component.
Collapse
Affiliation(s)
- Agnieszka Bierzynska
- Academic Renal Unit, School of Clinical Sciences, Bristol University, Bristol, UK
| | - Katrina Soderquest
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Ania Koziell
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, Faculty of Life Sciences and Medicine, King’s College London, London, UK
- *Correspondence: Ania Koziell, Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, Faculty of Life Sciences and Medicine, 5th Floor Tower Wing, Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK e-mail:
| |
Collapse
|
37
|
A compendium of urinary biomarkers indicative of glomerular podocytopathy. PATHOLOGY RESEARCH INTERNATIONAL 2013; 2013:782395. [PMID: 24327929 PMCID: PMC3845336 DOI: 10.1155/2013/782395] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/10/2013] [Indexed: 12/18/2022]
Abstract
It is well known that glomerular podocyte injury and loss are present in numerous nephropathies and that the pathophysiologic consecution of disease hinges upon the fate of the podocyte. While multiple factors play a hand in glomerulopathy progression, basic logic lends that if one monitors the podocyte's status, that may reflect the status of disease. Recent investigations have focused on what one can elucidate from the noninvasive collection of urine, and have proven that certain, specific biomarkers of podocytes can be readily identified via varying techniques. This paper has brought together all described urinary biomarkers of podocyte injury and is made to provide a concise summary of their utility and testing in laboratory and clinical theatres. While promising in the potential that they hold as tools for clinicians and investigators, the described biomarkers require further comprehensive vetting in the form of larger clinical trials and studies that would give their value true weight. These urinary biomarkers are put forth as novel indicators of glomerular disease presence, disease progression, and therapeutic efficacy that in some cases may be more advantageous than the established parameters/measures currently used in practice.
Collapse
|
38
|
Huang YT, Li FF, Ke C, Li Z, Li ZT, Zou XF, Zheng XX, Chen YP, Zhang H. PTPRO promoter methylation is predictive of poorer outcome for HER2-positive breast cancer: indication for personalized therapy. J Transl Med 2013; 11:245. [PMID: 24090193 PMCID: PMC3852714 DOI: 10.1186/1479-5876-11-245] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/25/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Protein Tyrosine Phosphatase Receptor-type O (PTPRO) has recently been in the spotlight as a tumor suppressor, whose encoding gene is frequently methylated in cancers. We examined the methylation status of the PTPRO gene promoter in breast cancer and evaluated the correlation between PTPRO promoter methylation and both clinicopathological parameters and prognosis of breast cancer patients. METHODS Two hundred twenty-one formalin-fixed, paraffin-embedded (FFPE) tumor tissues, 20 FFPE normal adjacent tissues and 24 matched plasma samples, collected from primary breast cancer patients, were assessed for PTPRO gene promoter methylation using methylation-specific PCR. Associations of promoter methylation with clinicopathological parameters were evaluated. Kaplan-Meier survival analysis and Cox proportional hazards models were used to estimate the effect on survival. RESULTS 175 samples gave identifiable PCR products, of which 130 cases (74.3%) had PTPRO gene promoter methylation. PTPRO methylation correlated with higher histological grade (P = 0.028), but not other clinical parameters. Multivariate analysis indicated that overall survival (OS) was significantly poorer in HER2-positive, but not ER-positive patients with methylated-PTPRO. Methylated-PTPRO was detectable in matched plasma samples and only observed in plasma from patients whose corresponding primary tumors were also methylated. CONCLUSIONS PTPRO methylation is a common event in the primary breast cancer and can be reliably detected in peripheral blood samples. PTPRO methylation is associated with poor survival only in HER2-positive patients, suggesting use of PTPRO methylation as a prognostic factor for breast cancer and for optimizing individualized therapy for HER2-positive patients.
Collapse
Affiliation(s)
- Yi-Teng Huang
- Department of Integrative Oncology, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| | - Fei-Fei Li
- Department of Integrative Oncology, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| | - Chen Ke
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| | - Zhou Li
- Department of Integrative Oncology, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| | - Zong-Tai Li
- Department of Integrative Oncology, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| | - Xiao-Fang Zou
- Department of Integrative Oncology, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| | - Xiao-Xuan Zheng
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| | - Yu-Ping Chen
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
| | - Hao Zhang
- Department of Integrative Oncology, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Tumor Tissue Bank, Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
39
|
Grahammer F, Schell C, Huber TB. Molecular understanding of the slit diaphragm. Pediatr Nephrol 2013; 28:1957-62. [PMID: 23233041 DOI: 10.1007/s00467-012-2375-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 10/16/2012] [Accepted: 10/17/2012] [Indexed: 11/30/2022]
Abstract
Glomerular filtration has always attracted the interest of nephrologists and renal researchers alike. Although several key questions on the structure and function of the kidney filter may have been answered within the last 40 years of intense research, there still remain crucial questions to be solved. The following article attempts to give a brief overview of recent developments in glomerular research highlighting particular advances in our understanding of the slit diaphragm.
Collapse
Affiliation(s)
- Florian Grahammer
- Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, Freiburg 79106, Germany
| | | | | |
Collapse
|
40
|
Lasagni L, Lazzeri E, Shankland SJ, Anders HJ, Romagnani P. Podocyte mitosis - a catastrophe. Curr Mol Med 2013; 13:13-23. [PMID: 23176147 PMCID: PMC3624791 DOI: 10.2174/1566524011307010013] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/14/2012] [Accepted: 11/20/2012] [Indexed: 12/20/2022]
Abstract
Podocyte loss plays a key role in the progression of glomerular disorders towards glomerulosclerosis and chronic kidney disease. Podocytes form unique cytoplasmic extensions, foot processes, which attach to the outer surface of the glomerular basement membrane and interdigitate with neighboring podocytes to form the slit diaphragm. Maintaining these sophisticated structural elements requires an intricate actin cytoskeleton. Genetic, mechanic, and immunologic or toxic forms of podocyte injury can cause podocyte loss, which causes glomerular filtration barrier dysfunction, leading to proteinuria. Cell migration and cell division are two processes that require a rearrangement of the actin cytoskeleton; this rearrangement would disrupt the podocyte foot processes, therefore, podocytes have a limited capacity to divide or migrate. Indeed, all cells need to rearrange their actin cytoskeleton to assemble a correct mitotic spindle and to complete mitosis. Podocytes, even when being forced to bypass cell cycle checkpoints to initiate DNA synthesis and chromosome segregation, cannot complete cytokinesis efficiently and thus usually generate aneuploid podocytes. Such aneuploid podocytes rapidly detach and die, a process referred to as mitotic catastrophe. Thus, detached or dead podocytes cannot be adequately replaced by the proliferation of adjacent podocytes. However, even glomerular disorders with severe podocyte injury can undergo regression and remission, suggesting alternative mechanisms to compensate for podocyte loss, such as podocyte hypertrophy or podocyte regeneration from resident renal progenitor cells. Together, mitosis of the terminally differentiated podocyte rather accelerates podocyte loss and therefore glomerulosclerosis. Finding ways to enhance podocyte regeneration from other sources remains a challenge goal to improve the treatment of chronic kidney disease in the future.
Collapse
Affiliation(s)
- L Lasagni
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Viale Pieraccini 6, 50139, Firenze, Italy.
| | | | | | | | | |
Collapse
|
41
|
Grahammer F, Schell C, Huber TB. The podocyte slit diaphragm--from a thin grey line to a complex signalling hub. Nat Rev Nephrol 2013; 9:587-98. [PMID: 23999399 DOI: 10.1038/nrneph.2013.169] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The architectural design of our kidneys is amazingly complex, and culminates in the 3D structure of the glomerular filter. During filtration, plasma passes through a sieve consisting of a fenestrated endothelium and a broad basement membrane before it reaches the most unique part, the slit diaphragm, a specialized type of intercellular junction that connects neighbouring podocyte foot processes. When podocytes become stressed, irrespective of the causative stimulus, they undergo foot process effacement and loss of slit diaphragms--two key steps leading to proteinuria. Thus, proteinuria is the unifying denominator of a broad spectrum of podocytopathies. With the rising prevalence of chronic kidney disease and the fact that glomerular diseases account for the majority of patients with end-stage renal disease, further investigation and elucidation of this unique structure is of paramount importance. This Review recounts how perception of the slit diaphragm has changed over time as a result of intense research, from its first anatomical description as a thin intercellular connection, to an appreciation of its role as a dynamic signalling hub. These observations led to the introduction of novel concepts in podocyte biology, which could pave the way to development of highly desired, specific therapeutic strategies for glomerular diseases.
Collapse
Affiliation(s)
- Florian Grahammer
- Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, Freiburg 79106, Germany
| | | | | |
Collapse
|
42
|
Takahashi S, Tomioka M, Hiromura K, Sakairi T, Hamatani H, Watanabe M, Ikeuchi H, Kaneko Y, Maeshima A, Aoki T, Ohnishi H, Matozaki T, Nojima Y. SIRPα signaling regulates podocyte structure and function. Am J Physiol Renal Physiol 2013; 305:F861-70. [PMID: 23842779 DOI: 10.1152/ajprenal.00597.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Signal-regulatory protein-α (SIRPα) is a transmembrane protein that contains tyrosine phosphorylation sites in its cytoplasmic region; two tyrosine phosphatases, SHP-1 and SHP-2, bind to these sites in a phosphorylation-dependent manner and transduce multiple intracellular signals. Recently, SIRPα was identified as one of the major tyrosine-phosphorylated proteins in the glomeruli and found to be expressed in podocytes. In the present study, we examined the role of SIRPα expression in podocytes using knockin mice (C57BL/6 background) expressing mutant SIRPα that lacks a cytoplasmic region (SIRPα-mutant mice). Light microscopic examination revealed no apparent morphological abnormalities in the kidneys of the SIRPα-mutant mice. On the other hand, electron microscopic examination revealed abnormal podocytes with irregular major processes and wider and flattened foot processes in the SIRPα-mutant mice compared with their wild-type counterparts. Significantly impaired renal functions and slight albuminuria were demonstrated in the SIRPα-mutant mice. In addition, adriamycin injection induced massive albuminuria together with focal glomerulosclerosis in the SIRPα-mutant mice, while their wild-type counterparts were resistant to adriamycin-induced nephropathy. These data demonstrate that SIRPα is involved in the regulation of podocyte structure and function as a filtration barrier under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Satoshi Takahashi
- Dept. of Medicine and Clinical Science, Gunma Univ. Graduate School of Medicine, 3-39-22 Showa, Maebashi, Gunma 371-8511, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Protein tyrosine phosphatase receptor type O inhibits trigeminal axon growth and branching by repressing TrkB and Ret signaling. J Neurosci 2013; 33:5399-410. [PMID: 23516305 DOI: 10.1523/jneurosci.4707-12.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Axonal branches of the trigeminal ganglion (TG) display characteristic growth and arborization patterns during development. Subsets of TG neurons express different receptors for growth factors, but these are unlikely to explain the unique patterns of axonal arborizations. Intrinsic modulators may restrict or enhance cellular responses to specific ligands and thereby contribute to the development of axon growth patterns. Protein tyrosine phosphatase receptor type O (PTPRO), which is required for Eph receptor-dependent retinotectal development in chick and for development of subsets of trunk sensory neurons in mouse, may be such an intrinsic modulator of TG neuron development. PTPRO is expressed mainly in TrkB-expressing (TrkB(+)) and Ret(+) mechanoreceptors within the TG during embryogenesis. In PTPRO mutant mice, subsets of TG neurons grow longer and more elaborate axonal branches. Cultured PTPRO(-/-) TG neurons display enhanced axonal outgrowth and branching in response to BDNF and GDNF compared with control neurons, indicating that PTPRO negatively controls the activity of BDNF/TrkB and GDNF/Ret signaling. Mouse PTPRO fails to regulate Eph signaling in retinocollicular development and in hindlimb motor axon guidance, suggesting that chick and mouse PTPRO have different substrate specificities. PTPRO has evolved to fine tune growth factor signaling in a cell-type-specific manner and to thereby increase the diversity of signaling output of a limited number of receptor tyrosine kinases to control the branch morphology of developing sensory neurons. The regulation of Eph receptor-mediated developmental processes by protein tyrosine phosphatases has diverged between chick and mouse.
Collapse
|
45
|
Hendriks WJAJ, Pulido R. Protein tyrosine phosphatase variants in human hereditary disorders and disease susceptibilities. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1673-96. [PMID: 23707412 DOI: 10.1016/j.bbadis.2013.05.022] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/14/2013] [Accepted: 05/16/2013] [Indexed: 12/18/2022]
Abstract
Reversible tyrosine phosphorylation of proteins is a key regulatory mechanism to steer normal development and physiological functioning of multicellular organisms. Phosphotyrosine dephosphorylation is exerted by members of the super-family of protein tyrosine phosphatase (PTP) enzymes and many play such essential roles that a wide variety of hereditary disorders and disease susceptibilities in man are caused by PTP alleles. More than two decades of PTP research has resulted in a collection of PTP genetic variants with corresponding consequences at the molecular, cellular and physiological level. Here we present a comprehensive overview of these PTP gene variants that have been linked to disease states in man. Although the findings have direct bearing for disease diagnostics and for research on disease etiology, more work is necessary to translate this into therapies that alleviate the burden of these hereditary disorders and disease susceptibilities in man.
Collapse
Affiliation(s)
- Wiljan J A J Hendriks
- Department of Cell Biology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | |
Collapse
|
46
|
Chai OH, Song CH, Park SK, Kim W, Cho ES. Molecular regulation of kidney development. Anat Cell Biol 2013; 46:19-31. [PMID: 23560233 PMCID: PMC3615609 DOI: 10.5115/acb.2013.46.1.19] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 01/25/2013] [Accepted: 02/04/2013] [Indexed: 12/21/2022] Open
Abstract
Genetically engineered mice have provided much information about gene function in the field of developmental biology. Recently, conditional gene targeting using the Cre/loxP system has been developed to control the cell type and timing of the target gene expression. The increase in number of kidney-specific Cre mice allows for the analysis of phenotypes that cannot be addressed by conventional gene targeting. The mammalian kidney is a vital organ that plays a critical homeostatic role in the regulation of body fluid composition and excretion of waste products. The interactions between epithelial and mesenchymal cells are very critical events in the field of developmental biology, especially renal development. Kidney development is a complex process, requiring inductive interactions between epithelial and mesenchymal cells that eventually lead to the growth and differentiation of multiple highly specialized stromal, vascular, and epithelial cell types. Through the use of genetically engineered mouse models, the molecular bases for many of the events in the developing kidney have been identified. Defective morphogenesis may result in clinical phenotypes that range from complete renal agenesis to diseases such as hypertension that exist in the setting of grossly normal kidneys. In this review, we focus on the growth and transcription factors that define kidney progenitor cell populations, initiate ureteric bud branching, induce nephron formation within the metanephric mesenchyme, and differentiate stromal and vascular progenitors in the metanephric mesenchyme.
Collapse
Affiliation(s)
- Ok-Hee Chai
- Department of Anatomy, Institute for Medical Sciences, Chonbuk National University Medical School, Jeonju, Korea
| | | | | | | | | |
Collapse
|
47
|
Liao WH, Cheng CH, Hung KS, Chiu WT, Chen GD, Hwang PP, Hwang SPL, Kuan YS, Huang CJ. Protein tyrosine phosphatase receptor type O (Ptpro) regulates cerebellar formation during zebrafish development through modulating Fgf signaling. Cell Mol Life Sci 2013; 70:2367-81. [PMID: 23361036 PMCID: PMC3676743 DOI: 10.1007/s00018-013-1259-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 12/13/2012] [Accepted: 01/03/2013] [Indexed: 02/04/2023]
Abstract
Protein activities controlled by receptor protein tyrosine phosphatases (RPTPs) play comparably important roles in transducing cell surface signals into the cytoplasm by protein tyrosine kinases. Previous studies showed that several RPTPs are involved in neuronal generation, migration, and axon guidance in Drosophila, and the vertebrate hippocampus, retina, and developing limbs. However, whether the protein tyrosine phosphatase type O (ptpro), one kind of RPTP, participates in regulating vertebrate brain development is largely unknown. We isolated the zebrafish ptpro gene and found that its transcripts are primarily expressed in the embryonic and adult central nervous system. Depletion of zebrafish embryonic Ptpro by antisense morpholino oligonucleotide knockdown resulted in prominent defects in the forebrain and cerebellum, and the injected larvae died on the 4th day post-fertilization (dpf). We further investigated the function of ptpro in cerebellar development and found that the expression of ephrin-A5b (efnA5b), a Fgf signaling induced cerebellum patterning factor, was decreased while the expression of dusp6, a negative-feedback gene of Fgf signaling in the midbrain-hindbrain boundary region, was notably induced in ptpro morphants. Further analyses demonstrated that cerebellar defects of ptpro morphants were partially rescued by inhibiting Fgf signaling. Moreover, Ptpro physically interacted with the Fgf receptor 1a (Fgfr1a) and dephosphorylated Fgfr1a in a dose-dependant manner. Therefore, our findings demonstrate that Ptpro activity is required for patterning the zebrafish embryonic brain. Specifically, Ptpro regulates cerebellar formation during zebrafish development through modulating Fgf signaling.
Collapse
Affiliation(s)
- Wei-Hao Liao
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 104, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
The glomerular filtration barrier is a highly specialized blood filtration interface that displays a high conductance to small and midsized solutes in plasma but retains relative impermeability to macromolecules. Its integrity is maintained by physicochemical and signalling interplay among its three core constituents-the glomerular endothelial cell, the basement membrane and visceral epithelial cell (podocyte). Understanding the pathomechanisms of inherited and acquired human diseases as well as experimental injury models of this barrier have helped to unravel this interdependence. Key among the consequences of interference with the integrity of the glomerular filtration barrier is the appearance of significant amounts of proteins in the urine. Proteinuria correlates with kidney disease progression and cardiovascular mortality. With specific reference to proteinuria in human and animal disease phenotypes, the following review explores the roles of the endothelial cell, glomerular basement membrane, and the podocyte and attempts to highlight examples of essential crosstalk within this barrier.
Collapse
|
49
|
The glomerular filtration barrier: components and crosstalk. Int J Nephrol 2012; 2012:749010. [PMID: 22934182 PMCID: PMC3426247 DOI: 10.1155/2012/749010] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 06/02/2012] [Accepted: 06/05/2012] [Indexed: 01/18/2023] Open
Abstract
The glomerular filtration barrier is a highly specialized blood filtration interface that displays a high conductance to small and midsized solutes in plasma but retains relative impermeability to macromolecules. Its integrity is maintained by physicochemical and signalling interplay among its three core constituents—the glomerular endothelial cell, the basement membrane and visceral epithelial cell (podocyte). Understanding the pathomechanisms of inherited and acquired human diseases as well as experimental injury models of this barrier have helped to unravel this interdependence. Key among the consequences of interference with the integrity of the glomerular filtration barrier is the appearance of significant amounts of proteins in the urine. Proteinuria correlates with kidney disease progression and cardiovascular mortality. With specific reference to proteinuria in human and animal disease phenotypes, the following review explores the roles of the endothelial cell, glomerular basement membrane, and the podocyte and attempts to highlight examples of essential crosstalk within this barrier.
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Focal segmental glomerulosclerosis (FSGS) is a major cause of nephrotic syndrome and renal failure. All forms of FSGS share podocyte injury and depletion as central mediators. This review focuses on new insights into pathogenesis from study of extrinsic toxins in experimental models, permeability factors in human disease, and novel genetic causes. RECENT FINDINGS Experimental toxin models have advanced our understanding of the threshold and dynamics of podocyte injury. Following initial podocyte depletion, spreading fields of podocyte injury through secondary mediators appear to be important in generating the segmental pathologic lesions. Proliferating glomerular epithelial cells are common in FSGS, although there are conflicting views about their identity. Evidence suggests potential contributions by mature parietal epithelial cells, facultative stem cells and podocytes. A number of novel candidate permeability factors that affect podocyte function and motility have been discovered in human FSGS and related podocytopathy minimal change disease. Exome capture has identified new monogenic causes of familial FSGS. Apolipoprotein L-1 (APOL1) is expressed in podocytes, and the prevalence of APOL1 risk alleles in patients of African descent with primary FSGS and HIV-associated nephropathy is high, implicating potential podocyte effects. SUMMARY FSGS is caused by a complex interplay of inherent genetic susceptibilities and external injurious factors acting on podocytes. Critical levels of podocyte stress eventuate in podocyte depletion, segmental glomerular scarring, and glomerular epithelial cell hyperplasia.
Collapse
|