1
|
Hackl A, Weber LT. The Ca 2+-actin-cytoskeleton axis in podocytes is an important, non-immunologic target of immunosuppressive therapy in proteinuric kidney diseases. Pediatr Nephrol 2025:10.1007/s00467-025-06670-z. [PMID: 39856247 DOI: 10.1007/s00467-025-06670-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025]
Abstract
The integrity of the filtration barrier of the kidney relies on the proper composition of podocyte interdigitating foot processes. Their architecture is supported by a complex actin-cytoskeleton. Following podocyte stress or injury, podocytes encounter structural changes, including rearrangement of the actin network and subsequent effacement of the foot processes. Immunosuppressive drugs, which are currently used as treatment in proteinuric kidney diseases, have been shown to exert not only immune-mediated effects. This review will focus on the direct effects of glucocorticoids, cyclosporine A, tacrolimus, mycophenolate mofetil, and rituximab on podocytes by regulation of Ca2+ ion channels and consecutive downstream signaling which prevent cytoskeletal rearrangements and ultimately proteinuria. In addition, the efficacy of these drugs in genetic nephrotic syndrome will be discussed.
Collapse
Affiliation(s)
- Agnes Hackl
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany.
| | - Lutz T Weber
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Street 62, 50937, Cologne, Germany
| |
Collapse
|
2
|
Palazzo E, Marabese I, Ricciardi F, Guida F, Luongo L, Maione S. The influence of glutamate receptors on insulin release and diabetic neuropathy. Pharmacol Ther 2024; 263:108724. [PMID: 39299577 DOI: 10.1016/j.pharmthera.2024.108724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Diabetes causes macrovascular and microvascular complications such as peripheral neuropathy. Glutamate regulates insulin secretion in pancreatic β-cells, and its increased activity in the central nervous system is associated with peripheral neuropathy in animal models of diabetes. One strategy to modulate glutamatergic activity consists in the pharmacological manipulation of metabotropic glutamate receptors (mGluRs), which, compared to the ionotropic receptors, allow for a fine-tuning of neurotransmission that is compatible with therapeutic interventions. mGluRs are a family of eight G-protein coupled receptors classified into three groups (I-III) based on sequence homology, transduction mechanisms, and pharmacology. Activation of group II and III or inhibition of group I represents a strategy to counteract the glutamatergic hyperactivity associated with diabetic neuropathy. In this review article, we will discuss the role of glutamate receptors in the release of insulin and the development/treatment of diabetic neuropathy, with particular emphasis on their manipulation to prevent the glutamatergic hyperactivity associated with diabetic neuropathy.
Collapse
Affiliation(s)
- Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", via Costantinopoli 16, 80138 Naples, Italy.
| | - Ida Marabese
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", via Costantinopoli 16, 80138 Naples, Italy
| | - Federica Ricciardi
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", via Costantinopoli 16, 80138 Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", via Costantinopoli 16, 80138 Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", via Costantinopoli 16, 80138 Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
3
|
Bohovyk R, Kravtsova O, Levchenko V, Klemens CA, Palygin O, Staruschenko A. Effects of zinc in podocytes and cortical collecting duct in vitro and Dahl salt-sensitive rats in vivo. J Biol Chem 2024; 300:107781. [PMID: 39276935 PMCID: PMC11736004 DOI: 10.1016/j.jbc.2024.107781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
Zinc is one of the essential divalent cations in the human body and a fundamental microelement involved in the regulation of many cellular and subcellular functions. Experimental studies reported that zinc deficiency is associated with renal damage and could increase blood pressure. It was proposed that zinc dietary supplementation plays a renoprotective role. Our study aimed to investigate the effects of zinc on intracellular signaling in renal cells and explore the correlation between dietary zinc and the progression of salt-induced hypertension. The impact of extracellular zinc concentrations on two different kidney epithelial cell types, podocytes and principal cells of the cortical collecting duct (CCD), was tested. In podocytes, a rise in extracellular zinc promotes TRPC6 channel-mediated calcium entry but not altered intracellular zinc levels. However, we observe the opposite effect in CCD cells with no alteration in calcium levels and steady-state elevation in intracellular zinc. Moreover, prolonged extracellular zinc exposure leads to cytotoxic insults in CCD cells but not in podocytes, characterized by increased cell death and disrupted cytoskeletal organization. Next, we tested if dietary zinc plays a role in the development of hypertension in Dahl salt-sensitive rats. Neither zinc-rich nor deficient diets impact the regular development of salt-sensitive hypertension. These results suggest specialized roles for zinc in renal function, implicating its involvement in proliferation and apoptosis in CCD cells and calcium signaling and cytoskeletal dynamics modulation in podocytes. Further research is required to elucidate the detailed mechanisms of zinc action and its implications in renal health and disease.
Collapse
Affiliation(s)
- Ruslan Bohovyk
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Olha Kravtsova
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| | - Christine A Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA; Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida, USA
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA; Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida, USA; James A. Haley Veterans' Hospital, Tampa, Florida, USA.
| |
Collapse
|
4
|
Ma R, Tao Y, Wade ML, Mallet RT. Non-voltage-gated Ca 2+ channel signaling in glomerular cells in kidney health and disease. Am J Physiol Renal Physiol 2024; 327:F249-F264. [PMID: 38867675 PMCID: PMC11460346 DOI: 10.1152/ajprenal.00130.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
Positioned at the head of the nephron, the renal corpuscle generates a plasma ultrafiltrate to initiate urine formation. Three major cell types within the renal corpuscle, the glomerular mesangial cells, podocytes, and glomerular capillary endothelial cells, communicate via endocrine- and paracrine-signaling mechanisms to maintain the structure and function of the glomerular capillary network and filtration barrier. Ca2+ signaling mediated by several distinct plasma membrane Ca2+ channels impacts the functions of all three cell types. The past two decades have witnessed pivotal advances in understanding of non-voltage-gated Ca2+ channel function and regulation in the renal corpuscle in health and renal disease. This review summarizes the current knowledge of the physiological and pathological impact of non-voltage-gated Ca2+ channel signaling in mesangial cells, podocytes and glomerular capillary endothelium. The main focus is on transient receptor potential and store-operated Ca2+ channels, but ionotropic N-methyl-d-aspartate receptors and purinergic receptors also are discussed. This update of Ca2+ channel functions and their cellular signaling cascades in the renal corpuscle is intended to inform the development of therapeutic strategies targeting these channels to treat kidney diseases, particularly diabetic nephropathy.
Collapse
Affiliation(s)
- Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Yu Tao
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Michael L Wade
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Robert T Mallet
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
5
|
Li C, Szeto CC. Urinary podocyte markers in diabetic kidney disease. Kidney Res Clin Pract 2024; 43:274-286. [PMID: 38325865 PMCID: PMC11181047 DOI: 10.23876/j.krcp.23.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/30/2023] [Accepted: 10/30/2023] [Indexed: 02/09/2024] Open
Abstract
Podocytes are involved in maintaining kidney function and are a major focus of research on diabetic kidney disease (DKD). Urinary biomarkers derived from podocyte fragments and molecules have been proposed for the diagnosis and monitoring of DKD. Various methods have been used to detect intact podocytes and podocyte-derived microvesicles in urine, including centrifugation, visualization, and molecular quantification. Quantification of podocyte-specific protein targets and messenger RNA levels can be performed by Western blotting or enzyme-linked immunosorbent assay and quantitative polymerase chain reaction, respectively. At present, many of these techniques are expensive and labor-intensive, all limiting their widespread use in routine clinical tests. While the potential of urinary podocyte markers for monitoring and risk stratification of DKD has been explored, systematic studies and external validation are lacking in the current literature. Standardization and automation of laboratory methods should be a priority for future research, and the added value of these methods to routine clinical tests should be defined.
Collapse
Affiliation(s)
- Chuanlei Li
- Carol & Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Cheuk-Chun Szeto
- Carol & Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
6
|
Chou X, Li X, Ma K, Shen Y, Min Z, Xiao W, Zhang J, Wu Q, Sun D. N-methyl-d-aspartate receptor 1 activation mediates cadmium-induced epithelial-mesenchymal transition in proximal tubular cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 904:166955. [PMID: 37704144 DOI: 10.1016/j.scitotenv.2023.166955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
Cadmium (Cd) is a commonly found environmental pollutant and is known to damage multiple organs with kidneys being the most common one. N-methyl-d-aspartate receptor 1 (NMDAR1) is a ligand-gated ion channel that is highly permeable to calcium ion (Ca2+). Because Cd2+ and Ca2+ have structural and physicochemical similarities, whether and how Cd could interfere NMDAR1 function to cause renal epithelial cells dysfunction remains unknown. In this study, we investigated the role of NMDAR1 in Cd-induced renal damage and found that Cd treatment upregulated NMDAR1 expression and promoted epithelial-mesenchymal transition (EMT) in mouse kidneys in vivo and human proximal tubular epithelial HK-2 cells in vitro, which were accompanied with activation of the inositol-requiring enzyme 1 (IRE-1α) / spliced X box binding protein-1 (XBP-1s) pathway, an indicative of endoplasmic reticulum (ER) stress. Mechanistically, NMDAR1 upregulation by Cd promoted Ca2+ channel opening and Ca2+ influx, resulting in ER stress and subsequently EMT in HK-2 cells. Inhibition of NMDAR1 by pharmacological antagonist MK-801 significantly attenuated Cd-induced Ca2+ influx, ER stress, and EMT. Pretreatment with the IRE-1α/XBP-1s pathway inhibitor STF-083010 also restored the epithelial phenotype of Cd-treated HK-2 cells. Therefore, our findings suggest that NMDAR1 activation mediates Cd-induced EMT in proximal epithelial cells likely through the IRE-1α/XBP-1s pathway, supporting the idea that NMDAR1 could be a potential therapeutic target for Cd-induced renal damage.
Collapse
Affiliation(s)
- Xin Chou
- Department of Occupational Disease, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Xiaohu Li
- Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430022, China
| | - Kunpeng Ma
- Department of Occupational Disease, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Yue Shen
- Department of Occupational Disease, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Zhen Min
- Department of Occupational Disease, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Wusheng Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, School of Public Health, Peking University, Beijing 100191, China
| | - Jingbo Zhang
- Department of Occupational Disease, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China
| | - Qing Wu
- Department of Toxicology, School of Public Health, Fudan University, 130 Dong'an Road, Shanghai 200032, China
| | - Daoyuan Sun
- Department of Occupational Disease, Shanghai Pulmonary Hospital affiliated to Tongji University, Shanghai 200433, China.
| |
Collapse
|
7
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
8
|
Pang Q, Chen H, Wu H, Wang Y, An C, Lai S, Xu J, Wang R, Zhou J, Xiao H. N6-methyladenosine regulators-related immune genes enable predict graft loss and discriminate T-cell mediate rejection in kidney transplantation biopsies for cause. Front Immunol 2022; 13:1039013. [PMID: 36483557 PMCID: PMC9722771 DOI: 10.3389/fimmu.2022.1039013] [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: 09/07/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
Objective The role of m6A modification in kidney transplant-associated immunity, especially in alloimmunity, still remains unknown. This study aims to explore the potential value of m6A-related immune genes in predicting graft loss and diagnosing T cell mediated rejection (TCMR), as well as the possible role they play in renal graft dysfunction. Methods Renal transplant-related cohorts and transcript expression data were obtained from the GEO database. First, we conducted correlation analysis in the discovery cohort to identify the m6A-related immune genes. Then, lasso regression and random forest were used respectively to build prediction models in the prognosis and diagnosis cohort, to predict graft loss and discriminate TCMR in dysfunctional renal grafts. Connectivity map (CMap) analysis was applied to identify potential therapeutic compounds for TCMR. Results The prognostic prediction model effectively predicts the prognosis and survival of renal grafts with clinical indications (P< 0.001) and applies to both rejection and non-rejection situations. The diagnostic prediction model discriminates TCMR in dysfunctional renal grafts with high accuracy (area under curve = 0.891). Meanwhile, the classifier score of the diagnostic model, as a continuity index, is positively correlated with the severity of main pathological injuries of TCMR. Furthermore, it is found that METTL3, FTO, WATP, and RBM15 are likely to play a pivotal part in the regulation of immune response in TCMR. By CMap analysis, several small molecular compounds are found to be able to reverse TCMR including fenoldopam, dextromethorphan, and so on. Conclusions Together, our findings explore the value of m6A-related immune genes in predicting the prognosis of renal grafts and diagnosis of TCMR.
Collapse
Affiliation(s)
- Qidan Pang
- Department of Nephrology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Hang Wu
- Department of Nephrology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Wang
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Changyong An
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Suhe Lai
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Jia Xu
- Department of Nephrology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Ruiqiong Wang
- Department of Nephrology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Juan Zhou
- Department of Nephrology, Bishan Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Hanyu Xiao, ; Juan Zhou,
| | - Hanyu Xiao
- Department of General Surgery/Gastrointestinal Surgery, Bishan Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Hanyu Xiao, ; Juan Zhou,
| |
Collapse
|
9
|
Grochowalska K, Pikul P, Piwkowska A. Insights into the regulation of podocyte and glomerular function by lactate and its metabolic sensor G-protein-coupled receptor 81. J Cell Physiol 2022; 237:4097-4111. [PMID: 36084306 DOI: 10.1002/jcp.30874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 11/10/2022]
Abstract
Podocytes and their foot processes are an important cellular layer of the renal filtration barrier that is involved in regulating glomerular permeability. Disturbances of podocyte function play a central role in the development of proteinuria in diabetic nephropathy. The retraction and effacement of podocyte foot processes that form slit diaphragms are a common feature of proteinuria. Correlations between the retraction of foot processes and the development of proteinuria are not well understood. Unraveling peculiarities of podocyte energy metabolism notably under diabetic conditions will provide insights into the pathogenesis of diabetic nephropathy. Intracellular metabolism in the cortical area of podocytes is regulated by glycolysis, whereas energy balance in the central area is controlled by oxidative phosphorylation and glycolysis. High glucose concentrations were recently reported to force podocytes to switch from mitochondrial oxidative phosphorylation to glycolysis, resulting in lactic acidosis. In this review, we hypothesize that the lactate receptor G-protein-coupled receptor 81 (also known as hydroxycarboxylic acid receptor 81) may contribute to the control of podocyte function in both health and disease.
Collapse
Affiliation(s)
- Klaudia Grochowalska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | - Piotr Pikul
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland
| | - Agnieszka Piwkowska
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Gdansk, Poland.,Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| |
Collapse
|
10
|
Saeedi M, Mehranfar F. Challenges and approaches of drugs such as Memantine, Donepezil, Rivastigmine and Aducanumab in the treatment, control and management of Alzheimer's disease. Recent Pat Biotechnol 2022; 16:102-121. [PMID: 35236274 DOI: 10.2174/1872208316666220302115901] [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: 08/25/2021] [Revised: 10/20/2021] [Accepted: 12/28/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a kinds of neuropsychiatric illnesses that affect the central nervous system. In this disease, the accumulation of amyloid-beta increases, and phosphorylated tau (P-tau) protein, one of the ways to treat this disease is to reduce the accumulation of amyloid-beta. Various studies have demonstrated that pharmacological approaches have considerable effects in the treatment of AD, despite the side effects and challenges. Cholinesterase inhibitors and the NMDA receptor antagonist memantine are presently authorized therapies for AD. Memantine and Donepezil are the most common drugs for the prevention and therapy of AD with mechanisms such as lessened β-amyloid plaque, effect on N-Methyl-D-aspartate (NMDA) receptors. Diminution glutamate and elevated acetylcholine are some of the influences of medications administrated to treat AD, and drugs can also play a role in slowing the progression of cognitive and memory impairment. A new pharmacological approach and strategy is required to control the future of AD. This review appraises the effects of memantine, donepezil, rivastigmine, and aducanumab in clinical trials, in vitro and animal model studies that have explored how these drugs versus AD development and also discuss possible mechanisms of influence on the brain. Research in clinical trials has substantial findings that support the role of these medications in AD treatment and ameliorate the safety and efficacy of AD therapy, although more clinical trials are required to prove their effectiveness.
Collapse
Affiliation(s)
- Mohammad Saeedi
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Mehranfar
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
11
|
Ismail CAN, Ghazali AK, Suppian R, Abd Aziz CB, Long I. Lower Formalin-Induced Pain Responses in Painless Diabetic Neuropathy Rat Correlate with the Reduced Spinal Cord NR2B Subunit of N-Methyl-D-Aspartate Receptor Activation. J Mol Neurosci 2021; 72:598-609. [PMID: 34727325 DOI: 10.1007/s12031-021-01929-0] [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: 12/24/2020] [Accepted: 10/11/2021] [Indexed: 11/30/2022]
Abstract
Diabetic neuropathy (DN) is a late complication of diabetic mellitus and may rise into painful and painless variants. Limited studies have looked at nociceptive mechanisms of painless DN variant. The study aimed to determine phosphorylation and total NR2B subunit of N-methyl-D-aspartate receptor in the spinal cord of painless DN rat during early phase following formalin injection. Thirty-six Sprague-Dawley male rats were randomly assigned into three groups: control, painful, and painless DN (n = 12). The rats were developed into the early phase of DN for 2 weeks following diabetic induction. Two weeks later, the rats were injected with 5% formalin solution and flinching and licking responses were recorded for 60 min. The rats were sacrificed 3 days later, and the spinal cord enlargement region was collected. Immunohistochemistry and Western blot procedures were conducted to determine the phosphorylated and total NR2B subunit expressions. The results showed reduced flinching and licking responses in painless DN rats compared to control and painful DN groups, followed by a significant reduction in phosphorylated and total NR2B expression at both ipsilateral and contralateral regions of the spinal cord. In conclusion, reduced pain behavior responses in painless DN rats following formalin injection is possibly contributed by the reduced expression of phosphorylated and total NR2B subunit in the spinal cord.
Collapse
Affiliation(s)
- Che Aishah Nazariah Ismail
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, 16150, Kelantan, Malaysia.
- Brain and Behaviour Clusters, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, 16150, Kelantan, Malaysia.
| | - Anis Kausar Ghazali
- Biostatistics and Research Methodology Unit, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, 16150, Kelantan, Malaysia
| | - Rapeah Suppian
- School of Health Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, 16150, Kelantan, Malaysia
| | - Che Badariah Abd Aziz
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, 16150, Kelantan, Malaysia
| | - Idris Long
- School of Health Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian, 16150, Kelantan, Malaysia
| |
Collapse
|
12
|
Scholz O, Otter S, Welters A, Wörmeyer L, Dolenšek J, Klemen MS, Pohorec V, Eberhard D, Mrugala J, Hamacher A, Koch A, Sanz M, Hoffmann T, Hogeback J, Herebian D, Klöcker N, Piechot A, Mayatepek E, Meissner T, Stožer A, Lammert E. Peripherally active dextromethorphan derivatives lower blood glucose levels by targeting pancreatic islets. Cell Chem Biol 2021; 28:1474-1488.e7. [PMID: 34118188 DOI: 10.1016/j.chembiol.2021.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/09/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022]
Abstract
Dextromethorphan (DXM) acts as cough suppressant via its central action. Cell-protective effects of this drug have been reported in peripheral tissues, making DXM potentially useful for treatment of several common human diseases, such as type 2 diabetes mellitus (T2DM). Pancreatic islets are among the peripheral tissues that positively respond to DXM, and anti-diabetic effects of DXM were observed in two placebo-controlled, randomized clinical trials in humans with T2DM. Since these effects were associated with central side effects, we here developed chemical derivatives of DXM that pass the blood-brain barrier to a significantly lower extent than the original drug. We show that basic nitrogen-containing residues block central adverse events of DXM without reducing its anti-diabetic effects, including the protection of human pancreatic islets from cell death. These results show how to chemically modify DXM, and possibly other morphinans, as to exclude central side effects, while targeting peripheral tissues, such as pancreatic islets.
Collapse
Affiliation(s)
- Okka Scholz
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Silke Otter
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany
| | - Alena Welters
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Laura Wörmeyer
- Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; Faculty of Natural Sciences and Mathematics, University of Maribor, Koroška cesta 160, 2000 Maribor, Slovenia
| | - Maša Skelin Klemen
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Viljem Pohorec
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Daniel Eberhard
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Jessica Mrugala
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Anna Hamacher
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Angela Koch
- Institute of Neuro- and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Miguel Sanz
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Torsten Hoffmann
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Jens Hogeback
- A&M Labor für Analytik und Metabolismusforschung Service GmbH, 50126 Bergheim, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Nikolaj Klöcker
- Institute of Neuro- and Sensory Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Alexander Piechot
- Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; Taros Chemicals GmbH & Co. KG, 44227 Dortmund, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Eckhard Lammert
- Institute for Vascular and Islet Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, 40225 Düsseldorf, Germany; Institute of Metabolic Physiology, Heinrich Heine University, 40225 Düsseldorf, Germany; Center of Competence for Innovative Diabetes Therapy (KomIT), German Diabetes Center (DDZ), 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD e.V.), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
| |
Collapse
|
13
|
Yeung JHY, Walby JL, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A. Glutamatergic receptor expression changes in the Alzheimer's disease hippocampus and entorhinal cortex. Brain Pathol 2021; 31:e13005. [PMID: 34269494 PMCID: PMC8549033 DOI: 10.1111/bpa.13005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's Disease (AD) is the leading form of dementia worldwide. Currently, the pathological mechanisms underlying AD are not well understood. Although the glutamatergic system is extensively implicated in its pathophysiology, there is a gap in knowledge regarding the expression of glutamate receptors in the AD brain. This study aimed to characterize the expression of specific glutamate receptor subunits in post‐mortem human brain tissue using immunohistochemistry and confocal microscopy. Free‐floating immunohistochemistry and confocal laser scanning microscopy were used to quantify the density of glutamate receptor subunits GluA2, GluN1, and GluN2A in specific cell layers of the hippocampal sub‐regions, subiculum, entorhinal cortex, and superior temporal gyrus. Quantification of GluA2 expression in human post‐mortem hippocampus revealed a significant increase in the stratum (str.) moleculare of the dentate gyrus (DG) in AD compared with control. Increased GluN1 receptor expression was found in the str. moleculare and hilus of the DG, str. oriens of the CA2 and CA3, str. pyramidale of the CA2, and str. radiatum of the CA1, CA2, and CA3 subregions and the entorhinal cortex. GluN2A expression was significantly increased in AD compared with control in the str. oriens, str. pyramidale, and str. radiatum of the CA1 subregion. These findings indicate that the expression of glutamatergic receptor subunits shows brain region‐specific changes in AD, suggesting possible pathological receptor functioning. These results provide evidence of specific glutamatergic receptor subunit changes in the AD hippocampus and entorhinal cortex, indicating the requirement for further research to elucidate the pathophysiological mechanisms it entails, and further highlight the potential of glutamatergic receptor subunits as therapeutic targets.
Collapse
Affiliation(s)
- Jason H Y Yeung
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Joshua L Walby
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Therapeutic Potential of Polyphenols in the Management of Diabetic Neuropathy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9940169. [PMID: 34093722 PMCID: PMC8137294 DOI: 10.1155/2021/9940169] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
Diabetic neuropathy (DN) is a common and serious diabetes-associated complication that primarily takes place because of neuronal dysfunction in patients with diabetes. Use of current therapeutic agents in DN treatment is quite challenging because of their severe adverse effects. Therefore, there is an increased need of identifying new safe and effective therapeutic agents. DN complications are associated with poor glycemic control and metabolic imbalances, primarily oxidative stress (OS) and inflammation. Various mediators and signaling pathways such as glutamate pathway, activation of channels, trophic factors, inflammation, OS, advanced glycation end products, and polyol pathway have a significant contribution to the progression and pathogenesis of DN. It has been indicated that polyphenols have the potential to affect DN pathogenesis and could be used as potential alternative therapy. Several polyphenols including kolaviron, resveratrol, naringenin, quercetin, kaempferol, and curcumin have been administered in patients with DN. Furthermore, chlorogenic acid can provide protection against glutamate neurotoxicity via its hydrolysate, caffeoyl acid group, and caffeic acid through regulating the entry of calcium into neurons. Epigallocatechin-3-gallate treatment can protect motor neurons by regulating the glutamate level. It has been demonstrated that these polyphenols can be promising in combating DN-associated damaging pathways. In this article, we have summarized DN-associated metabolic pathways and clinical manifestations. Finally, we have also focused on the roles of polyphenols in the treatment of DN.
Collapse
|
15
|
Rapid effects of neurosteroids on neuronal plasticity and their physiological and pathological implications. Neurosci Lett 2021; 750:135771. [PMID: 33636284 DOI: 10.1016/j.neulet.2021.135771] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 11/22/2022]
Abstract
Current neuroscience research on neurosteroids and their synthetic analogues - neuroactive steroids - clearly demonstrate their drug likeness in a variety of neurological and psychiatric conditions. Moreover, research on neurosteroids continues to provide novel mechanistic insights into receptor activation or inhibition of various receptors. This mini-review will provide a high-level overview of the research area and discuss the various classes of potential physiological and pathological implications discovered so far.
Collapse
|
16
|
Zhou J, Liu S, Guo L, Wang R, Chen J, Shen J. NMDA receptor-mediated CaMKII/ERK activation contributes to renal fibrosis. BMC Nephrol 2020; 21:392. [PMID: 32907546 PMCID: PMC7488001 DOI: 10.1186/s12882-020-02050-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 08/30/2020] [Indexed: 02/08/2023] Open
Abstract
Background This study aimed to understand the mechanistic role of N-methyl-D-aspartate receptor (NMDAR) in acute fibrogenesis using models of in vivo ureter obstruction and in vitro TGF-β administration. Methods Acute renal fibrosis (RF) was induced in mice by unilateral ureteral obstruction (UUO). Histological changes were observed using Masson’s trichrome staining. The expression levels of NR1, which is the functional subunit of NMDAR, and fibrotic and epithelial-to-mesenchymal transition markers were measured by immunohistochemical and Western blot analysis. HK-2 cells were incubated with TGF-β, and NMDAR antagonist MK-801 and Ca2+/calmodulin-dependent protein kinase II (CaMKII) antagonist KN-93 were administered for pathway determination. Chronic RF was introduced by sublethal ischemia–reperfusion injury in mice, and NMDAR inhibitor dextromethorphan hydrobromide (DXM) was administered orally. Results The expression of NR1 was upregulated in obstructed kidneys, while NR1 knockdown significantly reduced both interstitial volume expansion and the changes in the expression of α-smooth muscle actin, S100A4, fibronectin, COL1A1, Snail, and E-cadherin in acute RF. TGF-β1 treatment increased the elongation phenotype of HK-2 cells and the expression of membrane-located NR1 and phosphorylated CaMKII and extracellular signal–regulated kinase (ERK). MK801 and KN93 reduced CaMKII and ERK phosphorylation levels, while MK801, but not KN93, reduced the membrane NR1 signal. The levels of phosphorylated CaMKII and ERK also increased in kidneys with obstruction but were decreased by NR1 knockdown. The 4-week administration of DXM preserved renal cortex volume in kidneys with moderate ischemic–reperfusion injury. Conclusions NMDAR participates in both acute and chronic renal fibrogenesis potentially via CaMKII-induced ERK activation.
Collapse
Affiliation(s)
- Jingyi Zhou
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Shuaihui Liu
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Luying Guo
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Rending Wang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China. .,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China. .,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China. .,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China.
| | - Jia Shen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China. .,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China. .,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China. .,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China.
| |
Collapse
|
17
|
Glutamate-Gated NMDA Receptors: Insights into the Function and Signaling in the Kidney. Biomolecules 2020; 10:biom10071051. [PMID: 32679780 PMCID: PMC7407907 DOI: 10.3390/biom10071051] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022] Open
Abstract
N-Methyl-d-aspartate receptor (NMDAR) is a glutamate-gated ionotropic receptor that intervenes in most of the excitatory synaptic transmission within the central nervous system (CNS). Aside from being broadly distributed in the CNS and having indispensable functions in the brain, NMDAR has predominant roles in many physiological and pathological processes in a wide range of non-neuronal cells and tissues. The present review outlines current knowledge and understanding of the physiological and pathophysiological functions of NMDAR in the kidney, an essential excretory and endocrine organ responsible for the whole-body homeostasis. The review also explores the recent findings regarding signaling pathways involved in NMDAR-mediated responses in the kidney. As established from diverse lines of research reviewed here, basal levels of receptor activation within the kidney are essential for the maintenance of healthy tubular and glomerular function, while a disproportionate activation can lead to a disruption of NMDAR's downstream signaling pathways and a myriad of pathophysiological consequences.
Collapse
|
18
|
Naseri R, Farzaei F, Fakhri S, El-Senduny FF, Altouhamy M, Bahramsoltani R, Ebrahimi F, Rahimi R, Farzaei MH. Polyphenols for diabetes associated neuropathy: Pharmacological targets and clinical perspective. Daru 2019; 27:781-798. [PMID: 31352568 PMCID: PMC6895369 DOI: 10.1007/s40199-019-00289-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/01/2019] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Diabetic neuropathy (DNP) is a widespread and debilitating complication with complex pathophysiology that is caused by neuronal dysfunction in diabetic patients. Conventional therapeutics for DNP are quite challenging due to their serious adverse effects. Hence, there is a need to investigate novel effective and safe options. The novelty of the present study was to provide available therapeutic approaches, emerging molecular mechanisms, signaling pathways and future directions of DNP as well as polyphenols' effect, which accordingly, give new insights for paving the way for novel treatments in DNP. EVIDENCE ACQUISITION A comprehensive review was done in electronic databases including Medline, PubMed, Web of Science, Scopus, national database (Irandoc and SID), and related articles regarding metabolic pathways on the pathogenesis of DNP as well as the polyphenols' effect. The keywords "diabetic neuropathy" and "diabetes mellitus" in the title/abstract and "polyphenol" in the whole text were used. Data were collected from inception until May 2019. RESULTS DNP complications is mostly related to a poor glycemic control and metabolic imbalances mainly inflammation and oxidative stress. Several signaling and molecular pathways play key roles in the pathogenesis and progression of DNP. Among natural entities, polyphenols are suggested as multi-target alternatives affecting most of these pathogenesis mechanisms in DNP. CONCLUSION The findings revealed novel pathogenicity signaling pathways of DNP and affirmed the auspicious role of polyphenols to tackle these destructive pathways in order to prevent, manage, and treat various diseases. Graphical Abstract .
Collapse
Affiliation(s)
- Rozita Naseri
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fardous F El-Senduny
- Biochemistry division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Miram Altouhamy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Roodabeh Bahramsoltani
- Department of Pharmacy in Persian Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farnaz Ebrahimi
- Pharmacy students` research committee, School of pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roja Rahimi
- Department of Pharmacy in Persian Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Hassanzadeh Khayyat N, Kim EY, Dryer SE. TRPC6 inactivation does not protect against diabetic kidney disease in streptozotocin (STZ)-treated Sprague-Dawley rats. FASEB Bioadv 2019; 1:773-782. [PMID: 32123821 PMCID: PMC6996301 DOI: 10.1096/fba.2019-00077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/06/2019] [Accepted: 10/30/2019] [Indexed: 01/14/2023] Open
Abstract
Canonical transient receptor potential-6 (TRPC6) channels have been implicated in the progression of several forms of kidney disease (1). While there is strong evidence that glomerular TRPC6 channels are dysregulated in diabetic nephropathy (DN), there is no consensus as to whether deletion or inactivation of TRPC6 is protective in animal models of DN. A previous study in Dahl salt-sensitive rats suggests that TRPC6 knockout has a modest protective effect in streptozotocin (STZ)-induced DN (2). In the present study, we examined whether inactivation of TRPC6 channels by CRISPR/Cas9 editing (Trpc6 del/del rats) affects progression of STZ-induced DN in Sprague-Dawley rats. Wild-type littermates (Trpc6 wt/wt rats) were used as controls. We observed that a single injection of STZ resulted in severe hyperglycemia that was sustained over a 10-week period, accompanied by a marked reduction in circulating C-peptide, dyslipidemia, and failure to gain weight compared to vehicle-treated animals. Those effects were equally severe in Trpc6 wt/wt and Trpc6 del/del rats. STZ treatment resulted in increased urine albumin excretion at 4, 8, and 10 weeks after injection, and this effect was equally severe in Trpc6 wt/wt and Trpc6 del/del rats. TRPC6 inactivation had no effect on blood urea nitrogen (BUN), plasma creatinine concentration, urine nephrin excretion, or kidney weight:body weight ratio measured 10 weeks after STZ injection. STZ treatment evoked modest and equivalent mesangial expansion in Trpc6 wt/wt and Trpc6 del/del rats. In summary, we observed no protective effect of TRPC6 inactivation on STZ-induced DN in rats on the Sprague-Dawley background.
Collapse
Affiliation(s)
| | - Eun Young Kim
- Department of Biology and BiochemistryUniversity of HoustonHoustonTXUSA
| | - Stuart E. Dryer
- Department of Biology and BiochemistryUniversity of HoustonHoustonTXUSA
- Department of Biomedical SciencesUniversity of Houston College of MedicineHoustonTXUSA
| |
Collapse
|
20
|
Papu John AS, Kundu S, Pushpakumar S, Amin M, Tyagi SC, Sen U. Hydrogen sulfide inhibits Ca 2+-induced mitochondrial permeability transition pore opening in type-1 diabetes. Am J Physiol Endocrinol Metab 2019; 317:E269-E283. [PMID: 31039005 PMCID: PMC6732471 DOI: 10.1152/ajpendo.00251.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/29/2022]
Abstract
Hydrogen sulfide (H2S) attenuates N-methyl-d-aspartate receptor-R1 (NMDA-R1) and mitigates diabetic renal damage; however, the molecular mechanism is not well known. Whereas NMDA-R1 facilitates Ca2+ permeability, H2S is known to inhibit L-type Ca2+ channel. High Ca2+ activates cyclophilin D (CypD), a gatekeeper protein of mitochondrial permeability transition pore (MPTP), thus facilitating molecular exchange between matrix and cytoplasm causing oxidative outburst and cell death. We tested the hypothesis of whether NMDA-R1 mediates Ca2+ influx causing CypD activation and MPTP opening leading to oxidative stress and renal injury in diabetes. We also tested whether H2S treatment blocks Ca2+ channel and thus inhibits CypD and MPTP opening to prevent renal damage. C57BL/6J and Akita (C57BL/6J-Ins2Akita) mice were treated without or with H2S donor GYY4137 (0.25 mg·kg-1·day-1 ip) for 8 wk. In vitro studies were performed using mouse glomerular endothelial cells. Results indicated that low levels of H2S and increased expression of NMDA-R1 in diabetes induced Ca2+ permeability, which was ameliorated by H2S treatment. We observed cytosolic Ca2+ influx in hyperglycemic (HG) condition along with mitochondrial-CypD activation, increased MPTP opening, and oxidative outburst, which were mitigated with H2S treatment. Renal injury biomarker KIM-1 was upregulated in HG conditions and normalized following H2S treatment. Inhibition of NMDA-R1 by pharmacological blocker MK-801 revealed similar results. We conclude that NMDA-R1-mediated Ca2+ influx in diabetes induces MPTP opening via CypD activation leading to increased oxidative stress and renal injury, and H2S protects diabetic kidney from injury by blocking mitochondrial Ca2+ permeability through NMDA-R1 pathway.
Collapse
Affiliation(s)
- A Sashi Papu John
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Sourav Kundu
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Matthew Amin
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine , Louisville, Kentucky
| |
Collapse
|
21
|
Trpc6 inactivation confers protection in a model of severe nephrosis in rats. J Mol Med (Berl) 2018; 96:631-644. [PMID: 29785489 PMCID: PMC6015123 DOI: 10.1007/s00109-018-1648-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/25/2018] [Accepted: 05/07/2018] [Indexed: 12/11/2022]
Abstract
Abstract Mutations in canonical transient receptor potential-6 (TRPC6) channels give rise to rare familial forms of focal and segmental glomerulosclerosis (FSGS). Here we examined a possible role for TRPC6 in the progression of chronic puromycin aminonucleoside (PAN) nephrosis in Sprague-Dawley rats, a classic model of acquired nephrotic syndromes. We used CRISPR/Cas9 technology to delete a 239-bp region within exon 2 of the Trpc6 gene (Trpc6del allele). Trpc6del/del rats expressed detectable Trpc6 transcripts missing exon 2, and TRPC6 proteins could be detected by immunoblot of renal cortex. However, the abundance of Trpc6 transcripts and TRPC6 protein in renal cortex was much lower than in Trpc6wt/wt littermates, and functional TRPC6 channels could not be detected in whole-cell recordings from glomerular cells cultured from Trpc6del/del animals, possibly because of disruption of ankyrin repeats 1 and 2. During the chronic phase of PAN nephrosis, Trpc6del/del rats had reduced urine albumin excretion, reduced serum cholesterol and triglycerides, and improved azotemia compared to wild-type Trpc6wt/wt littermates. Glomerulosclerosis was severe during chronic PAN nephrosis in Trpc6wt/wt rats but was markedly reduced in Trpc6del/del littermates. Trpc6del/del animals also had less severe tubulointerstitial fibrosis as assessed by several biochemical and histological analyses, as well as reduced foot process effacement and glomerular basement thickening compared to Trpc6wtt/wt controls. None of the manipulations in this study affected the abundance of TRPC5 channels in renal cortex. TRPC3 was increased in PAN nephrosis and in Trpc6del/del rats. These data support a role for TRPC6 channels in driving an acquired form of secondary FSGS. Key messages We examined aminonucleoside nephrosis in rats with wild type and inactivated TRPC6. TRPC6 channels were inactivated by CRISPR/Cas9 editing of the Trpc6 gene. TRPC6 inactivation reduced albuminuria in the chronic but not the acute phase. TRPC6 inactivation reduced glomerulosclerosis and ultrastructural changes. TRPC6 inactivation also reduced interstitial changes and renal fibrosis.
Electronic supplementary material The online version of this article (10.1007/s00109-018-1648-3) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Ettcheto M, Sánchez-López E, Gómez-Mínguez Y, Cabrera H, Busquets O, Beas-Zarate C, García ML, Carro E, Casadesus G, Auladell C, Vázquez Carrera M, Folch J, Camins A. Peripheral and Central Effects of Memantine in a Mixed Preclinical Mice Model of Obesity and Familial Alzheimer's Disease. Mol Neurobiol 2018; 55:7327-7339. [PMID: 29404958 DOI: 10.1007/s12035-018-0868-4] [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] [Received: 07/10/2017] [Accepted: 01/07/2018] [Indexed: 01/01/2023]
Abstract
There is growing evidence that obesity associated with type 2 diabetes mellitus (T2DM) and aging are risk factors for the development of Alzheimer's disease (AD). However, the molecular mechanisms through which obesity interacts with β-amyloid (Aβ) to promote cognitive decline remains poorly understood. Memantine (MEM), a N-methyl-D-aspartate receptor antagonist, is currently used for the treatment of AD. Nonetheless, few studies have reported its effects on genetic preclinical models of this neurodegenerative disease exacerbated with high-fat diet (HFD)-induced obesity. Therefore, the present research aims to elucidate the effects of MEM on familial AD HFD-induced insulin resistance and learning and memory impairment. Furthermore, it aspires to determine the possible underlying mechanisms that connect AD to T2DM. Wild type and APPswe/PS1dE9 mice were used in this study. The animals were fed with either chow or HFD until 6 months of age, and they were treated with MEM-supplemented water (30 mg/kg) during the last 12 weeks. Our study demonstrates that MEM improves the metabolic consequences produced by HFD in this model of familial AD. Behavioural assessments confirmed that the treatment also improves animals learning abilities and decreases memory loss. Moreover, MEM treatment improves brain insulin signalling upregulating AKT, as well as cyclic adenosine monophosphate response element binding (CREB) expression, and modulates the amyloidogenic pathway, which, in turn, reduced the accumulation of Aβ. Moreover, this drug increases the activation of molecules involved with insulin signalling in the liver, such as insulin receptor substrate 2 (IRS2), which is a key protein regulating hepatic resistance to insulin. These results provide new insight into the role of MEM not only in the occurrence of AD treatment, but also in its potential application on peripheral metabolic disorders where Aβ plays a key role, as is the case of T2DM.
Collapse
Affiliation(s)
- Miren Ettcheto
- Departament de Farmacología, Toxicologia i Quimica Terapéutica, Unitat de Farmacologia i Farmacognosia, Facultat de Farmacia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII s/n, 08028, Barcelona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Unitat de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Institut de Neurociencias, Universitat de Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Unitat de Farmacia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Yaiza Gómez-Mínguez
- Departament de Farmacología, Toxicologia i Quimica Terapéutica, Unitat de Farmacologia i Farmacognosia, Facultat de Farmacia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII s/n, 08028, Barcelona, Spain
| | - Henrry Cabrera
- Departament de Farmacología, Toxicologia i Quimica Terapéutica, Unitat de Farmacologia i Farmacognosia, Facultat de Farmacia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII s/n, 08028, Barcelona, Spain
| | - Oriol Busquets
- Departament de Farmacología, Toxicologia i Quimica Terapéutica, Unitat de Farmacologia i Farmacognosia, Facultat de Farmacia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII s/n, 08028, Barcelona, Spain.,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Unitat de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain.,Institut de Neurociencias, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Departamento de Biología Celular y Molecular, C.U.C.B.A, Universidad de Guadalajara and División de Neurociencias, Sierra Mojada 800, Col. Independencia, 44340, Guadalajara, Jalisco, Mexico
| | - Maria Luisa García
- Unitat de Farmacia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Eva Carro
- Neurodegenerative Disorders Group, Instituto de Investigacion Hospital 12 de Octubre (i + 12), Madrid, Spain
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Carme Auladell
- Departament de Biologia Cel·lular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Manuel Vázquez Carrera
- Departament de Farmacología, Toxicologia i Quimica Terapéutica, Unitat de Farmacologia i Farmacognosia, Facultat de Farmacia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII s/n, 08028, Barcelona, Spain.,Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Barcelona, Spain.,Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Jaume Folch
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Unitat de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Tarragona, Spain
| | - Antoni Camins
- Departament de Farmacología, Toxicologia i Quimica Terapéutica, Unitat de Farmacologia i Farmacognosia, Facultat de Farmacia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII s/n, 08028, Barcelona, Spain. .,Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain. .,Institut de Neurociencias, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
23
|
Welters A, Klüppel C, Mrugala J, Wörmeyer L, Meissner T, Mayatepek E, Heiss C, Eberhard D, Lammert E. NMDAR antagonists for the treatment of diabetes mellitus-Current status and future directions. Diabetes Obes Metab 2017; 19 Suppl 1:95-106. [PMID: 28880473 DOI: 10.1111/dom.13017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/18/2017] [Accepted: 05/20/2017] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus is characterized by chronically elevated blood glucose levels accelerated by a progressive decline of insulin-producing β-cells in the pancreatic islets. Although medications are available to transiently adjust blood glucose to normal levels, the effects of current drugs are limited when it comes to preservation of a critical mass of functional β-cells to sustainably maintain normoglycemia. In this review, we recapitulate recent evidence on the role of pancreatic N-methyl-D-aspartate receptors (NMDARs) in β-cell physiology, and summarize effects of morphinan-based NMDAR antagonists that are beneficial for insulin secretion, glucose tolerance and islet cell survival. We further discuss NMDAR-mediated molecular pathways relevant for neuronal cell survival, which may also be important for the preservation of β-cell function and mass. Finally, we summarize the literature for evidence on the role of NMDARs in the development of diabetic long-term complications, and highlight beneficial pharmacologic aspects of NMDAR antagonists in diabetic nephropathy, retinopathy as well as neuropathy.
Collapse
Affiliation(s)
- Alena Welters
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Carina Klüppel
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
| | - Jessica Mrugala
- Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research, Helmholtz Zentrum München, Neuherberg, Düsseldorf, Germany
| | - Laura Wörmeyer
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Heiss
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Department of Biology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research, Helmholtz Zentrum München, Neuherberg, Düsseldorf, Germany
| |
Collapse
|
24
|
Srejovic I, Zivkovic V, Nikolic T, Jeremic N, Stojic I, Jeremic J, Djuric D, Jakovljevic V. Modulation of N-methyl-d-aspartate receptors in isolated rat heart. Can J Physiol Pharmacol 2017; 95:1327-1334. [PMID: 28758414 DOI: 10.1139/cjpp-2017-0056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Considering the limited data on the role of NMDA-Rs in the cardiovascular system, the aim of the present study was to examine the effects of NMDA and DL-Hcy TLHC, alone and in combination with glycine, memantine, and ifenprodil, in the isolated rat heart. The hearts of Wistar albino rats were retrogradely perfused according to the Langendorff technique at a constant perfusion pressure. The experimental protocol for all experimental groups included the stabilization period, application of estimated substance for 5 min, followed by a washout period of 10 min. Using a sensor placed in the left ventricle, we registered the following parameters of myocardial function: dp/dtmax, dp/dtmin, SLVP, DVLP, HR; CF was measured using flowmetry). We estimated the following oxidative stress biomarkers in the coronary venous effluent using spectrophotometry: TBARS, NO2-, O2-, and H2O2. NMDA alone did not induce any change in any of the observed parameters, while DL-Hcy TLHC alone, as well as a combined application of NMDA and DL-Hcy TLHC with glycine, induced a reduction of most cardiodynamic parameters. Memantine and ifenprodil induced a reduction of cardiodynamic parameters and CF, as well as some oxidative stress biomarkers.
Collapse
Affiliation(s)
- Ivan Srejovic
- a Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Vladimir Zivkovic
- a Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tamara Nikolic
- b Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena Jeremic
- b Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Isidora Stojic
- b Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Jovana Jeremic
- b Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragan Djuric
- c Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Jakovljevic
- a Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,d Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
25
|
Zakrocka I, Kocki T, Turski WA. The effect of three angiotensin-converting enzyme inhibitors on kynurenic acid production in rat kidney in vitro. Pharmacol Rep 2017; 69:536-541. [DOI: 10.1016/j.pharep.2017.01.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/11/2016] [Accepted: 01/18/2017] [Indexed: 01/19/2023]
|
26
|
Estradiol mitigates ischemia reperfusion-induced acute renal failure through NMDA receptor antagonism in rats. Mol Cell Biochem 2017; 434:33-40. [PMID: 28432550 DOI: 10.1007/s11010-017-3034-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
In the present study, we investigated possible involvement of N-methyl-D-aspartate receptors (NMDAR) in estradiol mediated protection against ischemia reperfusion (I/R)-induced acute renal failure (ARF) in rats. Bilateral renal ischemia of 40 min followed by reperfusion for 24 h induced ARF in male wistar rats. Quantification of serum creatinine, creatinine clearance (CrCl), blood urea nitrogen (BUN), uric acid, potassium, fractional excretion of sodium (FeNa), and urinary microproteins was done to assess I/R-induced renal damage in rats. Oxidative stress in kidneys was measured in terms of myeloperoxidase activity, thiobarbituric acid reactive substances, superoxide anion generation, and reduced glutathione levels. Hematoxylin & eosin and periodic acid Schiff stains were used to reveal structural changes in renal tissues. Estradiol benzoate (0.5 and 1.0 mg/kg, intraperitoneally, i.p.) was administered 1 h prior to I/R in rats. In separate groups, rats were treated with NMDAR agonists, glutamic acid (200 mg/kg, i.p.), and spermidine (20 mg/kg, i.p.) before administration of estradiol. Marked increase in serum creatinine, BUN, uric acid, serum potassium, FeNa, microproteinuria, and reduction in CrCl demonstrated I/R-induced ARF in rats. Treatment with estradiol mitigated I/R-induced changes in serum/urine parameters. Moreover, estrogen attenuated oxidative stress and structural changes in renal tissues. Prior administration of glutamic acid and spermidine abolished estradiol mediated renoprotection in rats. These results indicate the involvement of NMDAR in estradiol mediated renoprotective effect. In conclusion, we suggest that NMDAR antagonism serves as one of the mechanisms in estradiol-mediated protection against I/R-induced ARF in rats.
Collapse
|
27
|
Lan X, Lederman R, Eng JM, Shoshtari SSM, Saleem MA, Malhotra A, Singhal PC. Nicotine Induces Podocyte Apoptosis through Increasing Oxidative Stress. PLoS One 2016; 11:e0167071. [PMID: 27907022 PMCID: PMC5132002 DOI: 10.1371/journal.pone.0167071] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/20/2016] [Indexed: 12/15/2022] Open
Abstract
Background Cigarette smoking plays an important role in the progression of chronic kidney disease (CKD). Nicotine, one of the major components of cigarette smoking, has been demonstrated to increase proliferation of renal mesangial cells. In this study, we examined the effect of nicotine on podocyte injury. Methods To determine the expression of nicotinic acetylcholine receptors (nAChR subunits) in podocytes, cDNAs and cell lysate of cultured human podocytes were used for the expression of nAChR mRNAs and proteins, respectively; and mouse renal cortical sections were subjected to immunofluorescant staining. We also studied the effect of nicotine on podocyte nephrin expression, reactive oxygen species (ROS) generation (via DCFDA loading followed by fluorometric analysis), proliferation, and apoptosis (morphologic assays). We evaluated the effect of nicotine on podocyte downstream signaling including phosphorylation of ERK1/2, JNK, and p38 and established causal relationships by using respective inhibitors. We used nAChR antagonists to confirm the role of nicotine on podocyte injury. Results Human podocytes displayed robust mRNA and protein expression of nAChR in vitro studies. In vivo studies, mice renal cortical sections revealed co-localization of nAChRs along with synaptopodin. In vitro studies, nephrin expression in podocyte was decreased by nicotine. Nicotine stimulated podocyte ROS generation; nonetheless, antioxidants such as N-acetyl cysteine (NAC) and TEMPOL (superoxide dismutase mimetic agent) inhibited this effect of nicotine. Nicotine did not modulate proliferation but promoted apoptosis in podocytes. Nicotine enhanced podocyte phosphorylation of ERK1/2, JNK, and p38, and their specific inhibitors attenuated nicotine-induced apoptosis. nAChR antagonists significantly suppressed the effects of nicotine on podocyte. Conclusions Nicotine induces podocyte apoptosis through ROS generation and associated downstream MAPKs signaling. The present study provides insight into molecular mechanisms involved in smoking associated progression of chronic kidney disease.
Collapse
Affiliation(s)
- Xiqian Lan
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
- * E-mail: (XL); (PS)
| | - Rivka Lederman
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Judith M. Eng
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Seyedeh Shadafarin Marashi Shoshtari
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Moin A. Saleem
- Academic Renal Unit, Southmead Hospital, Bristol, United Kingdom
| | - Ashwani Malhotra
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
| | - Pravin C. Singhal
- Renal Molecular Research Laboratory, Feinstein Institute for Medical Research, Hofstra North Shore LIJ Medical School, New York, United States of America
- * E-mail: (XL); (PS)
| |
Collapse
|