1
|
Miyaki T, Homma N, Kawasaki Y, Kishi M, Yamaguchi J, Kakuta S, Shindo T, Sugiura M, Oliva Trejo JA, Kaneda H, Omotehara T, Takechi M, Negishi-Koga T, Ishijima M, Aoto K, Iseki S, Kitamura K, Muto S, Amagasa M, Hotchi S, Ogura K, Shibata S, Sakai T, Suzuki Y, Ichimura K. Ultrastructural analysis of whole glomeruli using array tomography. J Cell Sci 2024; 137:jcs262154. [PMID: 39171439 DOI: 10.1242/jcs.262154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
The renal glomerulus produces primary urine from blood plasma by ultrafiltration. The ultrastructure of the glomerulus is closely related to filtration function and disease development. The ultrastructure of glomeruli has mainly been evaluated using transmission electron microscopy; however, the volume that can be observed using transmission electron microscopy is extremely limited relative to the total volume of the glomerulus. Consequently, observing structures that exist in only one location in each glomerulus, such as the vascular pole, and evaluating low-density or localized lesions are challenging tasks. Array tomography (AT) is a technique used to analyze the ultrastructure of tissues and cells via scanning electron microscopy of serial sections. In this study, we present an AT workflow that is optimized for observing complete serial sections of the whole glomerulus, and we share several analytical examples that use the optimized AT workflow, demonstrating the usefulness of this approach. Overall, this AT workflow can be a powerful tool for structural and pathological evaluation of the glomerulus. This workflow is also expected to provide new insights into the ultrastructure of the glomerulus and its constituent cells.
Collapse
Affiliation(s)
- Takayuki Miyaki
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Nozomi Homma
- Department of Nephrology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yuto Kawasaki
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Mami Kishi
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Junji Yamaguchi
- Laboratory of Morphology and Image Analysis, Research Core Facilities , Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Research Core Facilities , Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Tomoko Shindo
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo 160-0016, Japan
| | - Makoto Sugiura
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Juan Alejandro Oliva Trejo
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Hisako Kaneda
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takuya Omotehara
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Masaki Takechi
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takako Negishi-Koga
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Department of Community Medicine and Research for Bone and Joint Diseases, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Department of Community Medicine and Research for Bone and Joint Diseases, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Kazushi Aoto
- Central Laboratory, Graduate School of Biomedical and Health Sciences , Hiroshima University, Hiroshima 734-8551, Japan
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences , Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Kosuke Kitamura
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Satoru Muto
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Mao Amagasa
- Department of Human Pathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Shiori Hotchi
- Department of Human Pathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Kanako Ogura
- Department of Human Pathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo 160-0016, Japan
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences , Niigata University, Niigata City 951-8510, Japan
| | - Tatsuo Sakai
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Laboratory of Morphology and Image Analysis, Research Core Facilities , Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| |
Collapse
|
2
|
Hu S, Hang X, Wei Y, Wang H, Zhang L, Zhao L. Crosstalk among podocytes, glomerular endothelial cells and mesangial cells in diabetic kidney disease: an updated review. Cell Commun Signal 2024; 22:136. [PMID: 38374141 PMCID: PMC10875896 DOI: 10.1186/s12964-024-01502-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 01/28/2024] [Indexed: 02/21/2024] Open
Abstract
Diabetic kidney disease (DKD) is a long-term and serious complication of diabetes that affects millions of people worldwide. It is characterized by proteinuria, glomerular damage, and renal fibrosis, leading to end-stage renal disease, and the pathogenesis is complex and involves multiple cellular and molecular mechanisms. Among three kinds of intraglomerular cells including podocytes, glomerular endothelial cells (GECs) and mesangial cells (MCs), the alterations in one cell type can produce changes in the others. The cell-to-cell crosstalk plays a crucial role in maintaining the glomerular filtration barrier (GFB) and homeostasis. In this review, we summarized the recent advances in understanding the pathological changes and interactions of these three types of cells in DKD and then focused on the signaling pathways and factors that mediate the crosstalk, such as angiopoietins, vascular endothelial growth factors, transforming growth factor-β, Krüppel-like factors, retinoic acid receptor response protein 1 and exosomes, etc. Furthermore, we also simply introduce the application of the latest technologies in studying cell interactions within glomerular cells and new promising mediators for cell crosstalk in DKD. In conclusion, this review provides a comprehensive and updated overview of the glomerular crosstalk in DKD and highlights its importance for the development of novel intervention approaches.
Collapse
Affiliation(s)
- Shiwan Hu
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xing Hang
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu Wei
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Han Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Lili Zhang
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
3
|
Wu Q, Zhou S, Xu D, Meng P, Chen Q, Wang X, Li X, Chen S, Ye H, Ye W, Xiong Y, Li J, Miao J, Shen W, Lin X, Hou FF, Liu Y, Zhang Y, Zhou L. The CXCR4-AT1 axis plays a vital role in glomerular injury via mediating the crosstalk between podocyte and mesangial cell. Transl Res 2024; 264:15-32. [PMID: 37696390 DOI: 10.1016/j.trsl.2023.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 08/18/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023]
Abstract
Glomeruli stand at the center of nephrons to accomplish filtration and albumin interception. Podocytes and mesangial cells are the major constituents in the glomeruli. However, their interdependency in glomerular injury has rarely been reported. Herein, we investigated the role of C-X-C chemokine receptor type 4 (CXCR4) in mediating the crosstalk between podocytes and mesangial cells. We found CXCR4 and angiotensin II (AngII) increased primarily in injured podocytes. However, type-1 receptor of angiotensin II (AT1) and stromal cell-derived factor 1α (SDF-1α), a ligand of CXCR4, were evidently upregulated in mesangial cells following the progression of podocyte injury. Ectopic expression of CXCR4 in 5/6 nephrectomy mice increased the decline of renal function and glomerular injury, accelerated podocyte injury and mesangial cell activation, and initiated CXCR4-AT1 axis signals. Additionally, treatment with losartan, an AT1 blocker, interrupted the cycle of podocyte injury and mesangial matrix deposition triggered by CXCR4. Podocyte-specific ablation of CXCR4 gene blocked podocyte injury and mesangial cell activation. In vitro, CXCR4 overexpression induced oxidative stress and renin angiotensin system (RAS) activation in podocytes, and triggered the communication between podocytes and mesangial cells. In cultured mesangial cells, AngII treatment induced the expression of SDF-1α, which was secreted into the supernatant to further promote oxidative stress and cell injury in podocytes. Collectively, these results demonstrate that the CXCR4-AT1 axis plays a vital role in glomerular injury via mediating pathologic crosstalk between podocytes and mesangial cells. Our findings uncover a novel pathogenic mechanism by which the CXCR4-AT1 axis promotes glomerular injury.
Collapse
Affiliation(s)
- Qinyu Wu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Shan Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Dan Xu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Ping Meng
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Qiurong Chen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Xiaoxu Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Xiaolong Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Shuangqin Chen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Huiyun Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Wenting Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Yabing Xiong
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Jiemei Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Jinhua Miao
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Weiwei Shen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Xu Lin
- Department of Nephrology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Yunfang Zhang
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.
| |
Collapse
|
4
|
Boi R, Ebefors K, Nyström J. The role of the mesangium in glomerular function. Acta Physiol (Oxf) 2023; 239:e14045. [PMID: 37658606 DOI: 10.1111/apha.14045] [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: 05/30/2023] [Revised: 07/12/2023] [Accepted: 08/01/2023] [Indexed: 09/03/2023]
Abstract
When discussing glomerular function, one cell type is often left out, the mesangial cell (MC), probably since it is not a part of the filtration barrier per se. The MCs are instead found between the glomerular capillaries, embedded in their mesangial matrix. They are in direct contact with the endothelial cells and in close contact with the podocytes and together they form the glomerulus. The MCs can produce and react to a multitude of growth factors, cytokines, and other signaling molecules and are in the perfect position to be a central hub for crosstalk communication between the cells in the glomerulus. In certain glomerular diseases, for example, in diabetic kidney disease or IgA nephropathy, the MCs become activated resulting in mesangial expansion. The expansion is normally due to matrix expansion in combination with either proliferation or hypertrophy. With time, this expansion can lead to fibrosis and decreased glomerular function. In addition, signs of complement activation are often seen in biopsies from patients with glomerular disease affecting the mesangium. This review aims to give a better understanding of the MCs in health and disease and their role in glomerular crosstalk and inflammation.
Collapse
Affiliation(s)
- Roberto Boi
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Li JS, Chen X, Luo A, Chen D. TFRC-RNA interactions show the regulation of gene expression and alternative splicing associated with IgAN in human renal tubule mesangial cells. Front Genet 2023; 14:1176118. [PMID: 37547464 PMCID: PMC10397801 DOI: 10.3389/fgene.2023.1176118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction: IgA nephropathy (IgAN) is the most common primary glomerular disease (PGD) which could progress to renal failure and is characterized by aberrant IgA immune complex deposition. Transferrin receptor1 (TFRC), an IgA receptor, is a potential RNA binding protein (RBP) which regulates expression of genes positively associated with the cell cycle and proliferation and is involved in IgAN. Molecular mechanisms by which TFRC affects IgAN development remain unclear. Methods: In this study, TFRC was overexpressed in human renal tubular mesangial cells (HRMCs) and RNA-sequencing (RNA-seq) and improved RNA immunoprecipitation sequencing (iRIP-seq) were performed. The aim was to identify potential RNA targets of TFRC at transcriptional and alternative splicing (AS) levels. Results: TFRC-regulated AS genes were enriched in mRNA splicing and DNA repair, consistent with global changes due to TFRC overexpression (TFRC-OE). Expression of TFRC-regulated genes potentially associated with IgAN, including CENPH, FOXM1, KIFC1, TOP2A, FABP4, ID1, KIF20A, ATF3, H19, IRF7, and H1-2, and with AS, CYGB, MCM7 and HNRNPH1, were investigated by RT-qPCR and iRIP-seq data analyzed to identify TFRC-bound RNA targets. RCC1 and RPPH1 were found to be TFRC-bound RNA targets involved in cell proliferation. Discussion: In conclusion, molecular TFRC targets were identified in HRMCs and TFRC found to regulate gene transcription and AS. TFRC is considered to have potential as a clinical therapeutic target.
Collapse
Affiliation(s)
- Jian-Si Li
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Chen
- Heilongjiang Provincial Hospital Affiliated to Harbin Institute of Technology, Harbin, China
| | - Ailing Luo
- Center for Genome Analysis, Wuhan Ruixing Biotechnology Co., Ltd., Wuhan, China
| | - Dong Chen
- Center for Genome Analysis, Wuhan Ruixing Biotechnology Co., Ltd., Wuhan, China
| |
Collapse
|
6
|
Ochiai K, Mochida Y, Nagase T, Fukuhara H, Yamaguchi Y, Nagase M. Upregulation of Piezo2 in the mesangial, renin, and perivascular mesenchymal cells of the kidney of Dahl salt-sensitive hypertensive rats and its reversal by esaxerenone. Hypertens Res 2023; 46:1234-1246. [PMID: 36810623 DOI: 10.1038/s41440-023-01219-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/23/2023]
Abstract
The recent discovery of mechanosensitive ion channels has promoted mechanobiological research in the field of hypertension and nephrology. We previously reported Piezo2 expression in mouse mesangial and juxtaglomerular renin-producing cells, and its modulation by dehydration. This study aimed to investigate how Piezo2 expression is altered in hypertensive nephropathy. The effects of the nonsteroidal mineralocorticoid receptor blocker, esaxerenone, were also analyzed. Four-week-old Dahl salt-sensitive rats were randomly assigned to three groups: rats fed a 0.3% NaCl diet (DSN), rats fed a high 8% NaCl diet (DSH), and rats fed a high salt diet supplemented with esaxerenone (DSH + E). After six weeks, DSH rats developed hypertension, albuminuria, glomerular and vascular injuries, and perivascular fibrosis. Esaxerenone effectively decreased blood pressure and ameliorated renal damage. In DSN rats, Piezo2 was expressed in Pdgfrb-positive mesangial and Ren1-positive cells. Piezo2 expression in these cells was enhanced in DSH rats. Moreover, Piezo2-positive cells accumulated in the adventitial layer of intrarenal small arteries and arterioles in DSH rats. These cells were positive for Pdgfrb, Col1a1, and Col3a1, but negative for Acta2 (αSMA), indicating that they were perivascular mesenchymal cells different from myofibroblasts. Piezo2 upregulation was reversed by esaxerenone treatment. Furthermore, Piezo2 inhibition by siRNA in the cultured mesangial cells resulted in upregulation of Tgfb1 expression. Cyclic stretch also upregulated Tgfb1 in both transfections of control siRNA and Piezo2 siRNA. Our findings suggest that Piezo2 may have a contributory role in modulating the pathogenesis of hypertensive nephrosclerosis and have also highlighted the therapeutic effects of esaxerenone on salt-induced hypertensive nephropathy. Mechanochannel Piezo2 is known to be expressed in the mouse mesangial cells and juxtaglomerular renin-producing cells, and this was confirmed in normotensive Dahl-S rats. In salt-induced hypertensive Dahl-S rats, Piezo2 upregulation was observed in the mesangial cells, renin cells, and notably, perivascular mesenchymal cells, suggesting its involvement in kidney fibrosis.
Collapse
Affiliation(s)
- Koji Ochiai
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.,Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Yuki Mochida
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.,Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Takashi Nagase
- Kunitachi Aoyagien Tachikawa Geriatric Health Services Facility, Tachikawa, Tokyo, Japan
| | - Hiroshi Fukuhara
- Department of Urology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Yoshihiro Yamaguchi
- Department of Trauma and Critical Care Medicine, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Miki Nagase
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.
| |
Collapse
|
7
|
Thurman JM, Harrison RA. The susceptibility of the kidney to alternative pathway activation-A hypothesis. Immunol Rev 2023; 313:327-338. [PMID: 36369971 DOI: 10.1111/imr.13168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The glomerulus is often the prime target of dysregulated alternative pathway (AP) activation. In particular, AP activation is the key driver of two severe kidney diseases: atypical hemolytic uremic syndrome and C3 glomerulopathy. Both conditions are associated with a variety of predisposing molecular defects in AP regulation, such as genetic variants in complement regulators, autoantibodies targeting AP proteins, or autoantibodies that stabilize the AP convertases (C3- and C5-activating enzymes). It is noteworthy that these are systemic AP defects, yet in both diseases pathologic complement activation primarily affects the kidneys. In particular, AP activation is often limited to the glomerular capillaries. This tropism of AP-mediated inflammation for the glomerulus points to a unique interaction between AP proteins in plasma and this particular anatomic structure. In this review, we discuss the pre-clinical and clinical data linking the molecular causes of aberrant control of the AP with activation in the glomerulus, and the possible causes of this tropism. Based on these data, we propose a model for why the kidney is so uniquely and frequently targeted in patients with AP defects. Finally, we discuss possible strategies for preventing pathologic AP activation in the kidney.
Collapse
Affiliation(s)
- Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | | |
Collapse
|
8
|
Kriz W, Wiech T, Gröne HJ. Mesangial Injury and Capillary Ballooning Precede Podocyte Damage in Nephrosclerosis. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1670-1682. [PMID: 36150506 DOI: 10.1016/j.ajpath.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/22/2022] [Accepted: 08/30/2022] [Indexed: 06/16/2023]
Abstract
The development of focal and segmental glomerulosclerosis (FSGS) as a consequence of glomerular hypertension resulting from arterial hypertension is widely considered a podocyte disease. However, the primary damage is encountered in the mesangium. In acute settings, mesangial cells disconnect from their insertions to the glomerular basement membrane, causing a ballooning of capillaries and severe changes of the folding pattern of the glomerular basement membrane, of the arrangement of the capillaries, and thereby of the architecture of the tuft. The displacement of capillaries led to contact of podocytes and parietal epithelial cells, initiating the formation of tuft adhesions to Bowman's capsule, the committed lesion to progress to FSGS. In addition, the displacement of capillaries also caused an abnormal stretching of podocytes, resulting in podocyte damage. Thus, the podocyte damage that starts the sequence to FSGS is predicted to develop secondary to the mesangial damage. This sequence was found in two hypertensive rat models of FSGS and in human hypertensive nephrosclerosis.
Collapse
Affiliation(s)
- Wilhelm Kriz
- Department of Neuroanatomy, Medical Faculty Mannheim, University Heidelberg, Mannheim, Germany.
| | - Thorsten Wiech
- Nephropathology Section, Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hermann-Josef Gröne
- Medical Faculty, University of Heidelberg, Heidelberg, Germany; Institute of Pharmacology, University of Marburg, Marburg, Germany
| |
Collapse
|
9
|
Luvizotto MJ, Menezes-Silva L, Woronik V, Monteiro RC, Câmara NOS. Gut-kidney axis in IgA nephropathy: Role on mesangial cell metabolism and inflammation. Front Cell Dev Biol 2022; 10:993716. [PMID: 36467425 PMCID: PMC9715425 DOI: 10.3389/fcell.2022.993716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2023] Open
Abstract
IgA Nephropathy (IgAN) is the commonest primary glomerular disease around the world and represents a significant cause of end-stage renal disease. IgAN is characterized by mesangial deposition of IgA-immune complexes and mesangial expansion. The pathophysiological process includes an abnormally glycosylated IgA1, which is an antigenic target. Autoantibodies specifically recognize galactose-deficient IgA1 forming immune complexes that are amplified in size by the soluble IgA Fc receptor CD89 leading to deposition in the mesangium through interaction with non-classical IgA receptors. The local production of cytokines promotes local inflammation and complement system activation, besides the stimulation of mesangial proliferation. The spectrum of clinical manifestations is quite variable from asymptomatic microscopic hematuria to rapidly progressive glomerulonephritis. Despite all the advances, the pathophysiology of the disease is still not fully elucidated. The mucosal immune system is quoted to be a factor in triggering IgAN and a "gut-kidney axis" is proposed in its development. Furthermore, many recent studies have demonstrated that food intake interferes directly with disease prognosis. In this review, we will discuss how mucosal immunity, microbiota, and nutritional status could be interfering directly with the activation of intrinsic pathways of the mesangial cells, directly resulting in changes in their function, inflammation and development of IgAN.
Collapse
Affiliation(s)
- Mateus Justi Luvizotto
- Department of Nephrology, Faculty of Medicine, University of Sao Paulo, São Paulo, Brazil
| | - Luísa Menezes-Silva
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Viktoria Woronik
- Department of Nephrology, Faculty of Medicine, University of Sao Paulo, São Paulo, Brazil
| | - Renato C. Monteiro
- Centre de Recherche sur l’Inflammation, INSERM and CNRS, Université Paris Cité, Paris, France
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Goligorsky MS. Emerging Insights into Glomerular Vascular Pole and Microcirculation. J Am Soc Nephrol 2022; 33:1641-1648. [PMID: 35853715 PMCID: PMC9529196 DOI: 10.1681/asn.2022030354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/16/2022] [Accepted: 07/06/2022] [Indexed: 01/14/2023] Open
Abstract
The glomerular vascular pole is the gate for the afferent and efferent arterioles and mesangial cells and a frequent location of peripolar cells with an unclear function. It has been studied in definitive detail for >30 years, and functionally interrogated in the context of signal transduction from the macula densa to the mesangial cells and afferent arteriolar smooth muscle cells from 10 to 20 years ago. Two recent discoveries shed additional light on the vascular pole, with possibly far-reaching implications. One, which uses novel serial section electron microscopy, reveals a shorter capillary pathway between the basins of the afferent and efferent arterioles. Such a pathway, when patent, may short-circuit the multitude of capillaries in the glomerular tuft. Notably, this shorter capillary route is enclosed within the glomerular mesangium. The second study used anti-Thy1.1-induced mesangiolysis and intravital microscopy to unequivocally establish in vivo the long-suspected contractile function of mesangial cells, which have the ability to change the geometry and curvature of glomerular capillaries. These studies led me to hypothesize the existence of a glomerular perfusion rheostat, in which the shorter path periodically fluctuates between being more and less patent. This action reduces or increases blood flow through the entire glomerular capillary tuft. A corollary is that the GFR is a net product of balance between the states of capillary perfusion, and that deviations from the balanced state would increase or decrease GFR. Taken together, these studies may pave the way to a more profound understanding of glomerular microcirculation under basal conditions and in progression of glomerulopathies.
Collapse
Affiliation(s)
- Michael S. Goligorsky
- Renal Research Institute, New York Medical College at the Touro University, Valhalla, New York
| |
Collapse
|
11
|
Thomas HY, Ford Versypt AN. Pathophysiology of mesangial expansion in diabetic nephropathy: mesangial structure, glomerular biomechanics, and biochemical signaling and regulation. J Biol Eng 2022; 16:19. [PMID: 35918708 PMCID: PMC9347079 DOI: 10.1186/s13036-022-00299-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/23/2022] [Indexed: 02/08/2023] Open
Abstract
Diabetic nephropathy, a kidney complication arising from diabetes, is the leading cause of death in diabetic patients. Unabated, the growing epidemic of diabetes is increasing instances of diabetic nephropathy. Although the main causes of diabetic nephropathy have been determined, the mechanisms of their combined effects on cellular and tissue function are not fully established. One of many damages of diabetic nephropathy is the development of fibrosis within the kidneys, termed mesangial expansion. Mesangial expansion is an important structural lesion that is characterized by the aberrant proliferation of mesangial cells and excess production of matrix proteins. Mesangial expansion is involved in the progression of kidney failure in diabetic nephropathy, yet its causes and mechanism of impact on kidney function are not well defined. Here, we review the literature on the causes of mesangial expansion and its impacts on cell and tissue function. We highlight the gaps that still remain and the potential areas where bioengineering studies can bring insight to mesangial expansion in diabetic nephropathy.
Collapse
Affiliation(s)
- Haryana Y Thomas
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Ashlee N Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA. .,Institute for Computational and Data Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA.
| |
Collapse
|
12
|
Piezo2 expression and its alteration by mechanical forces in mouse mesangial cells and renin-producing cells. Sci Rep 2022; 12:4197. [PMID: 35273307 PMCID: PMC8913706 DOI: 10.1038/s41598-022-07987-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 03/01/2022] [Indexed: 11/10/2022] Open
Abstract
The kidney plays a central role in body fluid homeostasis. Cells in the glomeruli and juxtaglomerular apparatus sense mechanical forces and modulate glomerular filtration and renin release. However, details of mechanosensory systems in these cells are unclear. Piezo2 is a recently identified mechanically activated ion channel found in various tissues, especially sensory neurons. Herein, we examined Piezo2 expression and regulation in mouse kidneys. RNAscope in situ hybridization revealed that Piezo2 expression was highly localized in mesangial cells and juxtaglomerular renin-producing cells. Immunofluorescence assays detected GFP signals in mesangial cells and juxtaglomerular renin-producing cells of Piezo2GFP reporter mice. Piezo2 transcripts were observed in the Foxd1-positive stromal progenitor cells of the metanephric mesenchyme in the developing mouse kidney, which are precursors of mesangial cells and renin-producing cells. In a mouse model of dehydration, Piezo2 expression was downregulated in mesangial cells and upregulated in juxtaglomerular renin-producing cells, along with the overproduction of renin and enlargement of the area of renin-producing cells. Furthermore, the expression of the renin coding gene Ren1 was reduced by Piezo2 knockdown in cultured juxtaglomerular As4.1 cells under static and stretched conditions. These data suggest pivotal roles for Piezo2 in the regulation of glomerular filtration and body fluid balance.
Collapse
|
13
|
Ebefors K, Bergwall L, Nyström J. The Glomerulus According to the Mesangium. Front Med (Lausanne) 2022; 8:740527. [PMID: 35155460 PMCID: PMC8825785 DOI: 10.3389/fmed.2021.740527] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
The glomerulus is the functional unit for filtration of blood and formation of primary urine. This intricate structure is composed of the endothelium with its glycocalyx facing the blood, the glomerular basement membrane and the podocytes facing the urinary space of Bowman's capsule. The mesangial cells are the central hub connecting and supporting all these structures. The components as a unit ensure a high permselectivity hindering large plasma proteins from passing into the urine while readily filtering water and small solutes. There has been a long-standing interest and discussion regarding the functional contribution of the different cellular components but the mesangial cells have been somewhat overlooked in this context. The mesangium is situated in close proximity to all other cellular components of the glomerulus and should be considered important in pathophysiological events leading to glomerular disease. This review will highlight the role of the mesangium in both glomerular function and intra-glomerular crosstalk. It also aims to explain the role of the mesangium as a central component involved in disease onset and progression as well as signaling to maintain the functions of other glomerular cells to uphold permselectivity and glomerular health.
Collapse
Affiliation(s)
- Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lovisa Bergwall
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Wang H, Zhang R, Wu X, Chen Y, Ji W, Wang J, Zhang Y, Xia Y, Tang Y, Yuan J. The Wnt Signaling Pathway in Diabetic Nephropathy. Front Cell Dev Biol 2022; 9:701547. [PMID: 35059392 PMCID: PMC8763969 DOI: 10.3389/fcell.2021.701547] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetic nephropathy (DN) is a serious kidney-related complication of both type 1 and type 2 diabetes mellitus (T1DM, T2DM) and the second major cause of end-stage kidney disease. DN can lead to hypertension, edema, and proteinuria. In some cases, DN can even progress to kidney failure, a life-threatening condition. The precise etiology and pathogenesis of DN remain unknown, although multiple factors are believed to be involved. The main pathological manifestations of DN include mesangial expansion, thickening of the glomerular basement membrane, and podocyte injury. Eventually, these pathological manifestations will lead to glomerulosclerosis, thus affecting renal function. There is an urgent need to develop new strategies for the prevention and treatment of DN. Existing evidence shows that the Wnt signaling cascade plays a key role in regulating the development of DN. Previous studies focused on the role of the Wnt canonical signaling pathway in DN. Subsequently, accumulated evidence on the mechanism of the Wnt non-canonical signaling indicated that Wnt/Ca2+ and Wnt/PCP also have essential roles in the progression of DN. In this review, we summarize the specific mechanisms of Wnt signaling in the occurrence and development of DN in podocyte injury, mesangial cell injury, and renal fibrosis. Also, to elucidate the significance of the Wnt canonical pathway in the process of DN, we uncovered evidence supporting that both Wnt/PCP and Wnt/Ca2+ signaling are critical for DN development.
Collapse
Affiliation(s)
- Haiying Wang
- Department of Physiology, Jining Medical University, Jining, China
| | - Ran Zhang
- Basic Medical School, Jining Medical University, Jining, China
| | - Xinjie Wu
- Basic Medical School, Jining Medical University, Jining, China
| | - Yafen Chen
- Basic Medical School, Jining Medical University, Jining, China
| | - Wei Ji
- Basic Medical School, Jining Medical University, Jining, China
| | - Jingsuo Wang
- Basic Medical School, Jining Medical University, Jining, China
| | - Yawen Zhang
- Basic Medical School, Jining Medical University, Jining, China
| | - Yong Xia
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Yiqun Tang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jinxiang Yuan
- Collaborative Innovation Center, Jining Medical University, Jining, China
| |
Collapse
|
15
|
Müller-Deile J, Sopel N, Ohs A, Rose V, Gröner M, Wrede C, Hegermann J, Daniel C, Amann K, Zahner G, Schiffer M. Glomerular Endothelial Cell-Derived microRNA-192 Regulates Nephronectin Expression in Idiopathic Membranous Glomerulonephritis. J Am Soc Nephrol 2021; 32:2777-2794. [PMID: 34716242 PMCID: PMC8806098 DOI: 10.1681/asn.2020121699] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 07/09/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Autoantibodies binding to podocyte antigens cause idiopathic membranous glomerulonephritis (iMGN). However, it remains elusive how autoantibodies reach the subepithelial space because the glomerular filtration barrier (GFB) is size selective and almost impermeable for antibodies. METHODS Kidney biopsies from patients with iMGN, cell culture, zebrafish, and mouse models were used to investigate the role of nephronectin (NPNT) regulating microRNAs (miRs) for the GFB. RESULTS Glomerular endothelial cell (GEC)-derived miR-192-5p and podocyte-derived miR-378a-3p are upregulated in urine and glomeruli of patients with iMGN, whereas glomerular NPNT is reduced. Overexpression of miR-192-5p and morpholino-mediated npnt knockdown induced edema, proteinuria, and podocyte effacement similar to podocyte-derived miR-378a-3p in zebrafish. Structural changes of the glomerular basement membrane (GBM) with increased lucidity, splitting, and lamellation, especially of the lamina rara interna, similar to ultrastructural findings seen in advanced stages of iMGN, were found. IgG-size nanoparticles accumulated in lucidity areas of the lamina rara interna and lamina densa of the GBM in npnt-knockdown zebrafish models. Loss of slit diaphragm proteins and severe structural impairment of the GBM were further confirmed in podocyte-specific Npnt knockout mice. GECs downregulate podocyte NPNT by transfer of miR-192-5p-containing exosomes in a paracrine manner. CONCLUSIONS Podocyte NPNT is important for proper glomerular filter function and GBM structure and is regulated by GEC-derived miR-192-5p and podocyte-derived miR-378a-3p. We hypothesize that loss of NPNT in the GBM is an important part of the initial pathophysiology of iMGN and enables autoantigenicity of podocyte antigens and subepithelial immune complex deposition in iMGN.
Collapse
Affiliation(s)
- Janina Müller-Deile
- Department of Nephrology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nina Sopel
- Department of Nephrology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Alexandra Ohs
- Department of Nephrology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Victoria Rose
- Department of Nephrology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marwin Gröner
- Research Center On Rare Kidney Diseases (RECORD), University Hospital Erlangen, Erlangen, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Medizinische Hochschule Hannover, Hannover, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Gunther Zahner
- Department of Medicine, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Mario Schiffer
- Department of Nephrology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
16
|
Sánchez Fernández de la Vega J, Martínez-Onsurbe MP, Alonso Garcia S, Alba Losada J, Alonso Riaño M, Pardo Mindán FJ. [Reinterpretation of the Malpighian body in light of the existence of a single glomerular arteriole (Trabucco and Marquez)]. REVISTA ESPANOLA DE PATOLOGIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ANATOMIA PATOLOGICA Y DE LA SOCIEDAD ESPANOLA DE CITOLOGIA 2021; 54:220-233. [PMID: 34544552 DOI: 10.1016/j.patol.2021.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 06/13/2023]
Abstract
INTRODUCTION In 1842 William Bowman described the microvascular system of the Malpighian body. Electron microscopic studies definitively revealed the spatial structure of its mesangial-capillary-epithelial component. In 1952-54 Trabucco and Marquez challenged the ideas of Bowman, demonstrating the existence of a single glomerular arteriole. Our study supports the finding of a single glomerular arteriole, leading to a definitive interpretation of the Malpighian body structure. MATERIALS AND METHODS Serial histological studies were carried out of the vascular pole in a case of oligomeganephrotic renal hypoplasia and the immunohistochemical study of embryonal glomerular development (15 embryos aged between 7 and 11weeks), with alpha-actin (smooth muscle marker), CD31 and CD34 (endothelial markers) and CD10 (podocyte marker). RESULTS The study of the glomerular vascular pole in the case of oligomeganephrotic renal hypoplasia supports the existence of a single glomerular arteriole. Our immunohistochemical study confirmed this finding and provided data on the morphogenesis of the mesangial-capillary-epithelial component of the Malpighian body. CONCLUSIONS There exist a single glomerular arteriole. Mesangial and endothelial cells originating from a single glomerular arteriole interact with an epithelial component derived from the nephrogenic vesicle which then generate the lobular glomerular tuft, providing the basis for a definitive interpretation of the structure of the Malpighian body. There is no scientific base to the interpretation of the glomerular microvascular system as having two glomerular arterioles with an intercalated capillary network.
Collapse
Affiliation(s)
| | - M Pilar Martínez-Onsurbe
- Anatomía Patológica, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, España
| | | | | | | | | |
Collapse
|
17
|
Ando K, Shih YH, Ebarasi L, Grosse A, Portman D, Chiba A, Mattonet K, Gerri C, Stainier DYR, Mochizuki N, Fukuhara S, Betsholtz C, Lawson ND. Conserved and context-dependent roles for pdgfrb signaling during zebrafish vascular mural cell development. Dev Biol 2021; 479:11-22. [PMID: 34310924 DOI: 10.1016/j.ydbio.2021.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/17/2021] [Indexed: 12/27/2022]
Abstract
Platelet derived growth factor beta and its receptor, Pdgfrb, play essential roles in the development of vascular mural cells, including pericytes and vascular smooth muscle cells. To determine if this role was conserved in zebrafish, we analyzed pdgfb and pdgfrb mutant lines. Similar to mouse, pdgfb and pdgfrb mutant zebrafish lack brain pericytes and exhibit anatomically selective loss of vascular smooth muscle coverage. Despite these defects, pdgfrb mutant zebrafish did not otherwise exhibit circulatory defects at larval stages. However, beginning at juvenile stages, we observed severe cranial hemorrhage and vessel dilation associated with loss of pericytes and vascular smooth muscle cells in pdgfrb mutants. Similar to mouse, pdgfrb mutant zebrafish also displayed structural defects in the glomerulus, but normal development of hepatic stellate cells. We also noted defective mural cell investment on coronary vessels with concomitant defects in their development. Together, our studies support a conserved requirement for Pdgfrb signaling in mural cells. In addition, these zebrafish mutants provide an important model for definitive investigation of mural cells during early embryonic stages without confounding secondary effects from circulatory defects.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden; Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Sendagi Bunkyo-ku, Tokyo, 113 8602, Japan.
| | - Yu-Huan Shih
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Lwaki Ebarasi
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden
| | - Ann Grosse
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Daneal Portman
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 564 8565, Japan
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Claudia Gerri
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany; Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 564 8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Sendagi Bunkyo-ku, Tokyo, 113 8602, Japan
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden; Department of Medicine Huddinge (MedH), Karolinska Institutet, Campus Flemingsberg, Neo, Blickagången 16, Hiss S, Plan 7, SE-141 57, Huddinge, Sweden
| | - Nathan D Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States.
| |
Collapse
|
18
|
Ziegler V, Fremter K, Helmchen J, Witzgall R, Castrop H. Mesangial cells regulate the single nephron GFR and preserve the integrity of the glomerular filtration barrier: An intravital multiphoton microscopy study. Acta Physiol (Oxf) 2021; 231:e13592. [PMID: 33269519 DOI: 10.1111/apha.13592] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/25/2022]
Abstract
AIM The intraglomerular mesangial cells are located between the glomerular capillaries. Here we hypothesized that mesangial cells regulate the single nephron glomerular filtration rate (snGFR) and that mesangial cells support the integrity of the glomerular filtration barrier. METHODS We assessed the function of mesangial cells in vivo by multiphoton microscopy. Mesangial cells were depleted in Munich Wistar Froemter rats using the Thy1.1 antibody model. RESULTS The Thy1.1 antibody caused the cell-specific loss of 82 ± 3% of mesangial cells. After mesangial cell depletion, the baseline snGFR was reduced to 12.0 ± 1.2 vs 32.4 ± 3.2 nL/min in controls. In control rats, the snGFR decreased after angiotensin II infusion by 61 ± 3% (P = .004), whereas it remained unchanged in Thy1.1-treated rats. The changes in the snGFR after angiotensin II infusion in control rats were accompanied by the marked rotation of the capillary loops within Bowman's space. This phenomenon was absent in anti-Thy1.1-treated rats. The glomerular sieving coefficient (GSCA ) for albumin, used as a measure of the integrity of the glomerular filtration barrier, was low in control rats (0.00061 ± 0.00004) and increased after angiotensin II infusion (0.00121 ± 0.00015). In Thy1.1-treated rats, the GSC was elevated (0.0032 ± 0.00059) and did not change in response to angiotensin II. Electron microscopy revealed the increased thickness of the glomerular basement membrane after mesangial cell depletion. CONCLUSION Our data suggest that mesangial cells actively contribute to the regulation of the snGFR. Furthermore, mesangial cells are crucially involved in maintaining the integrity of the glomerular filtration barrier, in part by modulating the thickness of the glomerular basement membrane.
Collapse
Affiliation(s)
- Vera Ziegler
- Institute of Physiology University of Regensburg Regensburg Germany
| | | | - Julia Helmchen
- Institute of Physiology University of Regensburg Regensburg Germany
| | - Ralph Witzgall
- Institute of Molecular and Cellular Anatomy University of Regensburg Regensburg Germany
| | - Hayo Castrop
- Institute of Physiology University of Regensburg Regensburg Germany
| |
Collapse
|
19
|
Kawasaki Y, Hosoyamada Y, Miyaki T, Yamaguchi J, Kakuta S, Sakai T, Ichimura K. Three-Dimensional Architecture of Glomerular Endothelial Cells Revealed by FIB-SEM Tomography. Front Cell Dev Biol 2021; 9:653472. [PMID: 33777962 PMCID: PMC7991748 DOI: 10.3389/fcell.2021.653472] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/04/2021] [Indexed: 11/29/2022] Open
Abstract
Focused-ion beam-scanning electron microscopic (FIB-SEM) tomography enables easier acquisition of a series of ultrastructural, sectional images directly from resin-embedded biological samples. In this study, to clarify the three-dimensional (3D) architecture of glomerular endothelial cells (GEnCs) in adult rats, we manually extracted GEnCs from serial FIB-SEM images and reconstructed them on an Amira reconstruction software. The luminal and basal surface structures were clearly visualized in the reconstructed GEnCs, although only the luminal surface structures could be observed by conventional SEM. The luminal surface visualized via the reconstructed GEnCs was quite similar to that observed through conventional SEM, indicating that 3D reconstruction could be performed with high accuracy. Thus, we successfully described the 3D architecture of normal GEnCs in adult rats more clearly and precisely than ever before. The GEnCs were found to consist of three major subcellular compartments, namely, the cell body, cytoplasmic ridges, and sieve plates, in addition to two associated subcellular compartments, namely, the globular protrusions and reticular porous structures. Furthermore, most individual GEnCs made up a “seamless” tubular shape, and some of them formed an autocellular junction to make up a tubular shape. FIB-SEM tomography with reconstruction is a powerful approach to better understand the 3D architecture of GEnCs. Moreover, the morphological information revealed in this study will be valuable for the 3D pathologic evaluation of GEnCs in animal and human glomerular diseases and the structural analysis of developmental processes in the glomerular capillary system.
Collapse
Affiliation(s)
- Yuto Kawasaki
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasue Hosoyamada
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Nutrition, Faculty of Health Care Sciences, Chiba Prefectural University of Health Sciences, Chiba, Japan
| | - Takayuki Miyaki
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Junji Yamaguchi
- Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tatsuo Sakai
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Activated mesangial cells acquire the function of antigen presentation. Cell Immunol 2020; 361:104279. [PMID: 33422698 DOI: 10.1016/j.cellimm.2020.104279] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/20/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022]
Abstract
Mesangial cells (MCs), as resident cells of the kidneys, play an important role in maintaining glomerular function. MCs are located between the capillary loops of the glomeruli and mainly support the capillary plexus, constrict blood vessels, extracellular matrix components, produce cytokines, and perform phagocytosis and clearance of macromolecular substances. When the glomerular environment changes, MCs are often affected, which can lead to functional transformation. The immune response is involved in the occurrence and development of various kidney diseases, in these diseases, antigen-presenting cells (APCs) play an important role. APCs can present antigens to T lymphocytes, causing them to become activated and proliferate. Studies have shown that MCs have phagocytic function and express APC markers on the cell surface. Additionally, MCs are stimulated by or produce various inflammatory factors to participate in the renal inflammatory response. Therefore, MCs have potential antigen presentation function and participate in the pathological changes of various kidney diseases as APCs upon activation. In this paper, by reviewing MC phagocytic function, activated MC expression of APC surface markers, and MC participation in the inflammatory response and local renal immune response, we confirm that activated MCs can act as APCs in renal disease.
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Earlier works of the glomerulogenesis described morphological steps and protein expression during in-vivo and in-vitro kidney development. Recent technologies using cell-specific or conditional knock-out mice for several factors provide important knowledge about cross-talk signaling among resident cells as local events. Based on the recent advancement, this review revisits comprehensive morphological development of the glomerulus. RECENT FINDINGS Interactions of presumptive podocyte vascular endothelial growth factor with vascular endothelial growth factor-2 on angioblasts initiate glomerular vascularization. In induced pluripotent stem cells or organoid-derived nephron formation, the lack of endothelium and mesangial cells under differentiated podocytes suggests the presence of another unknown mechanism for glomerular neovascularization. Mesangial cell migration is prerequisite for glomerular looping by interaction of endothelial platelet-derived grothe factor beta and mesangial platelet-derived growth factor receptor beta and requires the coreceptor neuropilin1. Development of the filtration barrier is promoted by cross-talk among resident cells and may need shear stress. The components of the glomerular basement membrane change during glomerulogenesis, and endothelium and podocytes produce laminin and type IV collagen α1 and α2, whereas type IV collagen α3, α4, α5 is derived only from podocytes. SUMMARY Glomerulogenesis progresses by dynamic cellular migration/differentiation induced by cross-talk signaling in resident cells. Glomerular vasculogenesis and subsequent capillary development provide insight into glomerular regeneration and remodeling for medical application.
Collapse
|
22
|
Abstract
The main cellular constituents in glomerular mesangium are mesangial cells, which account for approximately 30-40% of the total cells in the glomerulus. Together with the mesangial matrix, mesangial cells form the glomerular basement membrane (GBM) in the glomerulus, whose main function is to perform the filtration. Under the pathologic conditions, mesangial cells are activated, leading to hyperproliferation and excess extracellular matrix (ECM). Moreover, mesangial cells also secrete several kinds of inflammatory cytokines, adhesion molecules, chemokines, and enzymes, all of which participate in the process of renal glomerular fibrosis. During the past years, researchers have revealed the roles of mesangial cells and the associated signal pathways involved in renal fibrosis. In this section, we will discuss how mesangial cells are activated and its contributions to renal fibrosis, as well as the molecular mechanisms and novel anti-fibrotic agents. Full understanding of the contributions of mesangial cells to renal fibrosis will benefit the clinical drug developing.
Collapse
Affiliation(s)
- Jing-Hong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
23
|
Affiliation(s)
- Wilhelm Kriz
- Department of Neuroanatomy, Medical Faculty Mannheim, University of Heidelberg, Germany
| |
Collapse
|
24
|
Zimmerman SE, Hiremath C, Tsunezumi J, Yang Z, Finney B, Marciano DK. Nephronectin Regulates Mesangial Cell Adhesion and Behavior in Glomeruli. J Am Soc Nephrol 2018; 29:1128-1140. [PMID: 29335243 DOI: 10.1681/asn.2017070752] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/13/2017] [Indexed: 01/03/2023] Open
Abstract
A critical aspect of kidney function occurs at the glomerulus, the capillary network that filters the blood. The glomerular basement membrane (GBM) is a key component of filtration, yet our understanding of GBM interactions with mesangial cells, specialized pericytes that provide structural stability to glomeruli, is limited. We investigated the role of nephronectin (Npnt), a GBM component and known ligand of α8β1 integrin. Immunolocalization and in situ hybridization studies in kidneys of adult mice revealed that nephronectin is produced by podocytes and deposited into the GBM. Conditional deletion of Npnt from nephron progenitors caused a pronounced increase in mesangial cell number and mesangial sclerosis. Nephronectin colocalized with α8β1 integrin to novel, specialized adhesion structures that occurred at sites of mesangial cell protrusion at the base of the capillary loops. Absence of nephronectin disrupted these adhesion structures, leading to mislocalization of α8β1. Podocyte-specific deletion of Npnt also led to mesangial sclerosis in mice. These results demonstrate a novel role for nephronectin and α8β1 integrin in a newly described adhesion complex and begin to uncover the molecular interactions between the GBM and mesangial cells, which govern mesangial cell behavior and may have a role in pathologic states.
Collapse
Affiliation(s)
- Susan E Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chitkale Hiremath
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jun Tsunezumi
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhufeng Yang
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bronwyn Finney
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Denise K Marciano
- Division of Nephrology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
25
|
From tubular sublimate nephropathy via urinary concentrating mechanism to glomerular disease—Wilhelm Kriz’s contribution to modern nephrology. Pflugers Arch 2017. [DOI: 10.1007/s00424-017-2010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
26
|
Restricted nutrition-induced low birth weight, low number of nephrons and glomerular mesangium injury in Japanese quail. J Dev Orig Health Dis 2017; 8:287-300. [PMID: 28162133 DOI: 10.1017/s2040174416000787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Insufficient nutrition during the perinatal period causes structural alterations in humans and experimental animals, leading to increased vulnerability to diseases in later life. Japanese quail, Coturnix japonica, in which partial (8-10%) egg white was withdrawn (EwW) from eggs before incubation had lower birth weights than controls (CTs). EwW birds also had reduced hatching rates, smaller glomeruli and lower embryo weight. In EwW embryos, the surface condensate area containing mesenchymal cells was larger, suggesting that delayed but active nephrogenesis takes place. In mature EwW quail, the number of glomeruli in the cortical region (mm2) was significantly lower (CT 34.7±1.4, EwW 21.0±1.2); capillary loops showed focal ballooning, and mesangial areas were distinctly expanded. Immunoreactive cell junction proteins, N-cadherin and podocin, and slit diaphragms were clearly seen. With aging, the mesangial area and glomerular size continued to increase and were significantly larger in EwW quail, suggesting compensatory hypertrophy. Furthermore, apoptosis measured by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling analysis was higher in EwWs than in CTs on embryonic day 15 and postnatal day 4 (D4). Similarly, plasma glucocorticoid (corticosterone) was higher (P<0.01) on D4 in EwW quail. These results suggest that although nephrogenic activity is high in low-nutrition quail during the perinatal period, delayed development and increased apoptosis may result in a lower number of mature nephrons. Damaged or incompletely mature mesangium may trigger glomerular injury, leading in later life to nephrosclerosis. The present study shows that birds serve as a model for 'fetal programming,' which appears to have evolved phylogenetically early.
Collapse
|
27
|
Clark SD, Nabity MB, Cianciolo RE, Dufek B, Cosgrove D. X-Linked Alport Dogs Demonstrate Mesangial Filopodial Invasion of the Capillary Tuft as an Early Event in Glomerular Damage. PLoS One 2016; 11:e0168343. [PMID: 27959966 PMCID: PMC5154607 DOI: 10.1371/journal.pone.0168343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/29/2016] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND X-linked Alport syndrome (XLAS), caused by mutations in the type IV collagen COL4A5 gene, accounts for approximately 80% of human Alport syndrome. Dogs with XLAS have a similar clinical progression. Prior studies in autosomal recessive Alport mice demonstrated early mesangial cell invasion as the source of laminin 211 in the glomerular basement membrane (GBM), leading to proinflammatory signaling. The objective of this study was to verify this process in XLAS dogs. METHODS XLAS dogs and WT littermates were monitored with serial clinicopathologic data and kidney biopsies. Biopsies were obtained at set milestones defined by the onset of microalbuminuria (MA), overt proteinuria, onset of azotemia, moderate azotemia, and euthanasia. Kidney biopsies were analyzed by histopathology, immunohistochemistry, and electron microscopy. RESULTS XLAS dogs showed progressive decrease in renal function and progressive increase in interstitial fibrosis and glomerulosclerosis (based on light microscopy and immunostaining for fibronectin). The only identifiable structural abnormality at the time of microalbuminuria was ultrastructural evidence of mild segmental GBM multilamination, which was more extensive when overt proteinuria developed. Co-localization studies showed that mesangial laminin 211 and integrin α8β1 accumulated in the GBM at the onset of overt proteinuria and coincided with ultrastructural evidence of mild cellular interpositioning, consistent with invasion of the capillary loops by mesangial cell processes. CONCLUSION In a large animal model, the induction of mesangial filopodial invasion of the glomerular capillary loop leading to the irregular deposition of laminin 211 is an early initiating event in Alport glomerular pathology.
Collapse
Affiliation(s)
- Sabrina D. Clark
- Department of Veterinary Pathobiology, Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, Texas, United States of America
- * E-mail:
| | - Mary B. Nabity
- Department of Veterinary Pathobiology, Texas A&M University College of Veterinary Medicine & Biomedical Sciences, College Station, Texas, United States of America
| | - Rachel E. Cianciolo
- Department of Veterinary Biosciences, The Ohio State University Columbus, Ohio, United States of America
| | - Brianna Dufek
- Center for Basic Research, Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Dominic Cosgrove
- Center for Basic Research, Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| |
Collapse
|
28
|
Takahara H, Shirato I, Asanuma K, Yamashita M, Takeda Y, Tomino Y. Tensin Is Expressed in Glomerular Mesangial Cells and Is Related to Their Attachment to Surrounding Extracellular Matrix. J Histochem Cytochem 2016; 52:683-91. [PMID: 15100245 DOI: 10.1177/002215540405200512] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Glomerular expression of tensin was immunohistochemically studied in normal and diseased rat kidneys to determine whether tensin might be related to specific binding in individual glomerular cells. Normal rat kidneys displayed an intense immunofluorescence reaction for tensin along the basal aspects of proximal and distal tubule cells and parietal epithelial cells of Bowman's capsules. In glomeruli, a positive reaction for tensin was detected only in the mesangial areas. Immunoelectron microscopy revealed a positive reaction in the mesangial cell (MC) processes. RT-PCR and immunoprecipitation demonstrated mRNA and protein levels of tensin in cultured rat MCs. Mesangial tensin expression was decreased when the mesangium was injured by Habu snake venom. During the regenerative process after mesangiolysis, tensin expression was not detected in early-phase proliferating MCs that did not have extracellular matrix (ECM). The expression of tensin recovered in late-phase proliferating MCs, which became attached to regenerated ECM. It appears that tensin is related to MC attachment to surrounding ECM, which suggests that signal transduction regulated by tensin may be related to a specific mechanism of MC matrix regeneration. Furthermore, tensin can act as a marker for rat MCs because the expression of tensin was detected only in MCs in glomeruli.
Collapse
Affiliation(s)
- Hisatsugu Takahara
- Division of Nephrology, Department of Internal Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Endothelin A receptor activation on mesangial cells initiates Alport glomerular disease. Kidney Int 2016; 90:300-310. [PMID: 27165837 DOI: 10.1016/j.kint.2016.02.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 01/21/2016] [Accepted: 02/11/2016] [Indexed: 01/19/2023]
Abstract
Recent work demonstrates that Alport glomerular disease is mediated through a biomechanical strain-sensitive activation of mesangial actin dynamics. This occurs through a Rac1/CDC42 cross-talk mechanism that results in the invasion of the subcapillary spaces by mesangial filopodia. The filopodia deposit mesangial matrix proteins in the glomerular basement membrane, including laminin 211, which activates focal adhesion kinase in podocytes culminating in the up-regulation of proinflammatory cytokines and metalloproteinases. These events drive the progression of glomerulonephritis. Here we test whether endothelial cell-derived endothelin-1 is up-regulated in Alport glomeruli and further elevated by hypertension. Treatment of cultured mesangial cells with endothelin-1 activates the formation of drebrin-positive actin microspikes. These microspikes do not form when cells are treated with the endothelin A receptor antagonist sitaxentan or under conditions of small, interfering RNA knockdown of endothelin A receptor mRNA. Treatment of Alport mice with sitaxentan results in delayed onset of proteinuria, normalized glomerular basement membrane morphology, inhibition of mesangial filopodial invasion of the glomerular capillaries, normalization of glomerular expression of metalloproteinases and proinflammatory cytokines, increased life span, and prevention of glomerulosclerosis and interstitial fibrosis. Thus endothelin A receptor activation on mesangial cells is a key event in initiation of Alport glomerular disease in this model.
Collapse
|
30
|
Cell biology of mesangial cells: the third cell that maintains the glomerular capillary. Anat Sci Int 2016; 92:173-186. [PMID: 26910209 DOI: 10.1007/s12565-016-0334-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/14/2016] [Indexed: 10/22/2022]
Abstract
The renal glomerulus consists of glomerular endothelial cells, podocytes, and mesangial cells, which cooperate with each other for glomerular filtration. We have produced monoclonal antibodies against glomerular cells in order to identify different types of glomerular cells. Among these antibodies, the E30 clone specifically recognizes the Thy1.1 molecule expressed on mesangial cells. An injection of this antibody into rats resulted in mesangial cell-specific injury within 15 min, and induced mesangial proliferative glomerulonephritis in a reproducible manner. We examined the role of mesangial cells in glomerular function using several experimental tools, including an E30-induced nephritis model, mesangial cell culture, and the deletion of specific genes. Herein, we describe the characterization of E30-induced nephritis, formation of the glomerular capillary network, mesangial matrix turnover, and intercellular signaling between glomerular cells. New molecules that are involved in a wide variety of mesangial cell functions are also introduced.
Collapse
|
31
|
Hunt SE, Dorfman KD, Segal Y, Barocas VH. A computational model of flow and species transport in the mesangium. Am J Physiol Renal Physiol 2016; 310:F222-9. [PMID: 26831339 DOI: 10.1152/ajprenal.00182.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/06/2015] [Indexed: 01/17/2023] Open
Abstract
A variety of macromolecules accumulate in the glomerular mesangium in many different diseases, but the physics of the transport of these molecules within the mesangial matrix has not been extensively studied. We present a computational model of convection and diffusion within the porous mesangial matrix and apply this model to the specific instance of immunoglobulin A (IgA) transport in IgA nephropathy. We examine the influence of physiological factors including glomerular basement membrane (GBM) thickness and mesangial matrix density on the total accumulation of IgA. Our results suggest that IgA accumulation can be understood by relating convection and diffusion, thus demonstrating the importance of intrinsic glomerular factors.
Collapse
Affiliation(s)
- Sarah E Hunt
- Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota;
| | - Kevin D Dorfman
- Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota
| | - Yoav Segal
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; and Minneapolis Veterans Affairs Health Care System, Minneapolis, Minnesota
| | - Victor H Barocas
- Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
32
|
Hosoyamada Y, Ichimura K, Sakai T. Organ Specificity and Functional Relevance of the Arterial Structure: A Comparative Study in the Kidney and the Skeletal Muscle with Electron Microscopy. J Vasc Res 2016; 52:265-72. [PMID: 26824773 DOI: 10.1159/000442978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/01/2015] [Indexed: 11/19/2022] Open
Abstract
The differences in circulation among various organs are well known, but the structural differences have only been poorly investigated. In the present study the wall structure of arteries was observed with electron microscopy in rat kidney and skeletal muscle. The wall thickness was almost equal in both organs at about 40 μm in luminal diameter, while it was relatively thin in the larger arteries and thick in the smaller arteries in the kidney compared with that in the skeletal muscle. The smooth muscle cells were regularly arranged in parallel in circular or slightly spiral orientation in the kidney, whereas those in the skeletal muscle were irregularly arranged in heterogeneous orientations. Extracellular matrices were more abundant in the arterial media in the skeletal muscle than in the kidney. The inner elastic lamina was continuous in the kidney, and arranged in longitudinal bundles in the skeletal muscle. The adventitial collagen fibers were abundant and dense in the skeletal muscle, and were scattered in small bundles in the fluid-filled spaces in the kidney. While the arteries in the skeletal muscle are under severe mechanical stress during muscle contraction and dilate dramatically during exercise, those in the kidney regulate glomerular pressure almost constantly.
Collapse
Affiliation(s)
- Yasue Hosoyamada
- Department of Nutrition, Faculty of Health Care Sciences, Chiba Prefectural University of Health Sciences, Chiba, Japan
| | | | | |
Collapse
|
33
|
Ichimura K, Sakai T. Evolutionary morphology of podocytes and primary urine-producing apparatus. Anat Sci Int 2015; 92:161-172. [PMID: 26627098 PMCID: PMC5315740 DOI: 10.1007/s12565-015-0317-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/14/2015] [Indexed: 01/27/2023]
Abstract
Excretory organs were acquired in the early phase of metazoan evolution, and they play a crucial role in the maintenance of homeostasis of body fluids. In general, these organs consist of two functional components, the primary-urine producing apparatus and the modulating tubule. This basic organization of the excretory organs is conserved among most metazoans. Herein, we present an overview of the morphological evolution of the primary urine-producing apparatus in metazoans and describe the acquisition of the renal glomerulus—a specialized primary urine-producing apparatus—in vertebrates. We also describe the advancement of the glomerular structure and function in higher vertebrates.
Collapse
Affiliation(s)
- Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Tatsuo Sakai
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
34
|
Takahashi-Iwanaga H. Three-dimensional microanatomy of the pericapillary mesangial tissues in the renal glomerulus: Comparative observations in four vertebrate classes. Biomed Res 2015; 36:331-41. [PMID: 26522150 DOI: 10.2220/biomedres.36.331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The renal glomeruli in lower vertebrates display mesangium-like cells and matrices interposed between the capillary endothelium and the basement membrane, while those in mammals reportedly lack such interpositions except in pathological conditions. By combined scanning and transmission electron microscopic observations, the pericapillary mesangial tissues were comparatively analyzed in four vertebrate classes: mammals (rats and rabbits), reptiles (green iguanas), amphibians (bullfrogs), and teleosts (carps). The observations discriminated three types of pericapillary interposition. The first, acellular interpositions, occurred universally, with mammalians displaying rudimental ones. This tissue type corresponded with extracellular matrices held in subendothelial grooves which were supported by fine endothelial projections anchored to the basement membrane. In lower vertebrates these grooves constituted an anastomosed system of subendothelial channels that communicated with the mesangial region, to favor cleaning of the glomerular filter. The second, compound type was specific to reptiles and amphibians, affecting the entire capillary circumference in the latter. In this tissue type, fine mesangial processes--which accompanied considerable amounts of fibrillar matrices--were loosely associated with the endothelial bases, indicating their possible nature as a kind of myofibroblast. Occurrence of the third, cellular interpositions was confined to small incidental loci in mammalian and teleost glomeruli. This tissue type was mostly occupied by thick processes or main bodies of the mesangial cells that tightly interlocked their short marginal microvilli with corresponding indentations on the endothelial bases.
Collapse
|
35
|
Effect of Supercoiling on the Mechanical and Permeability Properties of Model Collagen IV Networks. Ann Biomed Eng 2014; 43:1695-705. [PMID: 25408357 DOI: 10.1007/s10439-014-1187-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/11/2014] [Indexed: 11/27/2022]
Abstract
Collagen IV networks in the glomerular basement membrane (GBM) are essential for the maintenance and regulation of blood filtration in the kidneys. The GBM contains two different types of collagen IV networks: [α1(IV)]2α2(IV) and α3(IV)α4(IV)α5(IV), the latter of which has a higher number of supercoils (two or more collagens coiling around each other). To investigate the effects of supercoiling on the mechanical and permeability properties of collagen IV networks, we generated model collagen IV networks in the GBM and reconnected them to create different levels of supercoiling. We found that supercoiling greatly increases the stiffness of collagen IV networks but only minimally decreases the permeability. Also, doubling the amount of supercoils in a network had a bigger effect than doubling the stiffness of the supercoils. Our results suggest that the formation of supercoils is a specialized mechanism by the GBM that provides with a network stiff and strong enough to withstand the high hydrostatic pressures of filtration, yet porous enough that filtration is not hindered. Clinically, understanding the effects of supercoiling gives us insight into the mechanisms of GBM failure in some disease states where the normal collagen IV structure is disrupted.
Collapse
|
36
|
Lal MA, Andersson AC, Katayama K, Xiao Z, Nukui M, Hultenby K, Wernerson A, Tryggvason K. Rhophilin-1 is a key regulator of the podocyte cytoskeleton and is essential for glomerular filtration. J Am Soc Nephrol 2014; 26:647-62. [PMID: 25071083 DOI: 10.1681/asn.2013111195] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Rhophilin-1 is a Rho GTPase-interacting protein, the biologic function of which is largely unknown. Here, we identify and describe the functional role of Rhophilin-1 as a novel podocyte-specific protein of the kidney glomerulus. Rhophilin-1 knockout mice were phenotypically normal at birth but developed albuminuria at about 2 weeks of age. Kidneys from severely albuminuric mice revealed widespread podocyte foot process effacement, thickening of the glomerular basement membrane, and FSGS-like lesions. The absence of any overt changes in the expression of podocyte proteins at the onset of proteinuria suggested that the primary cause of podocyte abnormalities in Rhpn1-null mice was the result of cell-autonomous, Rhophilin-1-dependent signaling events. In culture, Rhophilin-1 was detected at the plasma membrane leading edge of primary podocytes, where it elicited remodeling of the actin cytoskeleton network. This effect of Rhophilin-1 on actin cytoskeleton organization associated with inhibitory effects on Rho-dependent phosphorylation of the myosin regulatory light chain and stress fiber formation. Conversely, phosphorylation of myosin regulatory light chain increased in podocyte foot processes of Rhpn1(-/-) mice, implicating altered actinomyosin contractility in foot process effacement and compromised filtration capacity. Targeted deletion of RhoA in podocytes of Rhophilin-1 knockout mice exacerbated the renal injury. Taken together, our results indicate that Rhophilin-1 is essential for the integrity of the glomerular filtration barrier and that this protein is a key determinant of podocyte cytoskeleton architecture.
Collapse
Affiliation(s)
- Mark A Lal
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | | | - Kan Katayama
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Ziejie Xiao
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Masatoshi Nukui
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics
| | - Kjell Hultenby
- Department of Laboratory Medicine, Division of Clinical Research Center, and
| | - Annika Wernerson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics,
| |
Collapse
|
37
|
Kriz W, Hähnel B, Hosser H, Rösener S, Waldherr R. Structural analysis of how podocytes detach from the glomerular basement membrane under hypertrophic stress. Front Endocrinol (Lausanne) 2014; 5:207. [PMID: 25566184 PMCID: PMC4264519 DOI: 10.3389/fendo.2014.00207] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/17/2014] [Indexed: 01/09/2023] Open
Abstract
Podocytes are lost by detachment from the GBM as viable cells; details are largely unknown. We studied this process in the rat after growth stimulation with FGF-2. Endothelial and mesangial cells responded by hyperplasia, podocytes underwent hypertrophy, but, in the long run, developed various changes that could either be interpreted showing progressing stages in detachment from the GBM or stages leading to a tighter attachment by foot process effacement (FPE). This occurred in microdomains within the same podocyte; thus, features of detachment and of reinforced attachment may simultaneously be found in the same podocyte. (1) Initially, hypertrophied podocytes underwent cell body attenuation and formed large pseudocysts, i.e., expansions of the subpodocyte space. (2) Podocytes entered the process of FPE starting with the retraction of foot processes (FPs) and the replacement of the slit diaphragm by occluding junctions, thereby sealing the filtration slits. Successful completion of this process led to broad attachments of podocyte cell bodies to the GBM. (3) Failure of sealing the slits led to gaps of varying width between retracting FPs facilitating the outflow of the filtrate from the GBM. (4) Since those gaps are frequently overarched by broadened primary processes, the drainage of the filtrate into the Bowman's space may be hindered leading to the formation of small pseudocysts associated with bare areas of GBM. (5) The merging of pseudocysts created a system of communicating chambers through which the filtrate has to pass to reach Bowman's space. Multiple flow resistances in series likely generated an expansile force on podocytes contributing to detachment. (6) Such a situation appears to proceed to complete disconnection generally of a group of podocytes owing to the junctional connections between them. (7) Since such groups of detaching podocytes generally make contact to parietal cells, they start the formation of tuft adhesions to Bowman's capsule.
Collapse
Affiliation(s)
- Wilhelm Kriz
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Institute of Neuroanatomy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- *Correspondence: Wilhelm Kriz, Zentrum für Biomedizin und Medizintechnik Mannheim, Institut für Neuroanatomie, Ludolf-Krehl-Str. 13-17, Tridomus C, Ebene 6, Mannheim D68167, Germany e-mail:
| | - Brunhilde Hähnel
- Institute of Neuroanatomy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hiltraud Hosser
- Institute of Neuroanatomy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Pathologie Heidelberg, Heidelberg, Germany
| | - Sigrid Rösener
- Global Non-Clinical Safety, Merck KGaA, Merck Serono, Darmstadt, Germany
| | | |
Collapse
|
38
|
Are the precapillary sphincters and metarterioles universal components of the microcirculation? An historical review. J Physiol Sci 2013; 63:319-31. [PMID: 23824465 PMCID: PMC3751330 DOI: 10.1007/s12576-013-0274-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 06/03/2013] [Indexed: 12/17/2022]
Abstract
The microcirculation is a major topic in current physiology textbooks and is frequently explained with schematics including the precapillary sphincters and metarterioles. We re-evaluated the validity and applicability of the concepts precapillary sphincters and metarterioles by reviewing the historical context in which they were developed in physiology textbooks. The studies by Zweifach up until the 1950s revealed the unique features of the mesenteric microcirculation, illustrated with impressive schematics of the microcirculation with metarterioles and precapillary sphincters. Fulton, Guyton and other authors introduced or mimicked these schematics in their physiology textbooks as representative of the microcirculation in general. However, morphological and physiological studies have revealed that the microcirculation in the other organs and tissues contains no metarterioles or precapillary sphincters. The metarterioles and precapillary sphincters were not universal components of the microcirculation in general, but unique features of the mesenteric microcirculation.
Collapse
|
39
|
Gyoneva L, Segal Y, Dorfman KD, Barocas VH. Mechanical response of wild-type and Alport murine lens capsules during osmotic swelling. Exp Eye Res 2013; 113:87-91. [PMID: 23707242 DOI: 10.1016/j.exer.2013.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 05/06/2013] [Accepted: 05/13/2013] [Indexed: 01/05/2023]
Abstract
The mechanical support of basement membranes, such as the lens capsule, is believed to arise from one of their main constituents - collagen IV. The basement membranes of the lens, kidney, and ear normally contain two different types of collagen IV networks, referred to as the major and minor chain networks. In Alport syndrome, a mutation in one of the minor chain COL4 genes leads to the absence of the minor chain network, causing life-threatening disturbances. We hypothesized that the absence of the minor chain network increases basement membrane distensibility, as measured in wild-type (n = 25) and Alport syndrome (n = 21) mice using the lens capsule as a model. Osmotic swelling experiments revealed direction-dependent changes. As a reflection of lens capsule properties, Alport lenses strained significantly more than wild-type lenses in the anterior-posterior direction, i.e. along their thickness, but not in the equatorial direction (p = 0.03 and p = 0.08, respectively). This is consistent with clinical data: Alport patients develop conical protrusions on the anterior and posterior lenticular poles. There was no evidence of significant change in total amount of collagen between Alport and wild-type lenses (p = 0.6). The observed differences in distensibility could indicate that the major chain network alone cannot fully compensate for the absence of the more highly cross-linked minor chain network, which is believed to be stronger, more stable, and resistant to deformation. The addition of mechanical information on Alport syndrome to the currently available biological data provides a fuller picture into the progression of the disease.
Collapse
Affiliation(s)
- Lazarina Gyoneva
- Department of Biomedical Engineering, University of Minnesota, 7-105 Nils Hasselmo Hall, 312 Church St. SE, Minneapolis, MN 55455, United States.
| | | | | | | |
Collapse
|
40
|
Structural arrangement of collagen fibrils in the periarterial connective tissue of the kidney: their functional relevance as a structural stabilizer against arterial pressure. Anat Sci Int 2011; 87:80-7. [PMID: 22187217 PMCID: PMC3360868 DOI: 10.1007/s12565-011-0123-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 11/16/2011] [Indexed: 11/09/2022]
Abstract
Periarterial connective tissue with a moderate amount of collagen fibrils is known to be a specialized domain in the renal interstitium. This study aimed to clarify the microscopic architecture of the periarterial connective tissue as a mechanical supportive structure of the intrarenal arteries. Transmission and scanning electron microscopy revealed two populations of collagen fibrils in the periarterial connective tissue. The major one was composed of many bundles of collagen fibrils running in longitudinal directions, whereas the minor one was represented by a few circumferential bundles adjacent to the smooth muscles. The amount of collagen fibrils was obviously variable and correlated with the arterial caliber. The correlation between abundance of collagen fibrils and the arterial caliber was confirmed by morphometric analysis of the collagen fibril area per arterial perimeter on electron micrographs. The size of individual collagen fibrils was measured in periarterial connective tissue of arteries with various calibers. A positive correlation between the diameter of collagen fibrils and arterial caliber was confirmed, indicating the supportive function of collagen fibrils in the periarterial connective tissue. The accumulated morphological findings supported the hypothesis that the collagen fibrils in the periarterial connective tissue develop longitudinal tension with their tensile strength, whereas the smooth muscle cells in the media develop circumferential tension with active regulation of contracting force.
Collapse
|
41
|
Ichimura K, Kurihara H, Sakai T. Beta-cytoplasmic actin localization in vertebrate glomerular podocytes. ACTA ACUST UNITED AC 2010; 72:165-74. [PMID: 20513979 DOI: 10.1679/aohc.72.165] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The unique cytoarchitecture of glomerular podocytes is conserved in vertebrate evolution. Actin filaments play a crucial role in the formation of the conserved cytoarchitecture, though several isoforms of cytoplasmic actin have been found in vertebrates. The present study examined the expression and subcellular distribution of the beta-cytoplasmic actin (beta-actin) isoform in the podocytes of six vertebrate species by means of immunohistochemical techniques to reveal whether the beta-actin isoform is involved in the formation of podocyte cytoarchitecture throughout vertebrates. beta-actin was predominantly localized at the foot processes in carp, turtle, quail, and rat podocytes in addition to actin filament condensations, which were found only in carp and rat podocytes. The actin filament condensations in rats were in direct contact with the basal plasma membrane, but those in carp were found at the cell body and separated from the basal plasma membrane. In contrast with the above four species, beta-actin was not detected in podocytes in two amphibians-newt and frog, although podocyte foot processes are actin-filament based cytoplasmic protrusions in these species as well as in other vertebrates. In conclusion, the beta-actin isoform is involved in the formation of the podocyte actin cytoskeleton in vertebrates except for amphibians. Several kinds of unconventional cytoplasmic actins other than beta- and gamma-cytoplasmic actins are known to be expressed in amphibians, making it highly likely that one of these isoforms, instead of beta-actin, constructs actin filaments in the foot processes of newt and frog podocytes.
Collapse
Affiliation(s)
- Koichiro Ichimura
- Department of Anatomy, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan.
| | | | | |
Collapse
|
42
|
Wang Y, Heilig K, Saunders T, Minto A, Deb DK, Chang A, Brosius F, Monteiro C, Heilig CW. Transgenic overexpression of GLUT1 in mouse glomeruli produces renal disease resembling diabetic glomerulosclerosis. Am J Physiol Renal Physiol 2010; 299:F99-F111. [PMID: 20375117 DOI: 10.1152/ajprenal.00466.2009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous work identified an important role for hyperglycemia in diabetic nephropathy (The Diabetes Control and Complications Trial Research Group. N Engl J Med 329: 977-986, 1993; UK Prospective Diabetes Study Group. Lancet 352: 837-853, 1998), and increased glomerular GLUT1 has been implicated. However, the roles of GLUT1 and intracellular glucose have not been determined. Here, we developed transgenic GLUT1-overexpressing mice (GT1S) to characterize the roles of GLUT1 and intracellular glucose in the development of glomerular disease without diabetes. GLUT1 was overexpressed in glomerular mesangial cells (MC) of C57BL6 mice, a line relatively resistant to diabetic nephropathy. Blood pressure, blood glucose, glomerular morphometry, matrix proteins, cell signaling, transcription factors, and selected growth factors were examined. Kidneys of GT1S mice overexpressed GLUT1 in glomerular MCs and small vessels, rather than renal tubules. GT1S mice were neither diabetic nor hypertensive. Glomerular GLUT1, glucose uptake, mean capillary diameter, and mean glomerular volume were all increased in the GT1S mice. Moderately severe glomerulosclerosis (GS) was established by 26 wk of age in GT1S mice, with increased glomerular type IV collagen and fibronectin. Modest increases in glomerular basement membrane thickness and albuminuria were detected with podocyte foot processes largely preserved, in the absence of podocyte GLUT1 overexpression. Activation of glomerular PKC, along with increased transforming growth factor-beta1, VEGFR1, VEGFR2, and VEGF were all detected in glomeruli of GT1S mice, likely contributing to GS. The transcription factor NF-kappaB was also activated. Overexpression of glomerular GLUT1, mimicking the diabetic GLUT1 response, produced numerous features typical of diabetic glomerular disease, without diabetes or hypertension. This suggested GLUT1 may play an important role in the development of diabetic GS.
Collapse
Affiliation(s)
- Youli Wang
- Department of Medicine and Pathology, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hosoyamada Y, Ichimura K, Koizumi K, Sakai T. Structural organization of pulmonary veins in the rat lung, with special emphasis on the musculature consisting of cardiac and smooth muscles. Anat Sci Int 2010; 85:152-9. [PMID: 20082232 DOI: 10.1007/s12565-009-0071-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 12/07/2009] [Indexed: 10/20/2022]
Abstract
Recent physiological studies have indicated the significant role of pulmonary veins in the total resistance of pulmonary vasculature. The structure of pulmonary veins in the rat was reinvestigated to clarify the different venous segments and their ultrastructure with regard to the musculature including cardiac muscles and smooth muscles with light and electron microscopy. The cardiac muscles were located in the axial and the primary branches of the pulmonary veins within a certain distance limit from the hilum (CM segment) and not in the peripheral region (non-CM segment). The smooth muscles were found indifferent to the presence of cardiac muscles as a continuous layer in segments larger than 180 microm (continuous SM segment) or as a discontinuous layer of circular smooth muscle cells in segments between 50 and 180 microm (partial SM segment). The smooth muscle layer was extremely thin in the CM segments, whereas it became conspicuously thick in the non-CM segment with an irregularly undulating luminal outline, especially in the partial SM segments. There were two elastic laminae in the CM segments: a conspicuous one on the interstitial side of the smooth muscles, and a weaker one between the endothelium and smooth muscles. In the non-CM segment, one elastic lamina was found on the interstitial side of the smooth muscles. Considering the limited range of contraction of cardiac muscles and the thinness of smooth muscle cells in the CM segments, it was concluded that vasoconstriction in the pulmonary veins is executed by smooth muscle cells in the non-CM segments thicker than 50 microm.
Collapse
Affiliation(s)
- Yasue Hosoyamada
- Department of Nutrition, Chiba Prefectural University of Health Sciences, Japan
| | | | | | | |
Collapse
|
44
|
Nephron-deficient Fvb mice develop rapidly progressive renal failure and heavy albuminuria involving excess glomerular GLUT1 and VEGF. J Transl Med 2010; 90:83-97. [PMID: 19918242 PMCID: PMC4150870 DOI: 10.1038/labinvest.2009.95] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Reduced nephron numbers may predispose to renal failure. We hypothesized that glucose transporters (GLUTs) may contribute to progression of the renal disease, as GLUTs have been implicated in diabetic glomerulosclerosis and hypertensive renal disease with mesangial cell (MC) stretch. The Os (oligosyndactyly) allele that typically reduces nephron number by approximately 50%, was repeatedly backcrossed from ROP (Ra/+ (ragged), Os/+ (oligosyndactyly), and Pt/+ (pintail)) Os/+ mice more than six times into the Fvb mouse background to obtain Os/+ and +/+ mice with the Fvb background for study. Glomerular function, GLUT1, signaling, albumin excretion, and structural and ultrastructural changes were assessed. The FvbROP Os/+ mice (Fvb background) exhibited increased glomerular GLUT1, glucose uptake, VEGF, glomerular hypertrophy, hyperfiltration, extensive podocyte foot process effacement, marked albuminuria, severe extracellular matrix (ECM) protein deposition, and rapidly progressive renal failure leading to their early demise. Glomerular GLUT1 was increased 2.7-fold in the FvbROP Os/+ mice vs controls at 4 weeks of age, and glucose uptake was increased 2.7-fold. These changes were associated with the activation of glomerular PKCbeta1 and NF-kappaB p50 which contribute to ECM accumulation. The cyclic mechanical stretch of MCs in vitro, used as a model for increased MC stretch in vivo, reproduced increased GLUT1 at 48 h, a stimulus for increased VEGF expression which followed at 72 h. VEGF was also shown to act in a positive feedback manner on MC GLUT1, increasing GLUT1 expression, glucose uptake and fibronectin (FN) accumulation in vitro, whereas antisense suppression of GLUT1 largely blocked FN upregulation by VEGF. The FvbROP Os/+ mice exhibited an early increase in glomerular GLUT1 leading to increased glomerular glucose uptake PKCbeta1, and NF-kappaB activation, with excess ECM accumulation. A GLUT1-VEGF-GLUT1 positive feedback loop may play a key role in contributing to renal disease in this model of nondiabetic glomerulosclerosis.
Collapse
|
45
|
Ichimura K, Kurihara H, Sakai T. Actin filament organization of foot processes in vertebrate glomerular podocytes. Cell Tissue Res 2007; 329:541-57. [PMID: 17605050 DOI: 10.1007/s00441-007-0440-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2006] [Accepted: 05/16/2007] [Indexed: 11/30/2022]
Abstract
We investigated the actin filament organization and immunolocalization of actin-binding proteins (alpha-actinin and cortactin) in the podocyte foot processes of eight vertebrate species (lamprey, carp, newt, frog, gecko, turtle, quail, and rat). Three types of actin cytoskeleton were found in these foot processes. (1) A cortical actin network with cortactin filling the space between the plasma membrane and the other actin cytoskeletons described below was found in all of the species examined here. The data indicated that the cortical actin network was the minimal essential actin cytoskeleton for the formation and maintenance of the foot processes in vertebrate podocytes. (2) An actin bundle with alpha-actinin existing along the longitudinal axis of foot process above the level of slit diaphragms was only observed in quail and rat. (3) An actin fascicle consisting of much fewer numbers of actin filaments than that of the actin bundle was observed in the species other than quail and rat, but at various frequencies. These findings suggest that the actin bundle is an additional actin cytoskeleton reflecting a functional state peculiar to quail and rat glomeruli. Considering the higher intraglomerular pressure and the extremely thin filtration barrier in birds and mammals, the foot processes probably mainly protect the thinner filtration barrier from the higher internal pressure occurring in quail and rat glomeruli. Therefore, we consider that the actin bundle plays a crucial role in the mechanical protection of the filtration barrier. Moreover, the actin fascicle may be a potential precursor of the actin bundle.
Collapse
Affiliation(s)
- Koichiro Ichimura
- Department of Anatomy, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | | | | |
Collapse
|
46
|
Hosoyamada Y, Sakai T. Mechanical components of rat intestinal villi as revealed by ultrastructural analysis with special reference to the axial smooth muscle cells in the villi. ACTA ACUST UNITED AC 2007; 70:107-16. [PMID: 17827668 DOI: 10.1679/aohc.70.107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The ultrastructure of the rat intestinal interstitium with regard to the mechanical components was analyzed from a functional viewpoint utilizing serial horizontal as well as longitudinal sections through the lamina propria mucosae, including both villi and crypts. The axial smooth muscle cells in the villi (villus-axial SMs) exhibited different configurations at various levels of the wall. They were separated from the voluminous fluid-filled spaces by sheet-like processes of fibroblasts in the upper part of the intravillous interstitium, formed a sheet around the central lymphatics, and were covered by the sheet-like processes of fibroblasts in the lower part of the intravillous interstitium. These villus-axial SMs were poorly developed and associated with the lymphatic walls in the upper part of the pericryptal interstitium; they were tapered and connected to microtendons composed of fascicles of longitudinal collagen fibrils in the lower part of pericryptal interstitium. At the apical termination, the villus-axial SMs were connected to myofibloblasts, which sent off many processes into the subepithelial meshwork layer of fine cell processes and extracellular matrices. The villus-axial SMs possibly develop longitudinal tension against the intravillous hydraulic pressure developing from the transepithelial absorption through the intestinal epithelium.
Collapse
|
47
|
Ichimura K, Kurihara H, Sakai T. Involvement of mesangial cells expressing alpha-smooth muscle actin during restorative glomerular remodeling in Thy-1.1 nephritis. J Histochem Cytochem 2006; 54:1291-301. [PMID: 16924122 DOI: 10.1369/jhc.6a7000.2006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The function of actin cytoskeleton in mesangial cells (MCs) during the recovering process of injured glomeruli is not fully understood. MCs in injured glomeruli express alpha-smooth muscle actin (alpha-SMA), which is not detected in normal glomeruli. We focused on the localization of alpha-SMA in MCs of Thy-1.1 nephritic rat. Expression of alpha-SMA in the injured glomeruli peaked at day 5 after antibody injection and then declined gradually. At day 5, MCs, where alpha-SMA was localized at their cytoplasmic processes situated in various positions, occupied the expanded mesangium. MCs expressing alpha-SMA tended to be located at the peripheral region close to the glomerular basement membrane (GBM) or endothelial cells at day 8. Localization of alpha-SMA within the peripheral MCs was restricted to the cytoplasmic processes radiating toward the GBM and touching it with their tips at day 8. These alpha-SMA-containing processes are suitable to transmit the contractile force to GBM and may contribute to normalize the expanded glomerular volume. In addition, an actin-binding protein, drebrin, was localized in all MC processes extending toward various directions throughout the course of nephritis, suggesting that drebrin is involved in the formation of MC processes.
Collapse
Affiliation(s)
- Koichiro Ichimura
- Department of Anatomy, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | | | | |
Collapse
|
48
|
Hosoyamada Y, Sakai T. Structural and mechanical architecture of the intestinal villi and crypts in the rat intestine: integrative reevaluation from ultrastructural analysis. ACTA ACUST UNITED AC 2005; 210:1-12. [PMID: 16044319 DOI: 10.1007/s00429-005-0011-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2005] [Indexed: 10/25/2022]
Abstract
The ultrastructure of the rat intestinal interstitium was analyzed from the viewpoint of mechanical dynamics to stabilize the intestinal villi, crypts and mucosal folds. In the rat, the small intestine lacks circular folds, but the large intestine possesses spiral folds. The intestinal villi, the largest in the duodenum, decreased in size in the jejunum and ileum successively, and were absent in the large intestine. The intestinal interstitium consisted of lamina propria mucosae (LPM) and tela submucosa (TSM) separated by muscularis mucosae (MM), the LPM was subdivided into an upper part within the villi and a lower part among the crypts in the small intestine. The light microscopic density of interstitium in the intestinal wall was lowest in the upper LPM, moderately dense in the lower LPM and highest in the TSM, and that among the intestinal region was highest in the duodenum and decreased successively in the jejunum and ileum. In the large intestine, the TSM bulged to form spiral folds with very low density. The intestinal epithelium in the villi possessed wide intercellular spaces and that in the crypts had closed intercellular spaces. At electron microscopic level, the upper and lower LPM contained subepithelial supportive meshwork that consisted of collagen fibrils and myofibroblast processes. The lower LPM and TSM contained conspicuous bundles of collagen fibrils and, in addition, TSM contained minor populations of scattered collagen fibrils near the smooth muscle layer (SML). The diameter of collagen fibrils was the largest in the bundles of TSM, and decreased from the duodenum through the jejunum and ileum to the large intestine. On the basis of these observations, we hypothesize that the intestinal villi are mechanically stabilized by the balance between the expansive interstitial pressure and inward pull by the subepithelial supportive meshwork. This hypothesis explains the hitherto neglected fact that the intestinal epithelium possesses wide intercellular spaces only in the villi, and accounts for the counterforce against the perpendicular smooth muscle cells, which are supposed to contract the intestinal villi.
Collapse
Affiliation(s)
- Yasue Hosoyamada
- Department of Nutrition, Chiba College of Health Science, 2-10-1 Wakaba, Mihama-ku, Chiba-shi, Chiba, 261-0014, Japan
| | | |
Collapse
|
49
|
Tiedemann K, Sasaki T, Gustafsson E, Göhring W, Bätge B, Notbohm H, Timpl R, Wedel T, Schlötzer-Schrehardt U, Reinhardt DP. Microfibrils at basement membrane zones interact with perlecan via fibrillin-1. J Biol Chem 2005; 280:11404-12. [PMID: 15657057 DOI: 10.1074/jbc.m409882200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutational defects in fibrillin-rich microfibrils give rise to a number of heritable connective tissue disorders, generally termed microfibrillopathies. To understand the pathogenesis of these microfibrillopathies, it is important to elucidate the supramolecular composition of microfibrils and their interaction properties with extracellular matrix components. Here we demonstrate that the proteoglycan perlecan is an associated component of microfibrils typically close to basement membrane zones. Double immunofluorescence studies demonstrate colocalization of fibrillin-1, the major backbone component of microfibrils, with perlecan in fibroblast cultures as well as in dermal and ocular tissues. Double immunogold labeling further confirms colocalization of perlecan to microfibrils in various tissues at the ultrastructural level. Extraction studies revealed that perlecan is not covalently associated with microfibrils. High affinity interactions between fibrillin-1 and perlecan were found by kinetic binding studies with dissociation constants in the low nanomolar range. A detailed mapping study of the interaction epitopes by solid phase binding assays primarily revealed interactions of perlecan domains I and II with a central region of fibrillin-1. Analysis of perlecan null embryos showed less microfibrils at the dermal-epidermal junction as compared with wild-type littermates. The data presented indicate a functional significance for perlecan in anchoring microfibrils to basement membranes and in the biogenesis of microfibrils.
Collapse
Affiliation(s)
- Kerstin Tiedemann
- Department of Medical Molecular Biology, University of Lübeck, 23538 Lübeck, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kobayashi N, Gao SY, Chen J, Saito K, Miyawaki K, Li CY, Pan L, Saito S, Terashita T, Matsuda S. Process formation of the renal glomerular podocyte: is there common molecular machinery for processes of podocytes and neurons? Anat Sci Int 2004; 79:1-10. [PMID: 15088787 DOI: 10.1111/j.1447-073x.2004.00066.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The renal glomerular podocyte exhibits a highly arborized morphology. In comparison with the neuron, which is the best studied process-bearing cell, the podocyte major processes share many cell biological characteristics with neuronal dendrites. Both podocytes and neurons develop microtubule-based thick processes with branching morphology and both have thin actin-based projections (i.e. podocyte foot processes and dendritic spines). Formation of podocyte processes and neuronal dendrites depends on the assembly of microtubules. Because the assembly of microtubules is regulated by phosphorylation of microtubule-associated proteins, inhibition of protein phosphatases abolishes and inhibition of protein kinases promotes process formation. Podocytes and dendrites also share the machinery of intracellular traffic of membranous vesicles, as well as cytoskeletal elements, which is indispensable for the elongation of these processes. Furthermore, these two cell types share expression of various molecules working for signal transduction, transmembranous transport and intercellular contacts. Such common gene expression implies a similar transcriptional regulation in these cells. Concerning the formation of podocyte foot processes and dendritic branches, actin filaments are thought to play a central role in orchestrating the function of various molecules and the regulation of actin assembly is necessary to establish and maintain such sophisticated cellular architecture. The molecular mechanism of foot process formation seems to include Rho family small GTP-binding proteins, which are known to be responsible for the establishment of dendritic branching morphology.
Collapse
Affiliation(s)
- Naoto Kobayashi
- Division of Anatomy and Embryology, Department of Integrated Basic Medical Science, School of Medicine, University of Ehime, Ehime, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|