1
|
Priante G, Ceol M, Gianesello L, Radu CM, Mantese R, Stefanelli LF, Cacciapuoti M, Martino FK, Calò LA, Anglani F, Nalesso F, Del Prete D. Human parietal epithelial cells as Trojan horses in albumin overload. Sci Rep 2025; 15:1761. [PMID: 39800742 PMCID: PMC11725586 DOI: 10.1038/s41598-024-84972-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Parietal Epithelial Cells (PECs) activation and proliferation are common to several distinct forms of glomerulopathies. Due to several stimuli, PECs can change to a progenitor (CD24+ and CD133/2+) or a pro-sclerotic (CD44+) phenotype. In addition, PECs, which are constantly exposed to filtered albumin, are known to be involved in albumin internalization, but how this mechanism occurs is unknown. We hypothesized that PECs can transport albumin via receptor-mediated endocytosis and that albumin overload may affect the state of PECs. Conditionally immortalized human PECs (hPECs) were incubated with different albumin concentrations at different times. Albumin internalization studies were performed. Protein expression was assessed using In-Cell Western and immunofluorescence. Cell morphology was analyzed by phase-contrast microscopy and F-actin staining. We demonstrate that hPECs internalize albumin via receptor-mediated mechanisms. Under albumin stimulation, megalin, cubilin, ClC-5, CD133/2, CD24, and CD44 were upregulated. The increase of pERK1/2, the upregulation of ROCK1, ROCK2, caspase -3, -6, and -7, and the morphological changes associated with loss of F-actin fibers indicated that inflammation, proliferation and apoptosis mechanisms had been activated. Our results demonstrate that long-term exposure to high doses of albumin induces up-regulation of molecules involved in the tubular protein uptake machinery and suggest that albumin overload is able to trigger a regenerative process as well as an activation state which might lead in vivo to glomerular crescent formation.
Collapse
Affiliation(s)
- Giovanna Priante
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy.
| | - Monica Ceol
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Lisa Gianesello
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Claudia Maria Radu
- Thrombotic and Haemorrhagic Diseases Unit, Department of Medicine-DIMED, University of Padua, Padua, Italy
| | - Rachele Mantese
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Lucia Federica Stefanelli
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Martina Cacciapuoti
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Francesca K Martino
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Lorenzo Arcangelo Calò
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Franca Anglani
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Federico Nalesso
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| | - Dorella Del Prete
- Kidney Histomorphology and Molecular Biology Laboratory, Nephrology Unit, Department of Medicine - DIMED, University of Padua, Via Giustiniani 2, 35128, Padua, Italy
| |
Collapse
|
2
|
Rodrigues MC, Oliveira LBF, Vieira MAR, Caruso-Neves C, Peruchetti DB. Receptor-mediated endocytosis in kidney cells during physiological and pathological conditions. CURRENT TOPICS IN MEMBRANES 2024; 93:1-25. [PMID: 39181576 DOI: 10.1016/bs.ctm.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Mammalian cell membranes are very dynamic where they respond to several environmental stimuli by rearranging the membrane composition by basic biological processes, including endocytosis. In this context, receptor-mediated endocytosis, either clathrin-dependent or caveolae-dependent, is involved in different physiological and pathological conditions. In the last years, an important amount of evidence has been reported that kidney function involves the modulation of different types of endocytosis, including renal protein handling. In addition, the dysfunction of the endocytic machinery is involved with the development of proteinuria as well as glomerular and tubular injuries observed in kidney diseases associated with hypertension, diabetes, and others. In this present review, we will discuss the mechanisms underlying the receptor-mediated endocytosis in different glomerular cells and proximal tubule epithelial cells as well as their modulation by different factors during physiological and pathological conditions. These findings could help to expand the current understanding regarding renal protein handling as well as identify possible new therapeutic targets to halt the progression of kidney disease.
Collapse
Affiliation(s)
- Mariana C Rodrigues
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Laura B F Oliveira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria Aparecida R Vieira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAUDE/FAPERJ, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-Regenera, Conselho Nacional de Desenvolvimento Científico e Tecnológico/MCTI, Rio de Janeiro, RJ, Brazil
| | - Diogo B Peruchetti
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Nanobiofarmacêutica, INCT-NANOBiofar, Conselho Nacional de Desenvolvimento Científico e Tecnológico/MCTI, Belo Horizonte, MG, Brazil.
| |
Collapse
|
3
|
Han X, Yang F, Zhang Z, Hou Z, Sun Q, Su T, Lv W, Wang Z, Yuan C, Zhang G, Pi X, Long J, Liu H. 4EBP2-regulated protein translation has a critical role in high-fat diet-induced insulin resistance in hepatocytes. J Biol Chem 2023; 299:105315. [PMID: 37797700 PMCID: PMC10641227 DOI: 10.1016/j.jbc.2023.105315] [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: 06/28/2023] [Revised: 09/14/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023] Open
Abstract
A high-fat diet (HFD) plays a critical role in hepatocyte insulin resistance. Numerous models and factors have been proposed to elucidate the mechanism of palmitic acid (PA)-induced insulin resistance. However, proteomic studies of insulin resistance by HFD stimulation are usually performed under insulin conditions, leading to an unclear understanding of how a HFD alone affects hepatocytes. Here, we mapped the phosphorylation rewiring events in PA-stimulated HepG2 cells and found PA decreased the phosphorylation level of the eukaryotic translation initiation factor 4E-binding protein 2 (4EBP2) at S65/T70. Further experiments identified 4EBP2 as a key node of insulin resistance in either HFD mice or PA-treated cells. Reduced 4EBP2 levels increased glucose uptake and insulin sensitivity, whereas the 4EBP2_S65A/T70A mutation exacerbated PA-induced insulin resistance. Additionally, the nascent proteome revealed many glycolysis-related proteins translationally regulated by 4EBP2 such as hexokinase-2, pyruvate kinase PKM, TBC1 domain family member 4, and glucose-6-phosphate 1-dehydrogenase. In summary, we report the critical role of 4EBP2 in regulating HFD-stimulated insulin resistance in hepatocytes.
Collapse
Affiliation(s)
- Xiao Han
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Fei Yang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Zhengyi Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Zhanwu Hou
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Qiong Sun
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Tian Su
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Weiqiang Lv
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Zhen Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Chao Yuan
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Guanfei Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China
| | - Xin Pi
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi China.
| | - Huadong Liu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong China.
| |
Collapse
|
4
|
Chen M, Gu X. Emerging roles of proximal tubular endocytosis in renal fibrosis. Front Cell Dev Biol 2023; 11:1235716. [PMID: 37799275 PMCID: PMC10547866 DOI: 10.3389/fcell.2023.1235716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
Endocytosis is a crucial component of many pathological conditions. The proximal tubules are responsible for reabsorbing the majority of filtered water and glucose, as well as all the proteins filtered through the glomerular barrier via endocytosis, indicating an essential role in kidney diseases. Genetic mutations or acquired insults could affect the proximal tubule endocytosis processes, by disturbing or overstressing the endolysosomal system and subsequently activating different pathways, orchestrating renal fibrosis. This paper will review recent studies on proximal tubular endocytosis affected by other diseases and factors. Endocytosis plays a vital role in the development of renal fibrosis, and renal fibrosis could also, in turn, affect tubular endocytosis.
Collapse
Affiliation(s)
- Min Chen
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiangchen Gu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medicine, Shanghai Hospital of Civil Aviation Administration of China, Shanghai, China
| |
Collapse
|
5
|
Liu H, Ju A, Dong X, Luo Z, Tang J, Ma B, Fu Y, Luo Y. Young and undamaged recombinant albumin alleviates T2DM by improving hepatic glycolysis through EGFR and protecting islet β cells in mice. J Transl Med 2023; 21:89. [PMID: 36747238 PMCID: PMC9903539 DOI: 10.1186/s12967-023-03957-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Albumin is the most abundant protein in serum and serves as a transporter of free fatty acids (FFA) in blood vessels. In type 2 diabetes mellitus (T2DM) patients, the reduced serum albumin level is a risk factor for T2DM development and progression, although this conclusion is controversial. Moreover, there is no study on the effects and mechanisms of albumin administration to relieve T2DM. We examined whether the administration of young and undamaged recombinant albumin can alleviate T2DM in mice. METHODS The serum albumin levels and metabolic phenotypes including fasting blood glucose, glucose tolerance tests, and glucose-stimulated insulin secretion were studied in db/db mice or diet-induced obesity mice treated with saline or young, undamaged, and ultrapure rMSA. Apoptosis assays were performed at tissue and cell levels to determine the function of rMSA on islet β cell protection. Metabolic flux and glucose uptake assays were employed to investigate metabolic changes in saline-treated or rMSA-treated mouse hepatocytes and compared their sensitivity to insulin treatments. RESULTS In this study, treatment of T2DM mice with young, undamaged, and ultrapure recombinant mouse serum albumin (rMSA) increased their serum albumin levels, which resulted in a reversal of the disease including reduced fasting blood glucose levels, improved glucose tolerance, increased glucose-stimulated insulin secretion, and alleviated islet atrophy. At the cellular level, rMSA improved glucose uptake and glycolysis in hepatocytes. Mechanistically, rMSA reduced the binding between CAV1 and EGFR to increase EGFR activation leading to PI3K-AKT activation. Furthermore, rMSA extracellularly reduced the rate of fatty acid uptake by islet β-cells, which relieved the accumulation of intracellular ceramide, endoplasmic reticulum stress, and apoptosis. This study provided the first clear demonstration that injections of rMSA can alleviate T2DM in mice. CONCLUSION Our study demonstrates that increasing serum albumin levels can promote glucose homeostasis and protect islet β cells, which alleviates T2DM.
Collapse
Affiliation(s)
- Hongyi Liu
- grid.12527.330000 0001 0662 3178School of Life Sciences, Tsinghua University, Beijing, 100084 China ,grid.452723.50000 0004 7887 9190Tsinghua-Peking Joint Center for Life Sciences, Beijing, 100084 China ,The National Engineering Research Center for Protein Technology, Beijing, 100084 China ,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084 China
| | - Anji Ju
- grid.12527.330000 0001 0662 3178School of Life Sciences, Tsinghua University, Beijing, 100084 China ,The National Engineering Research Center for Protein Technology, Beijing, 100084 China ,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084 China
| | - Xuan Dong
- grid.12527.330000 0001 0662 3178School of Life Sciences, Tsinghua University, Beijing, 100084 China ,The National Engineering Research Center for Protein Technology, Beijing, 100084 China ,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084 China
| | - Zongrui Luo
- grid.12527.330000 0001 0662 3178School of Life Sciences, Tsinghua University, Beijing, 100084 China ,The National Engineering Research Center for Protein Technology, Beijing, 100084 China ,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084 China
| | - Jiaze Tang
- grid.12527.330000 0001 0662 3178School of Life Sciences, Tsinghua University, Beijing, 100084 China ,The National Engineering Research Center for Protein Technology, Beijing, 100084 China ,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084 China
| | - Boyuan Ma
- grid.12527.330000 0001 0662 3178School of Life Sciences, Tsinghua University, Beijing, 100084 China ,The National Engineering Research Center for Protein Technology, Beijing, 100084 China ,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084 China
| | - Yan Fu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,The National Engineering Research Center for Protein Technology, Beijing, 100084, China. .,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084, China. .,School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yongzhang Luo
- School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,Tsinghua-Peking Joint Center for Life Sciences, Beijing, 100084, China. .,The National Engineering Research Center for Protein Technology, Beijing, 100084, China. .,Beijing Key Laboratory for Protein Therapeutics, Beijing, 100084, China. .,School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
6
|
Nakamura M, Satoh N, Horita S, Nangaku M. Insulin-induced mTOR signaling and gluconeogenesis in renal proximal tubules: A mini-review of current evidence and therapeutic potential. Front Pharmacol 2022; 13:1015204. [PMID: 36299884 PMCID: PMC9589488 DOI: 10.3389/fphar.2022.1015204] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/27/2022] [Indexed: 12/02/2022] Open
Abstract
Energy is continuously expended in the body, and gluconeogenesis maintains glucose homeostasis during starvation. Gluconeogenesis occurs in the liver and kidneys. The proximal tubule is the primary location for renal gluconeogenesis, accounting for up to 25% and 60% of endogenous glucose production during fasting and after a meal, respectively. The mechanistic target of rapamycin (mTOR), which exists downstream of the insulin pathway, plays an important role in regulating proximal tubular gluconeogenesis. mTOR is an atypical serine/threonine kinase present in two complexes. mTORC1 phosphorylates substrates that enhance anabolic processes such as mRNA translation and lipid synthesis and catabolic processes such as autophagy. mTORC2 regulates cytoskeletal dynamics and controls ion transport and proliferation via phosphorylation of SGK1. Therefore, mTOR signaling defects have been implicated in various pathological conditions, including cancer, cardiovascular disease, and diabetes. However, concrete elucidations of the associated mechanisms are still unclear. This review provides an overview of mTOR and describes the relationship between mTOR and renal.
Collapse
Affiliation(s)
- Motonobu Nakamura
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
7
|
Silva-Aguiar RP, Peruchetti DB, Pinheiro AAS, Caruso-Neves C, Dias WB. O-GlcNAcylation in Renal (Patho)Physiology. Int J Mol Sci 2022; 23:ijms231911260. [PMID: 36232558 PMCID: PMC9569498 DOI: 10.3390/ijms231911260] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/29/2022] Open
Abstract
Kidneys maintain internal milieu homeostasis through a well-regulated manipulation of body fluid composition. This task is performed by the correlation between structure and function in the nephron. Kidney diseases are chronic conditions impacting healthcare programs globally, and despite efforts, therapeutic options for its treatment are limited. The development of chronic degenerative diseases is associated with changes in protein O-GlcNAcylation, a post-translation modification involved in the regulation of diverse cell function. O-GlcNAcylation is regulated by the enzymatic balance between O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) which add and remove GlcNAc residues on target proteins, respectively. Furthermore, the hexosamine biosynthetic pathway provides the substrate for protein O-GlcNAcylation. Beyond its physiological role, several reports indicate the participation of protein O-GlcNAcylation in cardiovascular, neurodegenerative, and metabolic diseases. In this review, we discuss the impact of protein O-GlcNAcylation on physiological renal function, disease conditions, and possible future directions in the field.
Collapse
Affiliation(s)
- Rodrigo P. Silva-Aguiar
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Diogo B. Peruchetti
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
| | - Ana Acacia S. Pinheiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro 21045-900, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro 21045-900, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-902, Brazil
| | - Wagner B. Dias
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil
- Correspondence:
| |
Collapse
|
8
|
Pellegrina D, Bahcheli AT, Krassowski M, Reimand J. Human phospho-signaling networks of SARS-CoV-2 infection are rewired by population genetic variants. Mol Syst Biol 2022; 18:e10823. [PMID: 35579274 PMCID: PMC9112486 DOI: 10.15252/msb.202110823] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
SARS-CoV-2 infection hijacks signaling pathways and induces protein-protein interactions between human and viral proteins. Human genetic variation may impact SARS-CoV-2 infection and COVID-19 pathology; however, the genetic variation in these signaling networks remains uncharacterized. Here, we studied human missense single nucleotide variants (SNVs) altering phosphorylation sites modulated by SARS-CoV-2 infection, using machine learning to identify amino acid substitutions altering kinase-bound sequence motifs. We found 2,033 infrequent phosphorylation-associated SNVs (pSNVs) that are enriched in sequence motif alterations, potentially reflecting the evolution of signaling networks regulating host defenses. Proteins with pSNVs are involved in viral life cycle and host responses, including RNA splicing, interferon response (TRIM28), and glucose homeostasis (TBC1D4) with potential associations with COVID-19 comorbidities. pSNVs disrupt CDK and MAPK substrate motifs and replace these with motifs of Tank Binding Kinase 1 (TBK1) involved in innate immune responses, indicating consistent rewiring of signaling networks. Several pSNVs associate with severe COVID-19 and hospitalization (STARD13, ARFGEF2). Our analysis highlights potential genetic factors contributing to inter-individual variation of SARS-CoV-2 infection and COVID-19 and suggests leads for mechanistic and translational studies.
Collapse
Affiliation(s)
- Diogo Pellegrina
- Computational Biology ProgramOntario Institute for Cancer ResearchTorontoONCanada
| | - Alexander T Bahcheli
- Computational Biology ProgramOntario Institute for Cancer ResearchTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Michal Krassowski
- Medical Sciences DivisionNuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Jüri Reimand
- Computational Biology ProgramOntario Institute for Cancer ResearchTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
| |
Collapse
|
9
|
Silva-Aguiar RP, Peruchetti DB, Florentino LS, Takiya CM, Marzolo MP, Dias WB, Pinheiro AAS, Caruso-Neves C. Albumin Expands Albumin Reabsorption Capacity in Proximal Tubule Epithelial Cells through a Positive Feedback Loop between AKT and Megalin. Int J Mol Sci 2022; 23:ijms23020848. [PMID: 35055044 PMCID: PMC8776186 DOI: 10.3390/ijms23020848] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/05/2022] [Accepted: 01/11/2022] [Indexed: 12/20/2022] Open
Abstract
Renal proximal tubule cells (PTECs) act as urine gatekeepers, constantly and efficiently avoiding urinary protein waste through receptor-mediated endocytosis. Despite its importance, little is known about how this process is modulated in physiologic conditions. Data suggest that the phosphoinositide-3-kinase (PI3K)/protein kinase B (AKT) pathway regulates PTEC protein reabsorption. Here, we worked on the hypothesis that the physiologic albumin concentration and PI3K/AKT pathway form a positive feedback loop to expand endocytic capacity. Using LLC-PK1 cells, a model of PTECs, we showed that the PI3K/AKT pathway is required for megalin recycling and surface expression, affecting albumin uptake. Inhibition of this pathway stalls megalin at EEA1+ endosomes. Physiologic albumin concentration (0.01 mg/mL) activated AKT; this depends on megalin-mediated albumin endocytosis and requires previous activation of PI3K/mTORC2. This effect is correlated to the increase in albumin endocytosis, a phenomenon that we refer to as “albumin-induced albumin endocytosis”. Mice treated with L-lysine present decreased albumin endocytosis leading to proteinuria and albuminuria associated with inhibition of AKT activity. Renal cortex explants obtained from control mice treated with MK-2206 decreased albumin uptake and promoted megalin internalization. Our data highlight the mechanism behind the capacity of PTECs to adapt albumin reabsorption to physiologic fluctuations in its filtration, avoiding urinary excretion.
Collapse
Affiliation(s)
- Rodrigo P. Silva-Aguiar
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.P.S.-A.); (D.B.P.); (L.S.F.); (C.M.T.); (W.B.D.); (A.A.S.P.)
| | - Diogo B. Peruchetti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.P.S.-A.); (D.B.P.); (L.S.F.); (C.M.T.); (W.B.D.); (A.A.S.P.)
| | - Lucas S. Florentino
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.P.S.-A.); (D.B.P.); (L.S.F.); (C.M.T.); (W.B.D.); (A.A.S.P.)
| | - Christina M. Takiya
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.P.S.-A.); (D.B.P.); (L.S.F.); (C.M.T.); (W.B.D.); (A.A.S.P.)
| | - María-Paz Marzolo
- Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile, Santiago 8330163, Chile;
| | - Wagner B. Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.P.S.-A.); (D.B.P.); (L.S.F.); (C.M.T.); (W.B.D.); (A.A.S.P.)
| | - Ana Acacia S. Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.P.S.-A.); (D.B.P.); (L.S.F.); (C.M.T.); (W.B.D.); (A.A.S.P.)
- Redes de Pesquisa em Nanotecnologia para Saúde, NanoSaúde/FAPERJ, Rio de Janeiro 21040-900, Brazil
| | - Celso Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (R.P.S.-A.); (D.B.P.); (L.S.F.); (C.M.T.); (W.B.D.); (A.A.S.P.)
- Redes de Pesquisa em Nanotecnologia para Saúde, NanoSaúde/FAPERJ, Rio de Janeiro 21040-900, Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-Regenera, Rio de Janeiro 21941-902, Brazil
- Correspondence: ; Tel.: +55-21-3938-6582
| |
Collapse
|
10
|
Lee Y, Kim MH, Alves DR, Kim S, Lee LP, Sung JH, Park S. Gut-Kidney Axis on Chip for Studying Effects of Antibiotics on Risk of Hemolytic Uremic Syndrome by Shiga Toxin-Producing Escherichia coli. Toxins (Basel) 2021; 13:toxins13110775. [PMID: 34822559 PMCID: PMC8622205 DOI: 10.3390/toxins13110775] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/23/2021] [Accepted: 10/30/2021] [Indexed: 12/30/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) infects humans by colonizing the large intestine, and causes kidney damage by secreting Shiga toxins (Stxs). The increased secretion of Shiga toxin 2 (Stx2) by some antibiotics, such as ciprofloxacin (CIP), increases the risk of hemolytic–uremic syndrome (HUS), which can be life-threatening. However, previous studies evaluating this relationship have been conflicting, owing to the low frequency of EHEC infection, very small number of patients, and lack of an appropriate animal model. In this study, we developed gut–kidney axis (GKA) on chip for co-culturing gut (Caco-2) and kidney (HKC-8) cells, and observed both STEC O157:H7 (O157) infection and Stx intoxication in the gut and kidney cells on the chip, respectively. Without any antibiotic treatment, O157 killed both gut and kidney cells in GKA on the chip. CIP treatment reduced O157 infection in the gut cells, but increased Stx2-induced damage in the kidney cells, whereas the gentamycin treatment reduced both O157 infection in the gut cells and Stx2-induced damage in the kidney cells. This is the first report to recapitulate a clinically relevant situation, i.e., that CIP treatment causes more damage than gentamicin treatment. These results suggest that GKA on chip is very useful for simultaneous observation of O157 infections and Stx2 poisoning in gut and kidney cells, making it suitable for studying the effects of antibiotics on the risk of HUS.
Collapse
Affiliation(s)
- Yugyeong Lee
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Department of Chemical Engineering, Hongik University, Seoul 04066, Korea
| | - David Rodrigues Alves
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1362035 Lisboa, Portugal
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Luke P. Lee
- Institute of Quantum Biophysics (IQB), Department of Biophysics, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, Korea
- Correspondence: (J.H.S.); (S.P.); Tel.: +82-2-320-3067 (J.H.S.); +82-31-290-7431 (S.P.)
| | - Sungsu Park
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Institute of Quantum Biophysics (IQB), Department of Biophysics, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- Correspondence: (J.H.S.); (S.P.); Tel.: +82-2-320-3067 (J.H.S.); +82-31-290-7431 (S.P.)
| |
Collapse
|
11
|
Faria J, Gerritsen KGF, Nguyen TQ, Mihaila SM, Masereeuw R. Diabetic proximal tubulopathy: Can we mimic the disease for in vitro screening of SGLT inhibitors? Eur J Pharmacol 2021; 908:174378. [PMID: 34303664 DOI: 10.1016/j.ejphar.2021.174378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 11/27/2022]
Abstract
Diabetic kidney disease (DKD) is the foremost cause of renal failure. While the glomeruli are severely affected in the course of the disease, the main determinant for disease progression is the tubulointerstitial compartment. DKD does not develop in the absence of hyperglycemia. Since the proximal tubule is the major player in glucose reabsorption, it has been widely studied as a therapeutic target for the development of new therapies. Currently, there are several proximal tubule cell lines available, being the human kidney-2 (HK-2) and human kidney clone-8 (HKC-8) cell lines the ones widely used for studying mechanisms of DKD. Studies in these models have pushed forward the understanding on how DKD unravels, however, these cell culture models possess limitations that hamper research, including lack of transporters and dedifferentiation. The sodium-glucose cotransporters (SGLT) are identified as key players in glucose reabsorption and pharmacological inhibitors have shown to be beneficial for the long-term clinical outcome in DKD. However, their mechanism of action has, as of yet, not been fully elucidated. To comprehend the protective effects of SGLT inhibitors, it is essential to understand the complete functional, structural, and molecular features of the disease, which until now have been difficult to recapitulate. This review addresses the molecular events of diabetic proximal tubulopathy. In addition, we evaluate the protective role of SGLT inhibitors in cardiovascular and renal outcomes, and provide an overview of various in vitro models mimicking diabetic proximal tubulopathy used so far. Finally, new insights on advanced in vitro systems to surpass past limitations are postulated.
Collapse
Affiliation(s)
- João Faria
- Div. Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands
| | - Karin G F Gerritsen
- Dept. Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands
| | - Tri Q Nguyen
- Dept. Pathology, University Medical Center Utrecht, the Netherlands
| | - Silvia M Mihaila
- Div. Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands; Dept. Nephrology and Hypertension, University Medical Center Utrecht, the Netherlands
| | - Rosalinde Masereeuw
- Div. Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, the Netherlands.
| |
Collapse
|
12
|
Urinary Exosomal MicroRNA Signatures in Nephrotic, Biopsy-Proven Diabetic Nephropathy. J Clin Med 2020; 9:jcm9041220. [PMID: 32340338 PMCID: PMC7231152 DOI: 10.3390/jcm9041220] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease (CKD). Elucidating the mechanisms underlying proteinuria in DKD is crucial because it is a common problem in DKD-related mortality and morbidity. MicroRNAs (miRs) associated with DKD have been detected in experimental diabetes models and in patients with both diabetes and CKD. Here, we aimed to investigate pathologic miRs in diabetic nephropathy (DN) by prospectively following six nephrotic, biopsy-proven isolated DN patients (enrolled between August 2015 and July 2017) for one year. The urinary exosomes were isolated at the time of the biopsy and the contained miRs were analyzed by next-generation sequencing. The results were compared to the control group, composed of age-, gender-, and CKD stage-matched patients with proteinuric CKD who did not present diabetes. Among the 72 identified miRs, we investigated eight (miR-188-5p, miR-150-3p, miR-760, miR-3677-3p, miR-548ah-3p, miR-548p, miR-320e, and miR-23c) exhibiting the strongest upregulation (13–15 fold) and two (miR-133a-3p and miR-153-3p) with the strongest downregulation (7–9 fold). The functional analysis of these miRs showed that they were involved in known and novel pathways of DN, supporting their pathologic roles. The bioinformatics-based prediction of the target genes of these miRs will inspire future research on the mechanisms underlying DN pathogenesis.
Collapse
|
13
|
Pereira-Moreira R, Muscelli E. Effect of Insulin on Proximal Tubules Handling of Glucose: A Systematic Review. J Diabetes Res 2020; 2020:8492467. [PMID: 32377524 PMCID: PMC7180501 DOI: 10.1155/2020/8492467] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/18/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023] Open
Abstract
Renal proximal tubules reabsorb glucose from the glomerular filtrate and release it back into the circulation. Modulation of glomerular filtration and renal glucose disposal are some of the insulin actions, but little is known about a possible insulin effect on tubular glucose reabsorption. This review is aimed at synthesizing the current knowledge about insulin action on glucose handling by proximal tubules. Method. A systematic article selection from Medline (PubMed) and Embase between 2008 and 2019. 180 selected articles were clustered into topics (renal insulin handling, proximal tubule glucose transport, renal gluconeogenesis, and renal insulin resistance). Summary of Results. Insulin upregulates its renal uptake and degradation, and there is probably a renal site-specific insulin action and resistance; studies in diabetic animal models suggest that insulin increases renal SGLT2 protein content; in vivo human studies on glucose transport are few, and results of glucose transporter protein and mRNA contents are conflicting in human kidney biopsies; maximum renal glucose reabsorptive capacity is higher in diabetic patients than in healthy subjects; glucose stimulates SGLT1, SGLT2, and GLUT2 in renal cell cultures while insulin raises SGLT2 protein availability and activity and seems to directly inhibit the SGLT1 activity despite it activating this transporter indirectly. Besides, insulin regulates SGLT2 inhibitor bioavailability, inhibits renal gluconeogenesis, and interferes with Na+K+ATPase activity impacting on glucose transport. Conclusion. Available data points to an important insulin participation in renal glucose handling, including tubular glucose transport, but human studies with reproducible and comparable method are still needed.
Collapse
Affiliation(s)
- Ricardo Pereira-Moreira
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Zip Code: 13083-887, Brazil
| | - Elza Muscelli
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Zip Code: 13083-887, Brazil
| |
Collapse
|
14
|
Singh S, Sharma R, Kumari M, Tiwari S. Insulin receptors in the kidneys in health and disease. World J Nephrol 2019; 8:11-22. [PMID: 30705868 PMCID: PMC6354081 DOI: 10.5527/wjn.v8.i1.11] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/15/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023] Open
Abstract
Insulin is an important hormone that affects various metabolic processes, including kidney function. Impairment in insulin’s action leads to insulin resistance in the target tissue. Besides defects in post-receptor insulin signaling, impairment at the receptor level could significantly affect insulin sensitivity of the target tissue. The kidney is a known target of insulin; however, whether the kidney develops “insulin resistance” is debatable. Regulation of the insulin receptor (IR) expression and its function is very well studied in major metabolic tissues like liver, skeletal muscles, and adipose tissue. The physiological relevance of IRs in the kidney has recently begun to be clarified. The credit goes to studies that showed a wide distribution of IR throughout the nephron segments and their reduced expression in the insulin resistance state. Moreover, altered renal and systemic metabolism observed in mice with targeted deletion of the IR from various epithelial cells of the kidney has strengthened this proposition. In this review, we recapitulate the crucial findings from literature that have expanded our knowledge regarding the significance of the renal IR in normal- and insulin-resistance states.
Collapse
Affiliation(s)
- Sarojini Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rajni Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Manju Kumari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Swasti Tiwari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
15
|
Kumari M, Sharma R, Pandey G, Ecelbarger CM, Mishra P, Tiwari S. Deletion of insulin receptor in the proximal tubule and fasting augment albumin excretion. J Cell Biochem 2019; 120:10688-10696. [PMID: 30644120 DOI: 10.1002/jcb.28359] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 11/29/2018] [Indexed: 12/29/2022]
Abstract
The contribution of proximal tubules (PT) to albumin uptake is now well recognized, however, its regulation is understudied area. There are reports suggesting that insulin resistance is associated with the development of albuminuria in nondiabetic individuals. We have previously reported reduced insulin receptor (IR) expression in renal-tubular-epithelial cells, including PT in various models of insulin resistance. However, the effect of a physiological fall in insulin levels and the role for IR in PT in tubular albumin uptake is not clear. To address these gaps in our understanding, we estimated urine excretion and renal uptake of albumin in fasted and fed C57Bl/6 mice injected with fluorescein isothiocyanate (FITC)-albumin (5 µg/mL/kg body weight, intraperitoneal, n = 6 per group). In addition, we compared spot urine analysis from 33 clinically healthy humans after overnight fasting (when insulin levels are lower than in the fed state) and then at 2 hours after 75 g oral glucose challenge (postprandial). Fasted mice had attenuated renal uptake of FITC-albumin and higher excretion in urine, relative to fed mice ( P = 0.04). Moreover, a significant drop in urine albumin-to-creatinine ratio (ACR) and urine albumin concentration (UAC) was observed in the postprandial state in these subjects ( P = 0.001 and P = 0.017, for ACR and UAC, respectively). The drop was negatively associated with postprandial blood glucose levels (ρ = -0.36, P = 0.03 for ΔUAC and ρ = -0.34, P = 0.05 for ΔACR). To test the role of IR in PT, we analyzed 24-hour urine albumin excretion in male mice with targeted deletion of IR from PT (insulin receptor knockout [IRKO]) and their wild-type (WT) littermates ( n = 7 per group). IRKO mice had significantly higher 24-hour urine albumin excretion relative to WT. Moreover, kidneys from KO mice revealed reduced expression of megalin and cubulin proteins in the PT relative to the WT. We also demonstrated insulin (100 nM) induced albumin internalization in human proximal tubule cells (hPT) and this effect of insulin was attenuated in hydroxy-2-naphthalenylmethylphosphonic acid (100 µM), a tyrosine kinase inhibitor, pretreated hPT. Our findings revealed albumin excretion was attenuated by glucose administration to fasting individuals implying a regulatory role for insulin in PT albumin reabsorption. Thus albuminuria associated with insulin resistance/diabetes may relate not only to glomerular dysfunction but also to impairment in insulin-mediated reabsorption.
Collapse
Affiliation(s)
- Manju Kumari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rajni Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Gaurav Pandey
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Carolyn M Ecelbarger
- Department of Medicine, Division of Endocrinology and Metabolism, Georgetown University, Washington, District of Columbia
| | - Prabhaker Mishra
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Swasti Tiwari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
16
|
Intracellular albumin overload elicits endoplasmic reticulum stress and PKC-delta/p38 MAPK pathway activation to induce podocyte apoptosis. Sci Rep 2018; 8:18012. [PMID: 30573754 PMCID: PMC6301950 DOI: 10.1038/s41598-018-36933-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022] Open
Abstract
Podocyte injury is closely related to proteinuria and the progression of chronic kidney disease (CKD). Currently, there is no conclusive understanding about the mechanisms involved in albumin overload and podocyte apoptosis response. In this study, we sought to explore the ways by which intracellular albumin can mediate podocyte apoptosis. Here, immortalized mouse podocytes were treated with bovine serum albumin (BSA) at different times and concentrations, in the presence or absence of SB203580 (0.1 µM, inhibitor of mitogen-activated-protein kinase – p38MAPK). Using immunofluorescence images, flow cytometry and immunoblotting, we observed a time-dependent intracellular accumulation of fluorescent albumin-FITC-BSA, followed by concentration-and time-dependent effect of intracellular albumin overload on podocyte apoptosis, which was mediated by increased expression of the chaperone glucose-regulated-protein 78 (GRP 78) and phosphorylated inositol-requiring enzyme 1 alpha (pIRE1-α), as well as protein kinase C delta (PKC-δ), p38MAPK and cleaved caspase 12 expression. SB203580 prevented the cleavage of caspase 12 and the albumin-mediated podocyte apoptosis. These results suggest that intracellular albumin overload is associated with endoplasmic reticulum (ER) stress and upregulation of PKC-δ/p38MAPK/caspase 12 pathway, which may be a target for future therapeutic of albumin-induced podocyte apoptosis.
Collapse
|
17
|
Bryniarski MA, Yee BM, Jaffri I, Chaves LD, Yu JA, Guan X, Ghavam N, Yacoub R, Morris ME. Increased megalin expression in early type 2 diabetes: role of insulin-signaling pathways. Am J Physiol Renal Physiol 2018; 315:F1191-F1207. [PMID: 29949391 DOI: 10.1152/ajprenal.00210.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The megalin/cubilin complex is responsible for the majority of serum protein reclamation in the proximal tubules. The current study examined if decreases in their renal expression, along with the albumin recycling protein neonatal Fc receptor (FcRn) could account for proteinuria/albuminuria in the Zucker diabetic fatty rat model of type 2 diabetes. Immunoblots of renal cortex samples obtained at worsening disease stages demonstrated no loss in megalin, cubilin, or FcRn, even when proteinuria was measured. Additionally, early diabetic rats exhibited significantly increased renal megalin expression when compared with controls (adjusted P < 0.01). Based on these results, the ability of insulin to increase megalin was examined in a clonal subpopulation of the opossum kidney proximal tubule cell line. Insulin treatments (24 h, 100 nM) under high glucose conditions significantly increased megalin protein ( P < 0.0001), mRNA ( P < 0.0001), and albumin endocytosis. The effect on megalin expression was prevented with inhibitors against key effectors of insulin intracellular signaling, phosphatidylinositide 3-kinase and Akt. Studies using rapamycin to inhibit the mechanistic target of rapamycin complex 1 (mTORC1) resulted in a loss of insulin-induced megalin expression. However, subsequent evaluation demonstrated these effects were independent of initial mTORC1 suppression. The presented results provide insight into the expression of megalin, cubilin, and FcRn in type 2 diabetes, which may be impacted by elevated insulin and glucose. Furthermore, proximal tubule endocytic activity in early diabetics may be enhanced, a process that could have a significant role in proteinuria-induced renal damage.
Collapse
Affiliation(s)
- Mark A Bryniarski
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo , New York
| | - Benjamin M Yee
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo , New York
| | - Irum Jaffri
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo , New York
| | - Lee D Chaves
- Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , New York
| | - Jin Ah Yu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo , New York
| | - Xiaowen Guan
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo , New York
| | - Nazanin Ghavam
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo , New York
| | - Rabi Yacoub
- Department of Internal Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , New York
| | - Marilyn E Morris
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo , New York
| |
Collapse
|
18
|
Vitlov Uljević M, Bočina I, Restović I, Kunac N, Mašek T, Kretzschmar G, Grobe M, Šarić M, Vukojević K, Saraga-Babić M, Filipović N. Reabsorption in the proximal tubuli-ultrastructural evidence for a novel aspect of renal VEGF trafficking. Cell Tissue Res 2018; 374:189-201. [PMID: 29804263 DOI: 10.1007/s00441-018-2850-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 04/03/2018] [Indexed: 10/16/2022]
Abstract
Many clinical and experimental studies have revealed VEGF as an important factor in the pathophysiology of renal damage during diabetes mellitus (DM). Anti-VEGF therapy is in clinical use for treatment of DM and other diabetes-related (and unrelated) diseases. Nevertheless, little is known about the metabolism of VEGF in the kidneys. In order to determine the ultrastructural localization of VEGF in the kidney, we study the distribution of VEGF in the kidney of rats by using immunogold immunohistochemistry. Our light-microscopic data showed remarkable re-distribution of VEGF in proximal tubular cells (PTCs) during prolonged hyperglycemia, a DM type 2 model (DM2), which was confirmed by transmission electron microscopy (TEM) findings. TEM findings revealed an initial presence of VEGF in the vesicular transport apparatus of PTCs in healthy rats and its gradual translocation to the apical membrane of PTCs after renal damage caused by high sucrose treatment. The presented data add to our understanding of kidney VEGF trafficking, providing novel insight into the renal metabolism and pharmacodynamics of the cytokine. This could have a high impact on the use of VEGF and anti-VEGF therapy in different renal diseases.
Collapse
Affiliation(s)
- Marija Vitlov Uljević
- Department of Anatomy, Histology and Embryology, Laboratory for Neurocardiology, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
| | - Ivana Bočina
- Department of Biology, University of Split Faculty of Science, Split, Croatia
| | - Ivana Restović
- Department of Teacher Education, University of Split Faculty of Humanities and Social Sciences, Split, Croatia
| | - Nenad Kunac
- Department of Pathology, University of Split School of Medicine, Split, Croatia
| | - Tomislav Mašek
- Department of Animal Nutrition and Dietetics, University of Zagreb Faculty of Veterinary Medicine, Zagreb, Croatia
| | - Genia Kretzschmar
- Department of Anatomy, Histology and Embryology, Laboratory for Neurocardiology, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
| | - Maximilian Grobe
- Department of Anatomy, Histology and Embryology, Laboratory for Neurocardiology, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
| | - Mia Šarić
- Department of Anatomy, Histology and Embryology, Laboratory for Neurocardiology, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, Laboratory for Neurocardiology, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia.,Department of Anatomy, Histology and Embryology, Laboratory for Early Human Development, University of Split School of Medicine, Split, Croatia
| | - Mirna Saraga-Babić
- Department of Anatomy, Histology and Embryology, Laboratory for Early Human Development, University of Split School of Medicine, Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, Laboratory for Neurocardiology, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia.
| |
Collapse
|
19
|
Kurosaki Y, Imoto A, Kawakami F, Yokoba M, Takenaka T, Ichikawa T, Katagiri M, Ishii N. Oxidative stress increases megalin expression in the renal proximal tubules during the normoalbuminuric stage of diabetes mellitus. Am J Physiol Renal Physiol 2018; 314:F462-F470. [DOI: 10.1152/ajprenal.00108.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Megalin, an endocytic receptor expressed in proximal tubule cells, plays a critical role in renal tubular protein reabsorption and is associated with the albuminuria observed in diabetic nephropathy. We have previously reported increased oxidant production in the renal cortex during the normoalbuminuric stage of diabetes mellitus (DM); however, the relationship between oxidative stress and renal megalin expression during the normoalbuminuric stage of DM remains unclear. In the present study, we evaluated whether oxidative stress affects megalin expression in the normoalbuminuric stage of DM in a streptozotocin-induced diabetic rat model and in immortalized human proximal tubular cells (HK-2). We demonstrated that increased expression of renal megalin accompanies oxidative stress during the early stage of DM, before albuminuria development. Telmisartan treatment prevented the diabetes-induced elevation in megalin level, possibly through an oxidative stress-dependent mechanism. In HK-2 cells, hydrogen peroxide significantly increased megalin levels in a dose- and time-dependent manner; however, the elevation in megalin expression was decreased following prolonged exposure to severe oxidative stress induced by 0.4 mmol/l hydrogen peroxide. High-glucose treatment also significantly increased megalin expression in HK-2 cells. Concurrent administration of the antioxidant N-acetyl-cysteine blocked the effects of high glucose on megalin expression. Furthermore, the hydrogen peroxide-induced increase in megalin expression was blocked by treatment with phosphatidylinositol 3-kinase and Akt inhibitors. Increase of phosphorylated Akt expression was also seen in the renal cortex of diabetic rats. Taken together, our results indicate that mild oxidative stress increases renal megalin expression through the phosphatidylinositol 3-kinase-Akt pathway in the normoalbuminuric stage of DM.
Collapse
Affiliation(s)
- Yoshifumi Kurosaki
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Akemi Imoto
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Fumitaka Kawakami
- Department of Pathological Biochemistry, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Masanori Yokoba
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Tsuneo Takenaka
- Department of Medicine, International University of Health and Welfare, Tokyo, Japan
| | - Takafumi Ichikawa
- Department of Pathological Biochemistry, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Masato Katagiri
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| | - Naohito Ishii
- Department of Medical Laboratory Sciences, Kitasato University School of Allied Health Sciences, Kanagawa, Japan
| |
Collapse
|
20
|
Fatah H, Benfaed N, Chana RS, Chunara MH, Barratt J, Baines RJ, Brunskill NJ. Reduced proximal tubular expression of protein endocytic receptors in proteinuria is associated with urinary receptor shedding. Nephrol Dial Transplant 2017; 33:934-943. [DOI: 10.1093/ndt/gfx321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 10/01/2017] [Indexed: 02/06/2023] Open
Affiliation(s)
- Hiwa Fatah
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Nura Benfaed
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Ravinder S Chana
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Mohamed H Chunara
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Jonathan Barratt
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, UK
- Department of Nephrology, Leicester General Hospital, Leicester, UK
| | - Richard J Baines
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, UK
- Department of Nephrology, Leicester General Hospital, Leicester, UK
| | - Nigel J Brunskill
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, UK
- Department of Nephrology, Leicester General Hospital, Leicester, UK
| |
Collapse
|
21
|
Gianesello L, Priante G, Ceol M, Radu CM, Saleem MA, Simioni P, Terrin L, Anglani F, Del Prete D. Albumin uptake in human podocytes: a possible role for the cubilin-amnionless (CUBAM) complex. Sci Rep 2017; 7:13705. [PMID: 29057905 PMCID: PMC5651885 DOI: 10.1038/s41598-017-13789-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 10/02/2017] [Indexed: 12/23/2022] Open
Abstract
Albumin re-uptake is a receptor-mediated pathway located in renal proximal tubuli. There is increasing evidence of glomerular protein handling by podocytes, but little is known about the mechanism behind this process. In this study, we found that human podocytes in vitro are committed to internalizing albumin through a receptor-mediated mechanism even after exposure to low doses of albumin. We show that these cells express cubilin, megalin, ClC-5, amnionless and Dab2, which are partners in the tubular machinery. Exposing human podocytes to albumin overload prompted an increase in CUBILIN, AMNIONLESS and CLCN5 gene expression. Inhibiting cubilin led to a reduction in albumin uptake, highlighting its importance in this mechanism. We demonstrated that human podocytes are committed to performing endocytosis via a receptor-mediated mechanism even in the presence of low doses of albumin. We also disclosed that protein overload first acts on the expression of the cubilin-amnionless (CUBAM) complex in these cells, then involves the ClC-5 channel, providing the first evidence for a possible role of the CUBAM complex in albumin endocytosis in human podocytes.
Collapse
Affiliation(s)
- Lisa Gianesello
- Clinical Nephrology, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy
| | - Giovanna Priante
- Clinical Nephrology, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy
| | - Monica Ceol
- Clinical Nephrology, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy
| | - Claudia M Radu
- Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy
| | - Moin A Saleem
- Academic and Children's Renal Unit, Dorothy Hodgkin Building, BS8 1TH, Bristol, United Kingdom
| | - Paolo Simioni
- Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy
| | - Liliana Terrin
- Clinical Nephrology, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy
| | - Franca Anglani
- Clinical Nephrology, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy
| | - Dorella Del Prete
- Clinical Nephrology, Department of Medicine - DIMED, University of Padua, 35129, Padua, Italy.
| |
Collapse
|
22
|
Kuczkowski A, Brinkkoetter PT. Metabolism and homeostasis in the kidney: metabolic regulation through insulin signaling in the kidney. Cell Tissue Res 2017; 369:199-210. [DOI: 10.1007/s00441-017-2619-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 03/22/2017] [Indexed: 02/07/2023]
|
23
|
Wen L, Andersen PK, Husum DMU, Nørregaard R, Zhao Z, Liu Z, Birn H. MicroRNA-148b regulates megalin expression and is associated with receptor downregulation in mice with unilateral ureteral obstruction. Am J Physiol Renal Physiol 2017; 313:F210-F217. [PMID: 28331063 DOI: 10.1152/ajprenal.00585.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/02/2017] [Accepted: 03/16/2017] [Indexed: 11/22/2022] Open
Abstract
Megalin is a multiligand, endocytic receptor that is important for the normal, proximal tubule reabsorption of filtered proteins, hormones, enzymes, essential nutrients, and nephrotoxins. Megalin dysfunction has been associated with acute, as well as chronic kidney diseases. Tubular proteinuria has been observed following unilateral ureteral obstruction (UUO), suggesting megalin dysfunction; however, the pathophysiological mechanism has not been determined. To identify potential regulators of megalin expression, we examined renal microRNAs (miRNAs) expression and observed an upregulation of microRNA-148b (miR-148b) in obstructed mouse kidneys 7 days after UUO, which was associated with a significant reduction in proximal tubule megalin expression and accumulation of megalin ligands. By in silico miRNA target prediction analysis, we identified megalin messenger RNA (mRNA) as a potential target of miR-148b and confirmed using a dual-luciferase reporter assay that miR-148b targeted the 3'-untranslated region of the megalin gene. Transfection of LLC-PK1 cells with miR-148b mimic reduced endogenous megalin mRNA and protein levels in a concentration-dependent manner, while transfection with miR-148b inhibitor resulted in an increase. Our findings suggest that miR-148b directly downregulates megalin expression and that miR-148b negatively regulates megalin expression in UUO-induced kidney injury. Furthermore, the identification of a miRNA regulating megalin expression may allow for targeted interventions to modulate megalin function and proximal tubule uptake of proteins, as well as other ligands.
Collapse
Affiliation(s)
- Lu Wen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pia K Andersen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Dina M U Husum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; and
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; and
| | - Zhanzheng Zhao
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhangsuo Liu
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Henrik Birn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; .,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|