1
|
Wang N, Li J, Tian E, Li S, Liu S, Cao F, Kong J, Yue B. Renin-angiotensin-aldosterone system variations in type 2 diabetes mellitus patients with different complications and treatments: Implications for glucose metabolism. PLoS One 2025; 20:e0316049. [PMID: 40106408 PMCID: PMC11922211 DOI: 10.1371/journal.pone.0316049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The presence of hypertension and various acute or chronic complications may affect the renin-angiotensin-aldosterone system (RAAS) in patients with type 2 diabetes mellitus (T2DM), which plays a crucial role in the regulation of glucose metabolism. However, the quantitative distribution of the RAAS components in relation to the progression of T2DM and the treatment of hyperglycemia and hypertension, as well as their association with different stages of complications and glucose metabolism, has not been well studied. METHODS We enrolled a total of 151 patients with T2DM and essential hypertension, 40 patients with T2DM and normotension, and 46 healthy controls in the study. They were categorized into subgroups based on criteria for diabetic complications. Statistical analyses, including Spearman rank correlation and multiple linear regression, were conducted to assess the relationship between RAAS components and glucose metabolism indexes such as HbA1c, FBG, CP, HOMA-β, HOMA-IR, and UACR. RESULTS The results revealed significant differences in AII, ALD, REN, and ARR levels across various complication subgroups. Notably, the concentrations of ALD and REN exhibited a consistent trend, while ARR showed an opposite trend to the REN concentration. More than 60% of hypertensive patients were treated with ACEI/ARBs and calcium channel blockers, while 29.8% of the patients were prescribed β-blockers, resulting in decreased REN and increased ARR levels. All T2DM patients received antidiabetic treatment, among which 95 (49.7%) took SGLT-2is, 40 (20.9%) took GLP-1RAs injection and 55(28.8%) took DPP-4is. The subsequent analysis revealed that SGLT-2is, GLP-1RAs, DPP-4is and other glucose-lowering agents had no statistically significant effect on the RAAS system (p > 0.05). The correlation matrix analysis indicated positive associations between ALD, REN, CP, and HOMA-IR. Furthermore, the REN levels were negatively correlated with UACR in the hypertensive group and positively correlated with HbA1c and FBG levels in the normotensive group. Multiple linear regression analysis demonstrated that ALD levels increased with higher levels of CP and HOMA-IR, independently of the RAAS system, anti-RAAS treatment and antidiabetic therapy. REN levels decreased with increasing UACR and β-blocker usage in the hypertensive group, while they increased with higher levels of HbA1c, FBG, and HOMA-IR in the normotensive group, independently of the RAAS system and antidiabetic therapy. CONCLUSIONS The activation status of the RAAS system varied among T2DM patients with different complications, highlighting the need for clinical differentiation. ALD was positively associated with insulin resistance and glucose metabolism impairment, while REN exhibited negative correlations with urinary microalbumin and β-blocker usage, and positive correlations with hyperglycemia and insulin resistance. Blocking the RAAS system holds promise for improving insulin sensitivity and β-cell function, and potentially reversing abnormal glucose tolerance or ameliorating glucose metabolism disorders.
Collapse
Affiliation(s)
- Ningning Wang
- Department of Laboratory Medicine, The First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Junhui Li
- Department of Laboratory Medicine, The First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Erjun Tian
- Department of Laboratory Medicine, The First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Shutong Li
- Department of Laboratory Medicine, The First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Shuai Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Faculty of Laboratory Medicine, The First Clinical Medical College, Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory Medicine of Henan Province, Zhengzhou, Henan, China
| | - Fei Cao
- Department of Orthopedics, The First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Junfeng Kong
- Department of Laboratory Medicine, The First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Baohong Yue
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Faculty of Laboratory Medicine, The First Clinical Medical College, Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory Medicine of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Singhal PC, Skorecki K. APOL1 Dynamics in Diabetic Kidney Disease and Hypertension. Biomolecules 2025; 15:205. [PMID: 40001508 PMCID: PMC11853202 DOI: 10.3390/biom15020205] [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: 12/18/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
APOL1 Renal Risk Variants (APOL1RRVs, G1, and G2) are known to be toxic to glomerular podocytes and causally associated with an enhanced prevalence and progression of many different etiologies of chronic kidney disease (CKD), leading to the delineation of a new disease designation of APOL1-Mediated Kidney Disease (AMKD). Notably, APOL1RRVs have not consistently been shown to increase the prevalence or severity of diabetic kidney disease (DKD) progression, which is the most common cause of End-Stage Kidney Disease (ESKD). While this apparent discrepancy seems perplexing, its clarification should provide important mechanistic and therapeutic insights. Activation of the Renin-Angiotensin System (RAS) plays a critical role in the development and progression of DKD. Recent in vitro and in vivo studies also demonstrated that RAS activation contributes to kidney cell injury in AMKD experimental models. Both high glucose, as well as APOL1RRVs escalate the podocyte expression of miR193a, a known mediator of glomerulosclerosis, including idiopathic Focal Segmental Glomerular Sclerosis (FSGS) and DKD. We propose that either the RAS and/or miR193a levels in the diabetic milieu are already maximally conducive to kidney target cell injury and, therefore, are agnostic to further injury in response to APOL1RRVs. Similarly, the contributory role of hypertension (which is frequently reported as the second most common cause of ESKD) in the progression of AMKD remains a controversial issue. Since several clinical reports have shown that controlling hypertension does not consistently slow the progression of AMKD, this has led to a formulation wherein APOL1-RRVs primarily lead to kidney injury with accompanying hypertension. Notably, half a decade later, the notion that hypertension is not a cause but rather a consequence of kidney injury was contested by investigators analyzing the Mount Sinai BioMe repository, a comprehensive clinical and genetic database including participants with APOL1RRVs. These investigators observed that hypertension predated the observed decline in GFR in individuals with APOL1RRVs by ten years. In the present study, we discuss the mechanistic forces that may underpin the gaps in these clinical manifestations, which did not allow the temporal association of hypertension with AMKD to be translated into causation and may also dissociate DKD and AMKD. We have hypothesized models that need to be validated in future experimental studies.
Collapse
Affiliation(s)
- Pravin C. Singhal
- Department of Medicine, Feinstein Institute for Medical Research, Zucker School of Medicine, Hempstead, NY 11549, USA
| | - Karl Skorecki
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525433, Israel
- Department of Nephrology, Rambam Health Care Campus, Haifa 3109601, Israel
| |
Collapse
|
3
|
Suresh V, Stillman IE, Campbell KN, Meliambro K. Focal Segmental Glomerulosclerosis. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:275-289. [PMID: 39084753 DOI: 10.1053/j.akdh.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 08/02/2024]
Abstract
Focal segmental glomerular sclerosis (FSGS) is a histological lesion characterized by sclerosis in sections (segmental) of some glomeruli (focal) in association with podocyte injury. Historically, FSGS has often been characterized as a disease, but it is a heterogeneous entity based on etiology, clinical course, and therapeutic approach. A unifying feature is podocyte injury and loss, which can be primary or the result of secondary maladaptive responses to glomerular stressors. FSGS has been demonstrated over time to carry a large health burden and remains a leading glomerular cause of ESRD globally. Recent clinical practice guidelines highlight the unmet scientific need for better understanding of disease pathogenesis, particularly for immunologic etiologies, as well as more targeted therapeutic drug development. In this review, we will discuss the current FSGS classification scheme, pathophysiologic mechanisms of injury, and treatment guidelines, along with emerging and investigational therapeutics.
Collapse
Affiliation(s)
- Varsha Suresh
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Isaac E Stillman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kirk N Campbell
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Kristin Meliambro
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
4
|
Daher G, Santos-Bezerra DP, Cavaleiro AM, Pelaes TS, Admoni SN, Perez RV, Machado CG, do Amaral FG, Cipolla-Neto J, Correa-Giannella ML. Rs4862705 in the melatonin receptor 1A gene is associated with renal function decline in type 1 diabetes individuals. Front Endocrinol (Lausanne) 2024; 15:1331012. [PMID: 38549765 PMCID: PMC10972958 DOI: 10.3389/fendo.2024.1331012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/21/2024] [Indexed: 04/02/2024] Open
Abstract
Aim The pathogenesis of chronic diabetes complications has oxidative stress as one of the major elements, and single-nucleotide polymorphisms (SNPs) in genes belonging to antioxidant pathways modulate susceptibility to these complications. Considering that melatonin is a powerful antioxidant compound, our aim was to explore, in a longitudinal cohort study of type 1 diabetes (T1D) individuals, the association of microvascular complications and SNPs in the gene encoding melatonin receptor 1A (MTNR1A). Methods Eight SNPs in MTNR1A were genotyped in 489 T1D individuals. Besides cross-sectional analyses of SNPs with each one of the microvascular complications (distal polyneuropathy, cardiovascular autonomic neuropathy, retinopathy, and diabetic kidney disease), a longitudinal analysis evaluated the associations of SNPs with renal function decline in 411 individuals followed up for a median of 8 years. In a subgroup of participants, the association of complications with urinary 6-sulfatoxymelatonin (aMT6s) concentration was investigated. Results The group of individuals with a renal function decline ≥ 5 mL min-1 1.73 m-2 year-1 presented a higher frequency of the A allele of rs4862705 in comparison with nondecliners, even after adjustment for confounding variables (OR = 1.84, 95% CI = 1.20-2.82; p = 0.0046). No other significant associations were found. Conclusions This is the first study showing an association between a variant in a gene belonging to the melatonin system and renal function decline in the diabetic setting.
Collapse
Affiliation(s)
- Gustavo Daher
- Laboratório de Carboidratos e Radioimunoensaios (LIM-18), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Daniele Pereira Santos-Bezerra
- Laboratório de Carboidratos e Radioimunoensaios (LIM-18), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Mercedes Cavaleiro
- Laboratório de Carboidratos e Radioimunoensaios (LIM-18), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Tatiana Souza Pelaes
- Laboratório de Carboidratos e Radioimunoensaios (LIM-18), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Sharon Nina Admoni
- Laboratório de Carboidratos e Radioimunoensaios (LIM-18), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo Vessoni Perez
- Laboratório de Carboidratos e Radioimunoensaios (LIM-18), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Cleide Guimarães Machado
- Divisão de Oftalmologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda Gaspar do Amaral
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Lúcia Correa-Giannella
- Laboratório de Carboidratos e Radioimunoensaios (LIM-18), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Sinha SK, Nicholas SB. Pathomechanisms of Diabetic Kidney Disease. J Clin Med 2023; 12:7349. [PMID: 38068400 PMCID: PMC10707303 DOI: 10.3390/jcm12237349] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 03/15/2024] Open
Abstract
The worldwide occurrence of diabetic kidney disease (DKD) is swiftly rising, primarily attributed to the growing population of individuals affected by type 2 diabetes. This surge has been transformed into a substantial global concern, placing additional strain on healthcare systems already grappling with significant demands. The pathogenesis of DKD is intricate, originating with hyperglycemia, which triggers various mechanisms and pathways: metabolic, hemodynamic, inflammatory, and fibrotic which ultimately lead to renal damage. Within each pathway, several mediators contribute to the development of renal structural and functional changes. Some of these mediators, such as inflammatory cytokines, reactive oxygen species, and transforming growth factor β are shared among the different pathways, leading to significant overlap and interaction between them. While current treatment options for DKD have shown advancement over previous strategies, their effectiveness remains somewhat constrained as patients still experience residual risk of disease progression. Therefore, a comprehensive grasp of the molecular mechanisms underlying the onset and progression of DKD is imperative for the continued creation of novel and groundbreaking therapies for this condition. In this review, we discuss the current achievements in fundamental research, with a particular emphasis on individual factors and recent developments in DKD treatment.
Collapse
Affiliation(s)
- Satyesh K. Sinha
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
- College of Medicine, Charles R Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Susanne B. Nicholas
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| |
Collapse
|
6
|
Tabei A, Sakairi T, Hamatani H, Ohishi Y, Watanabe M, Nakasatomi M, Ikeuchi H, Kaneko Y, Kopp JB, Hiromura K. The miR-143/145 cluster induced by TGF-β1 suppresses Wilms' tumor 1 expression in cultured human podocytes. Am J Physiol Renal Physiol 2023; 325:F121-F133. [PMID: 37167274 PMCID: PMC10511167 DOI: 10.1152/ajprenal.00313.2022] [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: 12/23/2022] [Revised: 05/01/2023] [Accepted: 05/01/2023] [Indexed: 05/13/2023] Open
Abstract
Transforming growth factor (TGF)-β1 contributes to podocyte injury in various glomerular diseases, including diabetic kidney disease, probably at least in part by attenuating the expression of Wilms' tumor 1 (WT1). However, the precise mechanisms remain to be defined. We performed miRNA microarray analysis in a human podocyte cell line cultured with TGF-β1 to examine the roles of miRNAs in podocyte damage. The microarray analysis identified miR-143-3p as the miRNA with the greatest increase following exposure to TGF-β1. Quantitative RT-PCR confirmed a significant increase in the miR-143-3p/145-5p cluster in TGF-β1-supplemented cultured podocytes and demonstrated upregulation of miR-143-3p in the glomeruli of mice with type 2 diabetes. Ectopic expression of miR-143-3p and miR-145-5p suppressed WT1 expression in cultured podocytes. Furthermore, inhibition of Smad or mammalian target of rapamycin signaling each partially reversed the TGF-β1-induced increase in miR-143-3p/145-5p and decrease in WT1. In conclusion, TGF-β1 induces expression of miR-143-3p/145-5p in part through Smad and mammalian target of rapamycin pathways, and miR-143-3p/145-5p reduces expression of WT1 in cultured human podocytes. miR-143-3p/145-5p may contribute to TGF-β1-induced podocyte injury.NEW & NOTEWORTHY This study by miRNA microarray analysis demonstrated that miR-143-3p expression was upregulated in cultured human podocytes following exposure to transforming growth factor (TGF)-β1. Furthermore, we report that the miR-143/145 cluster contributes to decreased expression of Wilms' tumor 1, which represents a possible mechanism for podocyte injury induced by TGF-β1. This study is important because it presents a novel mechanism for TGF-β-associated glomerular diseases, including diabetic kidney disease (DKD), and suggests potential therapeutic strategies targeting miR-143-3p/145-5p.
Collapse
Affiliation(s)
- Akifumi Tabei
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Toru Sakairi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroko Hamatani
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yuko Ohishi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Mitsuharu Watanabe
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Masao Nakasatomi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hidekazu Ikeuchi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yoriaki Kaneko
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Keiju Hiromura
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|
7
|
Arman K, Dalloul Z, Bozgeyik E. Emerging role of microRNAs and long non-coding RNAs in COVID-19 with implications to therapeutics. Gene 2023; 861:147232. [PMID: 36736508 PMCID: PMC9892334 DOI: 10.1016/j.gene.2023.147232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection which is commonly known as COVID-19 (COronaVIrus Disease 2019) has creeped into the human population taking tolls of life and causing tremendous economic crisis. It is indeed crucial to gain knowledge about their characteristics and interactions with human host cells. It has been shown that the majority of our genome consists of non-coding RNAs. Non-coding RNAs including micro RNAs (miRNAs) and long non-coding RNAs (lncRNAs) display significant roles in regulating gene expression in almost all cancers and viral diseases. It is intriguing that miRNAs and lncRNAs remarkably regulate the function and expression of major immune components of SARS-CoV-2. MiRNAs act via RNA interference mechanism in which they bind to the complementary sequences of the viral RNA strand, inducing the formation of silencing complex that eventually degrades or inhibits the viral RNA and viral protein expression. LncRNAs have been extensively shown to regulate gene expression in cytokine storm and thus emerges as a critical target for COVID-19 treatment. These lncRNAs also act as competing endogenous RNAs (ceRNAs) by sponging miRNAs and thus affecting the expression of downstream targets during SARS-CoV-2 infection. In this review, we extensively discuss the role of miRNAs and lncRNAs, describe their mechanism of action and their different interacting human targets cells during SARS-CoV-2 infection. Finally, we discuss possible ways how an interference with their molecular function could be exploited for new therapies against SARS-CoV-2.
Collapse
Affiliation(s)
- Kaifee Arman
- Institut de recherches cliniques de Montréal, Montréal, QC H2W 1R7, Canada.
| | - Zeinab Dalloul
- Institut de recherches cliniques de Montréal, Montréal, QC H2W 1R7, Canada
| | - Esra Bozgeyik
- Department of Medical Services and Techniques, Vocational School of Health Services, Adiyaman University, Adiyaman, Turkey
| |
Collapse
|
8
|
Chen X, Tan H, Xu J, Tian Y, Yuan Q, Zuo Y, Chen Q, Hong X, Fu H, Hou FF, Zhou L, Liu Y. Klotho-derived peptide 6 ameliorates diabetic kidney disease by targeting Wnt/β-catenin signaling. Kidney Int 2022; 102:506-520. [PMID: 35644285 DOI: 10.1016/j.kint.2022.04.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/01/2022] [Accepted: 04/27/2022] [Indexed: 01/02/2023]
Abstract
Diabetic kidney disease (DKD) is one of the most common and devastating complications of diabetic mellitus, and its prevalence is rising worldwide. Klotho, an anti-aging protein, is kidney protective in DKD. However, its large size, prohibitive cost and structural complexity hamper its potential utility in clinics. Here we report that Klotho-derived peptide 6 (KP6) mimics Klotho function and ameliorates DKD. In either an accelerated model of DKD induced by streptozotocin and advanced oxidation protein products in unilateral nephrectomized mice or db/db mice genetically prone to diabetes, chronic infusion of KP6 reversed established proteinuria, attenuated glomerular hypertrophy, mitigated podocyte damage, and ameliorated glomerulosclerosis and interstitial fibrotic lesions, but did not affect serum phosphorus and calcium levels. KP6 inhibited β-catenin activation in vivo and blocked the expression of its downstream target genes in glomerular podocytes and tubular epithelial cells. In vitro, KP6 prevented podocyte injury and inhibited β-catenin activation induced by high glucose without affecting Wnt expression. Co-immunoprecipitation revealed that KP6 bound to Wnt ligands and disrupted the engagement of Wnts with low density lipoprotein receptor-related protein 6, thereby interrupting Wnt/β-catenin signaling. Mutated KP6 with a scrambled amino acid sequence failed to bind Wnts and did not alleviate DKD in db/db mice. Thus, our studies identified KP6 as a novel Klotho-derived peptide that ameliorated DKD by blocking Wnt/β-catenin. Hence, our findings also suggest a new therapeutic strategy for the treatment of patients with DKD.
Collapse
Affiliation(s)
- Xiaowen Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Huishi Tan
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Jie Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Yuan Tian
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Qian Yuan
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Yangyang Zuo
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Qiyan Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Xue Hong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Haiyan Fu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China.
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory, Guangzhou, China; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
9
|
Khaksari M, Raji-Amirhasani A, Bashiri H, Ebrahimi MN, Azizian H. Protective effects of combining SERMs with estrogen on metabolic parameters in postmenopausal diabetic cardiovascular dysfunction: The role of cytokines and angiotensin II. Steroids 2022; 183:109023. [PMID: 35358567 DOI: 10.1016/j.steroids.2022.109023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The beneficial effects of the administration of selective estrogen receptor modulators (SERMs) and estrogen (E2), alone or in combination with each other, have been reported in postmenopausal diabetic cardiovascular dysfunction. In the present study, we determined the mechanism of action of SERMs and E2 on inflammatory balance, angiotensin II (Ang II) serum levels, and glycemic profile in a postmenopausal diabetic rat model. METHODS Ovariectomized rats with type 2 diabetes received daily SERMs (tamoxifen and raloxifene) and E2 for one month. After treatment, cardiovascular risk indices, glycemic profile, and serum Ang II, TNF-α and IL-10 levels were measured. RESULTS Type 2 diabetes caused an abnormal glycemic profile, which was exacerbated by ovariectomy. All treatments inhibited the effects of diabetes and ovariectomy on the glycemic profile, with combined treatments (SERMs + E2) showing stronger effects. Cardiovascular risk indices that became abnormal by diabetes and worsened by ovariectomy were improved in all treatment modalities. Also, combined treatment reduced serum Ang II, TNF-α, and the ratio of TNF-α to IL-10, indicating an improvement in inflammatory balance. CONCLUSION Our study showed the administration of SERMs and E2, alone or in combination, could be an effective alternative in the treatment of menopausal diabetes, and generally, the beneficial effects of combined treatments were more effective than the effects of E2 or SERMs alone. It appears that E2 or SERMs benefit the cardiovascular system by improving inflammatory balance and reducing Ang II levels.
Collapse
Affiliation(s)
- Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alireza Raji-Amirhasani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Cardiovascular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Hossein Azizian
- Neurobiomedical Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
10
|
Bai L, Sun S, Sun Y, Wang F, Nishiyama A. N-type calcium channel and renal injury. Int Urol Nephrol 2022; 54:2871-2879. [PMID: 35416563 PMCID: PMC9534814 DOI: 10.1007/s11255-022-03183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/15/2022] [Indexed: 11/26/2022]
Abstract
Accumulating evidences indicated that voltage-gated calcium channels (VDCC), including L-, T-, N-, and P/Q-type, are present in kidney and contribute to renal injury during various chronic diseases trough different mechanisms. As a voltage-gated calcium channel, N-type calcium channel was firstly been founded predominately distributed on nerve endings which control neurotransmitter releases. Since sympathetic nerve is distributed along renal afferent and efferent arterioles, N-type calcium channel blockade on sympathetic nerve terminals would bring renal dynamic improvement by dilating both arterioles and reducing glomerular pressure. In addition, large body of scientific research indicated that neurotransmitters, such as norepinephrine, releases by activating N-type calcium channel can trigger inflammatory and fibrotic signaling pathways in kidney. Interestingly, we recently demonstrated that N-type calcium channel is also expressed on podocytes and may directly contribute to podocyte injury in denervated animal models. In this paper, we will summarize our current knowledge regarding renal N-type calcium channels, and discuss how they might contribute to the river that terminates in renal injury.
Collapse
Affiliation(s)
- Lei Bai
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, People's Republic of China.
| | - Shichao Sun
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Heping Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Yao Sun
- Department of Medical Image, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, People's Republic of China
| | - Fujun Wang
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, People's Republic of China
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Medical School, Kagawa, 761-0793, Japan
| |
Collapse
|
11
|
Liu H, Sridhar VS, Boulet J, Dharia A, Khan A, Lawler PR, Cherney DZI. Cardiorenal protection with SGLT2 inhibitors in patients with diabetes mellitus: from biomarkers to clinical outcomes in heart failure and diabetic kidney disease. Metabolism 2022; 126:154918. [PMID: 34699838 DOI: 10.1016/j.metabol.2021.154918] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/23/2022]
Abstract
Type 2 diabetes (T2D) is one of the most common causes of chronic kidney disease (CKD) and cardiovascular (CV) disease. Until recently, glycemic and BP control were the cornerstones for preventing progression of CKD and CV disease associated with T2D. However, there has been a paradigm shift in treatment since the publication of the first clinical trial demonstrating benefits of sodium glucose cotransporter 2 (SGLT2) inhibitors in 2015. SGLT2 inhibitors have been shown to reduce the risk of major adverse CV events and progression of kidney disease in the setting of T2D. However, the elucidation of mechanisms of underlying these clinical benefits is the subject of ongoing investigation. Experimental studies have shown that SGLT2 inhibitors have diverse pleiotropic effects such as modulation of neurohormones such as the renin-angiotensin-aldosterone system, increasing hematocrit, altering energy substrate use, and attenuating systemic inflammation and oxidative stress, all of which have been implicated in the CV and kidney protective effects of SGLT2 inhibitors. In this review, we highlight biomarkers linked with diabetic kidney disease and heart failure and discuss how SGLT2 inhibitor-associated changes potentially mediate the cardiorenal protection observed with these therapies.
Collapse
Affiliation(s)
- Hongyan Liu
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Vikas S Sridhar
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jacinthe Boulet
- Department of Medicine, Division of Cardiology, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Atit Dharia
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada
| | - Abid Khan
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada; Division of Cardiology and Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Ravindran S, Munusamy S. Renoprotective mechanisms of sodium-glucose co-transporter 2 (SGLT2) inhibitors against the progression of diabetic kidney disease. J Cell Physiol 2021; 237:1182-1205. [PMID: 34713897 DOI: 10.1002/jcp.30621] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/02/2021] [Accepted: 10/08/2021] [Indexed: 12/19/2022]
Abstract
Sodium-glucose co-transporter 2 inhibitors (SGLT2-Is) have emerged as a promising class of antidiabetic drugs with cardioprotective and renoprotective effects in patients with type 2 diabetes (T2D). The sodium-glucose co-transporters 1 and 2 (SGLT 1 and SGLT2) located in the renal proximal tubules are responsible for glucose reabsorption from the glomerular filtrate back into the systemic circulation. Inhibition of SGLT2, which accounts for about 90% of the glucose reabsorption, leads to a significant reduction in blood glucose levels and a concomitant increase in the urinary excretion of glucose (glycosuria). Multiple mechanisms contribute to the nephroprotective effects of SGLT2-Is in T2D patients. These include: (1) Restoration of the tubuloglomerular feedback by increasing sodium delivery at macula densa, leading to afferent arteriolar constriction and reduced glomerular hyperfiltration, (2) Decreased activation of the intra-renal renin-angiotensin-aldosterone system, which also contributes to reducing glomerular hyperfiltration, (3) Increased production of ketone bodies, which serves as an alternate fuel for adenosine triphosphate production in mitochondria, which helps in attenuating inflammation, and (4) Protection against hypoxia, oxidative stress, and fibrosis. This review elaborates on the key mechanisms that underlie the nephroprotective effects and the adverse effects of SGLT2-Is in T2D patients with progressive diabetic kidney disease.
Collapse
Affiliation(s)
| | - Shankar Munusamy
- Department of Pharmaceutical and Administrative Sciences, Drake University College of Pharmacy and Health Sciences, Des Moines, Iowa, USA
| |
Collapse
|
13
|
Lehtonen S. Metformin Protects against Podocyte Injury in Diabetic Kidney Disease. Pharmaceuticals (Basel) 2020; 13:ph13120452. [PMID: 33321755 PMCID: PMC7764076 DOI: 10.3390/ph13120452] [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: 10/31/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Metformin is the most commonly prescribed drug for treating type 2 diabetes mellitus (T2D). Its mechanisms of action have been under extensive investigation, revealing that it has multiple cellular targets, either direct or indirect ones, via which it regulates numerous cellular pathways. Diabetic kidney disease (DKD), the serious complication of T2D, develops in up to 50% of the individuals with T2D. Various mechanisms contribute to the development of DKD, including hyperglycaemia, dyslipidemia, oxidative stress, chronic low-grade inflammation, altered autophagic activity and insulin resistance, among others. Metformin has been shown to affect these pathways, and thus, it could slow down or prevent the progression of DKD. Despite several animal studies demonstrating the renoprotective effects of metformin, there is no concrete evidence in clinical settings. This review summarizes the renoprotective effects of metformin in experimental settings. Special emphasis is on the effects of metformin on podocytes, the glomerular epithelial cells that are central in maintaining the glomerular ultrafiltration function.
Collapse
Affiliation(s)
- Sanna Lehtonen
- Research Program for Clinical and Molecular Metabolism and Department of Pathology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
14
|
Widiasta A, Sribudiani Y, Nugrahapraja H, Hilmanto D, Sekarwana N, Rachmadi D. Potential role of ACE2-related microRNAs in COVID-19-associated nephropathy. Noncoding RNA Res 2020; 5:153-166. [PMID: 32923747 PMCID: PMC7480227 DOI: 10.1016/j.ncrna.2020.09.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is responsible for coronavirus disease (COVID-19), potentially have severe kidney adverse effects. This organ expressed angiotensin-converting enzyme 2 (ACE2), the transmembrane protein which facilitate the entering of the virus into the cell. Therefore, early detection of the kidney manifestations of COVID-19 is crucial. Previous studies showed ACE2 role in various indications of this disease, especially in kidney effects. The MicroRNAs (miRNAs) in this organ affected ACE2 expression. Therefore, this review aims at summarizing the literature of a novel miRNA-based therapy and its potential applications in COVID-19-associated nephropathy. Furthermore, previous studies were analyzed for the kidney manifestations of COVID-19 and the miRNAs role that were published on the online databases, namely MEDLINE (PubMed) and Scopus. Several miRNAs, particularly miR-18 (which was upregulated in nephropathy), played a crucial role in ACE2 expression. Therefore, the antimiR-18 roles were summarized in various primate models that aided in developing the therapy for ACE2 related diseases.
Collapse
Affiliation(s)
- Ahmedz Widiasta
- Pediatric Nephrology Division, Child Health Department, Faculty of Medicine, Universitas Padjadjaran, Indonesia
- Medical Genetic Research Center, Faculty of Medicine, Universitas Padjadjaran, Indonesia
| | - Yunia Sribudiani
- Medical Genetic Research Center, Faculty of Medicine, Universitas Padjadjaran, Indonesia
- Department of Biomedical Sciences, Division of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Padjadjaran, Indonesia
| | - Husna Nugrahapraja
- Life Science and Biotechnology, Bandung Institute of Technology, Indonesia
| | - Dany Hilmanto
- Pediatric Nephrology Division, Child Health Department, Faculty of Medicine, Universitas Padjadjaran, Indonesia
| | - Nanan Sekarwana
- Pediatric Nephrology Division, Child Health Department, Faculty of Medicine, Universitas Padjadjaran, Indonesia
| | - Dedi Rachmadi
- Pediatric Nephrology Division, Child Health Department, Faculty of Medicine, Universitas Padjadjaran, Indonesia
- Medical Genetic Research Center, Faculty of Medicine, Universitas Padjadjaran, Indonesia
| |
Collapse
|
15
|
Juettner NE, Bogen JP, Bauer TA, Knapp S, Pfeifer F, Huettenhain SH, Meusinger R, Kraemer A, Fuchsbauer HL. Decoding the Papain Inhibitor from Streptomyces mobaraensis as Being Hydroxylated Chymostatin Derivatives: Purification, Structure Analysis, and Putative Biosynthetic Pathway. JOURNAL OF NATURAL PRODUCTS 2020; 83:2983-2995. [PMID: 32998509 DOI: 10.1021/acs.jnatprod.0c00201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Streptomyces mobaraensis produces the papain inhibitor SPI consisting of a 12 kDa protein and small active compounds (SPIac). Purification of the papain inhibitory compounds resulted in four diverse chymostatin derivatives that were characterized by NMR and MS analysis. Chymostatins are hydrophobic tetrapeptide aldehydes from streptomycetes, e.g., S. lavendulae and S. hygroscopicus, that reverse chymosin-mediated angiotensin activation and inhibit other serine and cysteine proteases. Chymotrypsin and papain were both inhibited by the SPIac compounds in the low nanomolar range. SPIac differs from the characterized chymostatins by the exchange of phenylalanine for tyrosine. The crystal structure of one of these chymostatin variants confirmed its molecular structure and revealed a S-configured hemithioacetal bond with the catalytic Cys25 thiolate as well as close interactions with hydrophobic S1 and S2 subsite amino acids. A model for chymostatin biosynthesis is provided based on the discovery of clustered genes encoding several putative nonribosomal peptide synthetases; among them, there is the unusual CstF enzyme that accommodates two canonical amino acid activation domains as well as three peptide carrier protein domains.
Collapse
Affiliation(s)
- Norbert E Juettner
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
- The Department of Biology, Technische Universität Darmstadt, Schnittspahnstraße 10, 64287 Darmstadt, Germany
| | - Jan P Bogen
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| | - Tobias A Bauer
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Felicitas Pfeifer
- The Department of Biology, Technische Universität Darmstadt, Schnittspahnstraße 10, 64287 Darmstadt, Germany
| | - Stefan H Huettenhain
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| | - Reinhard Meusinger
- The Department of Chemistry, Technische Universität Darmstadt, Alarich-Weiss-Straße 8, 64287 Darmstadt, Germany
| | - Andreas Kraemer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt am Main, Germany
| | - Hans-Lothar Fuchsbauer
- The Department of Chemical Engineering and Biotechnology, University of Applied Sciences of Darmstadt, Stephanstraße 7, 64295 Darmstadt, Germany
| |
Collapse
|
16
|
Chymase as a Possible Therapeutic Target for Amelioration of Non-Alcoholic Steatohepatitis. Int J Mol Sci 2020; 21:ijms21207543. [PMID: 33066113 PMCID: PMC7589185 DOI: 10.3390/ijms21207543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
The development and progression of non-alcoholic steatohepatitis (NASH) are linked to oxidative stress, inflammation, and fibrosis of the liver. Chymase, a chymotrypsin-like enzyme produced in mast cells, has various enzymatic actions. These actions include activation of angiotensin II, matrix metalloproteinase (MMP)-9, and transforming growth factor (TGF)-β, which are associated with oxidative stress, inflammation, and fibrosis, respectively. Augmentation of chymase activity in the liver has been reported in various NASH models. Generation of hepatic angiotensin II and related oxidative stress is upregulated in NASH but attenuated by treatment with a chymase inhibitor. Additionally, increases in MMP-9 and accumulation of inflammatory cells are observed in NASH but are decreased by chymase inhibitor administration. TGF-β and collagen I upregulation in NASH is also attenuated by chymase inhibition. These results in experimental NASH models demonstrate that a chymase inhibitor can effectively ameliorate NASH via the reduction of oxidative stress, inflammation, and fibrosis. Thus, chymase may be a therapeutic target for amelioration of NASH.
Collapse
|
17
|
Mechanism of Albuminuria Reduction by Chymase Inhibition in Diabetic Mice. Int J Mol Sci 2020; 21:ijms21207495. [PMID: 33050674 PMCID: PMC7589797 DOI: 10.3390/ijms21207495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/29/2022] Open
Abstract
Chymase has several functions, such as angiotensin II formation, which can promote diabetic kidney disease (DKD). In this study, we evaluated the effect of the chymase inhibitor TY-51469 on DKD in diabetic db/db mice. Diabetic mice were administered TY-51469 (10 mg/kg/day) or placebo for 4 weeks. No significant difference was observed in body weight and fasting blood glucose between TY-51469- and placebo-treated groups. However, a significant reduction in urinary albumin/creatinine ratio was observed in the TY-51469-treated group compared with the placebo-treated group. In the renal extract, chymase activity was significantly higher in placebo-treated mice than in non-diabetic db/m mice, but it was reduced by treatment with TY-51469. Both NADPH oxidase 4 expression and the oxidative stress marker malondialdehyde were significantly augmented in the placebo-treated group, but they were attenuated in the TY-51469-treated group. Significant increases of tumor necrosis factor-α and transforming growth factor-β mRNA levels in the placebo-treated group were significantly reduced by treatment with TY-51469. Furthermore, the expression of nephrin, which is a podocyte-specific protein, was significantly reduced in the placebo-treated group, but it was restored in the TY-51469-treated group. These findings demonstrated that chymase inhibition reduced albuminuria via attenuation of podocyte injury by oxidative stress.
Collapse
|
18
|
Bivona BJ, Takai S, Seth DM, Satou R, Harrison-Bernard LM. Chymase inhibition retards albuminuria in type 2 diabetes. Physiol Rep 2020; 7:e14302. [PMID: 31872559 PMCID: PMC6928241 DOI: 10.14814/phy2.14302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Chymase released from mast cells produces pro‐fibrotic, inflammatory, and vasoconstrictor agents. Studies were performed to test the hypothesis that chronic chymase inhibition provides a renal protective effect in type 2 diabetes. Diabetic (db/db) and control mice (db/m) were chronically infused with a chymase‐specific inhibitor or vehicle for 8 weeks. Baseline urinary albumin excretion (UalbV) averaged 42 ± 3 and 442 ± 32 microg/d in control (n = 22) and diabetic mice (n = 27), respectively (p < .05). After administration of chymase inhibitor to diabetic mice, the change in UalbV was significantly lower (459 ± 57 microg/d) than in vehicle‐treated diabetic mice (645 ± 108 microg/d). UNGALV was not different at baseline between diabetic mice that would receive the chymase inhibitor (349 ± 56 ng/d, n = 6) and vehicle (373 ± 99 ng/d, n = 6) infusions, but increased significantly only in the vehicle‐treated diabetic mice (p < .05). Glomeruli of diabetic kidneys treated chronically with chymase inhibition demonstrated reduced mesangial matrix expansion compared to glomeruli from untreated diabetic mice. Plasma angiotensin II levels were not altered by chymase inhibitor treatment. In summary, chronic chymase inhibition slowed the progression of urinary albumin excretion in diabetic mice. In conclusion, renal chymase may contribute to the progression of albuminuria in type 2 diabetes renal disease.
Collapse
Affiliation(s)
- Benjamin J Bivona
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Shinji Takai
- Department of Innovative Medicine, Osaka Medical College, Takatsuki City, Osaka, Japan
| | - Dale M Seth
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lisa M Harrison-Bernard
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
19
|
Stoll D, Yokota R, Sanches Aragão D, Casarini DE. Both aldosterone and spironolactone can modulate the intracellular ACE/ANG II/AT1 and ACE2/ANG (1-7)/MAS receptor axes in human mesangial cells. Physiol Rep 2020; 7:e14105. [PMID: 31165585 PMCID: PMC6548847 DOI: 10.14814/phy2.14105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 04/10/2019] [Accepted: 04/24/2019] [Indexed: 12/01/2022] Open
Abstract
The kidney is an important target of the renin‐ANG‐aldosterone system (RAAS). To date, several studies have demonstrated the existence of a local RAAS in various tissues, including the renal tissue. The mineralocorticoid aldosterone is known to play a critical role in the classical RAAS; however, its effect on mesangial cells (MCs) remains to be elucidated. Based on this, our aim was to investigate whether aldosterone stimulation can modulate the intracellular RAAS of immortalized human MCs by evaluating ANG‐converting enzyme (ACE)/ANG II/ANG II receptor type 1 (AT1) and ANG‐converting enzyme 2 (ACE2)/ANG (1‐7)/MAS receptor axes. To realise this, protein expression, enzyme activity, and immunofluorescence were performed under aldosterone stimulation and in the presence of the mineralocorticoid receptor (MR) antagonist spironolactone (SPI). We observed that high doses of aldosterone increase ACE activity. The effect of aldosterone on the catalytic activity of ACE was completely abolished with the pretreatment of SPI suggesting that the aldosterone‐induced cell injuries through ANG II release were attenuated. Aldosterone treatment also decreased the expression of MAS receptor, but did not alter the expression or the catalytic activity of ACE 2 and ANG (1‐7) levels. Spironolactone modulated the localization of ANG II and AT1 receptor and decreased ANG (1‐7) and MAS receptor levels. Our data suggest that both aldosterone and the MR receptor antagonist can modulate both of these axes and that spironolactone can protect MCs from the damage induced by aldosterone.
Collapse
Affiliation(s)
- Danielle Stoll
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| | - Rodrigo Yokota
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| | - Danielle Sanches Aragão
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| | - Dulce E Casarini
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| |
Collapse
|
20
|
Aleksova A, Ferro F, Gagno G, Padoan L, Saro R, Santon D, Stenner E, Barbati G, Cappelletto C, Rossi M, Beltrami AP, Sinagra G. Diabetes Mellitus and Vitamin D Deficiency:Comparable Effect on Survival and a DeadlyAssociation after a Myocardial Infarction. J Clin Med 2020; 9:E2127. [PMID: 32640692 PMCID: PMC7408858 DOI: 10.3390/jcm9072127] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Survivors after a myocardial infarction (MI), especially those with diabetes mellitus (DM),remain at high risk of further events. Identifying and treating factors that may influence survivalmay open new therapeutic strategies. We assessed the impact on prognosis of DM andhypovitaminosis D (hypovitD), alone or combined. In this prospective, observational study, 1081patients were enrolled surviving an MI and divided into four groups according to their diabetic andVitD status. The primary end-point was composite of all-cause mortality, angina/MI and heartfailure (HF). Secondary outcomes were mortality, HF and angina/MI. During a follow-up of 26.1months (IQR 6.6-64.5), 391 subjects experienced the primary end-point. Patients with DM orhypovitD had similar rate of the composite end-point. Patients with only hypovitD or DM did notdiffer regarding components of composite end-point (angina p = 0.97, HF p = 0.29, mortality p = 0.62).DM and VitD deficiency had similarly adjusted risks for primary end-point (HR 1.3, 95%CI 1.05-1.61; HR 1.3, 95% CI 1.04-1.64). The adjusted HR for primary composite end-point for patients withhypovitD and DM was 1.69 (95%CI 1.25-2.29, p = 0.001) in comparison to patients with neitherhypoD nor DM. In conclusion, DM and hypovitD, individually and synergistically, are associatedwith a worse outcome after MI.
Collapse
Affiliation(s)
- Aneta Aleksova
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy; (F.F.); (G.G.); (R.S.); (C.C.); (M.R.); (G.S.)
| | - Federico Ferro
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy; (F.F.); (G.G.); (R.S.); (C.C.); (M.R.); (G.S.)
| | - Giulia Gagno
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy; (F.F.); (G.G.); (R.S.); (C.C.); (M.R.); (G.S.)
| | - Laura Padoan
- Azienda Ospedaliera di Perugia and University of Perugia, Cardiology and Cardiovascular Physiopathology, 06156 Perugia, Italy;
| | - Riccardo Saro
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy; (F.F.); (G.G.); (R.S.); (C.C.); (M.R.); (G.S.)
| | - Daniela Santon
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), 34100 Trieste, Italy; (D.S.); (E.S.)
| | - Elisabetta Stenner
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), 34100 Trieste, Italy; (D.S.); (E.S.)
| | - Giulia Barbati
- Biostatistics Unit, Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy;
| | - Chiara Cappelletto
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy; (F.F.); (G.G.); (R.S.); (C.C.); (M.R.); (G.S.)
| | - Maddalena Rossi
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy; (F.F.); (G.G.); (R.S.); (C.C.); (M.R.); (G.S.)
| | | | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI) and Department of Medical Surgical and Health Sciences, University of Trieste, 34100 Trieste, Italy; (F.F.); (G.G.); (R.S.); (C.C.); (M.R.); (G.S.)
| |
Collapse
|
21
|
Wysocki J, Schulze A, Batlle D. Novel Variants of Angiotensin Converting Enzyme-2 of Shorter Molecular Size to Target the Kidney Renin Angiotensin System. Biomolecules 2019; 9:E886. [PMID: 31861139 PMCID: PMC6995632 DOI: 10.3390/biom9120886] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/27/2019] [Accepted: 12/10/2019] [Indexed: 12/29/2022] Open
Abstract
ACE2 is a monocarboxypeptidase which generates Angiotensin (1-7) from Angiotensin II (1-8). Attempts to target the kidney Renin Angiotensin System using native ACE2 to treat kidney disease are hampered by its large molecular size, 100 kDa, which precludes its glomerular filtration and subsequent tubular uptake. Here, we show that both urine and kidney lysates are capable of digesting native ACE2 into shorter proteins of ~60-75 kDa and then demonstrate that they are enzymatically very active. We then truncated the native ACE2 by design from the C-terminus to generate two short recombinant (r)ACE2 variants (1-605 and 1-619AA). These two truncates have a molecular size of ~70 kDa, as expected from the amino acid sequence and as shown by Western blot. ACE2 enzyme activity, measured using a specific substrate, was higher than that of the native rACE2 (1-740 AA). When infused to mice with genetic ACE2 deficiency, a single i.v. injection of 1-619 resulted in detectable ACE2 activity in urine, whereas infusion of the native ACE2 did not. Moreover, ACE2 activity was recovered in harvested kidneys from ACE2-deficient mice infused with 1-619, but not in controls (23.1 ± 4.3 RFU/µg creatinine/h and 1.96 ± 0.73 RFU/µg protein/hr, respectively). In addition, the kidneys of ACE2-null mice infused with 1-619 studied ex vivo formed more Ang (1-7) from exogenous Ang II than those infused with vehicle (AUC 8555 ± 1933 vs. 3439 ± 753 ng/mL, respectively, p < 0.05) further demonstrating the functional effect of increasing kidney ACE2 activity after the infusion of our short ACE2 1-619 variant. We conclude that our novel short recombinant ACE2 variants undergo glomerular filtration, which is associated with kidney uptake of enzymatically active proteins that can enhance the formation of Ang (1-7) from Ang II. These small ACE2 variants may offer a potentially useful approach to target kidney RAS overactivity to combat kidney injury.
Collapse
Affiliation(s)
- Jan Wysocki
- Department of Medicine, Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008, USA
| | - Arndt Schulze
- Department of Medicine, Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008, USA
- Department of Medicine, Charité-Universitätsmedizin, D-10117 Berlin, Germany
| | - Daniel Batlle
- Department of Medicine, Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611-3008, USA
| |
Collapse
|
22
|
Abstract
Finding new therapeutic targets of glomerulosclerosis treatment is an ongoing quest. Due to a living environment of various stresses and pathological stimuli, podocytes are prone to injuries; moreover, as a cell without proliferative potential, loss of podocytes is vital in the pathogenesis of glomerulosclerosis. Thus, sufficient understanding of factors and underlying mechanisms of podocyte injury facilitates the advancement of treating and prevention of glomerulosclerosis. The clinical symptom of podocyte injury is proteinuria, sometimes with loss of kidney functions progressing to glomerulosclerosis. Injury-induced changes in podocyte physiology and function are actually not a simple passive process, but a complex interaction of proteins that comprise the anatomical structure of podocytes at molecular levels. This chapter lists several aspects of podocyte injuries along with potential mechanisms, including glucose and lipid metabolism disorder, hypertension, RAS activation, micro-inflammation, immune disorder, and other factors. These aspects are not technically separated items, but intertwined with each other in the pathogenesis of podocyte injuries.
Collapse
|
23
|
A Glucose-Dependent Pharmacokinetic/ Pharmacodynamic Model of ACE Inhibition in Kidney Cells. Processes (Basel) 2019. [DOI: 10.3390/pr7030131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is a major cause of renal failure. Podocytes are terminally differentiated renal epithelial cells that are key targets of damage due to DKD. Podocytes express a glucose-stimulated local renin-angiotensin system (RAS) that produces angiotensin II (ANG II). Local RAS differs from systemic RAS, which has been studied widely. Hyperglycemia increases the production of ANG II by podocyte cells, leading to podocyte injury. Angiotensin-converting enzyme (ACE) is involved in the production of ANG II, and ACE inhibitors are drugs used to suppress elevated ANG II concentration. As systemic RAS differs from the local RAS in podocytes, ACE inhibitor drugs should act differently in local versus systemic contexts. Experimental and computational studies have considered the pharmacokinetics (PK) and pharmacodynamics (PD) of ACE inhibition of the systemic RAS. Here, a PK/PD model for ACE inhibition is developed for the local RAS in podocytes. The model takes constant or dynamic subject-specific glucose concentration input to predict the ANG II concentration and the corresponding effects of drug doses locally and systemically. The model is developed for normal and impaired renal function in combination with different glucose conditions, thus enabling the study of various pathophysiological conditions. Parameter uncertainty is also analyzed. Such a model can improve the study of the effects of drugs at the cellular level and can aid in development of therapeutic approaches to slow the progression of DKD.
Collapse
|
24
|
Ma R, Xu Y, Zhou H, Zhang D, Yao D, Song L, Liu Y. Participation of the AngII/TRPC6/NFAT axis in the pathogenesis of podocyte injury in rats with type 2 diabetes. Mol Med Rep 2019; 19:2421-2430. [PMID: 30664212 DOI: 10.3892/mmr.2019.9871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 12/19/2018] [Indexed: 11/06/2022] Open
Abstract
The canonical transient receptor potential channel 6 ion channel is expressed in podocytes and is an important component of the glomerular slit diaphragm. Focal segmental glomerulosclerosis is closely associated with TRPC6 gene mutations, and TRPC6 mediates podocyte injury induced by high glucose. Angiotensin II (AngII) has been revealed to enhance TRPC6 currents in certain types of cells, including podocytes and ventricular myocytes. It has been reported that glucose regulated TRPC6 expression in an AngII‑dependent manner in podocytes and that this pathway is critical in diabetic nephropathy. In the present study, the role of TRPC6 detected by western blotting and reverse transcription‑quantitative polymerase chain reaction in AngII‑mediated podocyte injury was evaluated in rats with type 2 diabetes induced by high‑calorie diets and streptozotocin. The results demonstrated that urinary albumin excretion was elevated, and morphological changes, including glomerular basement membrane thickening and podocyte process effacement, were observed. There was increased expression of AngII and TRPC6 in diabetic rats. The angiotensin receptor blocker valsartan significantly reduced TRPC6 and nuclear factor of activated T‑cells (NFAT) overexpression in diabetic rats. These results in vivo were confirmed by studies in vitro, which demonstrated that inhibition of TRPC6 ameliorated high glucose‑induced podocyte injury by decreasing NFAT mRNA levels. Taken together, the present results suggested that the AngII/TRPC6/NFAT axis may be a crucial signaling pathway in podocytes that is necessary for maintaining the integrity of the glomerular filtration barrier. In addition, TRPC6 may represent a potential therapeutic target for diabetic nephropathy.
Collapse
Affiliation(s)
- Ruixia Ma
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Yan Xu
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Hanyan Zhou
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Di Zhang
- Department of Special Medicine, School of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Dandan Yao
- Department of Nephrology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Limin Song
- Department of Special Medicine, School of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yuan Liu
- Department of Special Medicine, School of Medicine, Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
25
|
Meng XM. Inflammatory Mediators and Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:381-406. [PMID: 31399975 DOI: 10.1007/978-981-13-8871-2_18] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Renal inflammation is the initial, healthy response to renal injury. However, prolonged inflammation promotes the fibrosis process, which leads to chronic pathology and eventually end-stage kidney disease. There are two major sources of inflammatory cells: first, bone marrow-derived leukocytes that include neutrophils, macrophages, fibrocytes and mast cells, and second, locally activated kidney cells such as mesangial cells, podocytes, tubular epithelial cells, endothelial cells and fibroblasts. These activated cells produce many profibrotic cytokines and growth factors that cause accumulation and activation of myofibroblasts, and enhance the production of the extracellular matrix. In particular, activated macrophages are key mediators that drive acute inflammation into chronic kidney disease. They produce large amounts of profibrotic factors and modify the microenvironment via a paracrine effect, and they also transdifferentiate to myofibroblasts directly, although the origin of myofibroblasts in the fibrosing kidney remains controversial. Collectively, understanding inflammatory cell functions and mechanisms during renal fibrosis is paramount to improving diagnosis and treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Xiao-Ming Meng
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
26
|
Mohamed EA, Ahmed HI, Zaky HS. Protective effect of irbesartan against doxorubicin-induced nephrotoxicity in rats: implication of AMPK, PI3K/Akt, and mTOR signaling pathways. Can J Physiol Pharmacol 2018; 96:1209-1217. [DOI: 10.1139/cjpp-2018-0259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nephrotoxicity is one of the serious undesirable effects related to doxorubicin (DOX). Herein, we have investigated the potential protective effect of irbesartan (IRB) against chronic nephrotoxicity induced by DOX, and the implication of different mechanistic pathways underlying these effects. Rats were treated with either DOX (2.5 mg/kg i.p., 3 times/week) for 2 weeks, and (or) IRB (40 mg/kg, daily) for 3 weeks. IRB prohibited nephrotoxicity induced by DOX, which was evident by the increase in blood urea nitrogen and creatinine levels and histopathological changes. IRB improved DOX-induced alterations in oxidative status by diminishing lipid peroxidation and upregulating the antioxidant enzymes. Also, upon DOX treatment, the renal expression of tumor necrosis factor-α, interleukin-6, and caspase-3 were significantly increased; IRB diminished DOX-induced alterations in these parameters. Moreover, DOX significantly decreased the expression level of AMP-activated protein kinase (AMPK). Meanwhile, DOX induced activation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt/PKB) and mammalian target of rapamycin (mTOR) pathways that cross talked with AMPK. On the contrary, IRB successfully counterbalanced all these effects. Collectively, these outcomes suggest that the modulation of AMPK, PI3K, Akt, and mTOR pathways plays a critical role in conferring the protective effects of IRB against DOX nephrotoxicity.
Collapse
Affiliation(s)
- Eman A. Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Hebatalla I. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Heba S. Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
27
|
Mathematical Model for Glucose Dependence of the Local Renin-Angiotensin System in Podocytes. Bull Math Biol 2018. [PMID: 29520569 DOI: 10.1007/s11538-018-0408-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Diabetic kidney disease (DKD) is the primary cause of kidney failure. Diabetic hyperglycemia primarily damages podocyte cells. Podocytes express a local renin-angiotensin system (RAS) that produces angiotensin II (ANG II). ANG II levels are elevated by hyperglycemia, triggering podocyte injury. Quantitative descriptions of glucose dose dependency of ANG II are scarce in the literature. For better understanding of the mechanism of glycemic injury in DKD, a mathematical model is developed to describe the glucose-stimulated local RAS in podocytes. The model of the RAS signaling pathway in podocytes tracks peptides and enzymes without explicit glucose dependence. Local and global sensitivity analyses are used to identify the key parameters to be estimated in the model. Three approaches are explored to incorporate glucose dependency through linear ramp functions for the sensitive parameters. The first approach uses inferences from literature data to estimate the parameter values, while the other approaches reduce the number of assumptions by using least-squares regression to estimate all or a subset of the parameters. Physiological parameter values and RAS peptide concentrations ranges are used to discriminate between plausible models for the glucose dose dependency. This is the first model of the theory of the local RAS mechanism specific to podocyte cells to track ANG II levels in a range of glycemic conditions that may contribute to podocyte damage in DKD. The ability to track ANG II behavior could enable prediction of its downstream effects on podocytes and provide opportunities to better characterize pathophysiological features of DKD progression.
Collapse
|
28
|
Patney V, Chaudhary K, Whaley-Connell A. Treatment of Diabetic Kidney Disease With Hypertension Control and Renin Angiotensin System Inhibition. Adv Chronic Kidney Dis 2018; 25:158-165. [PMID: 29580580 DOI: 10.1053/j.ackd.2017.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/16/2017] [Accepted: 11/28/2017] [Indexed: 02/07/2023]
Abstract
The global incidence and prevalence of diabetes continues to expand due primarily to the influences of obesity and the contribution of obesity to the progression of type 2 diabetes mellitus. The rising prevalence of type 2 diabetes has driven an increase in rates of CKD in the past 3 decades in the United States. In turn, so have the rates for complications related to type 2 diabetes including CKD, eg, diabetic kidney disease (DKD). Although incident rates for DKD have stabilized in the recent years, diabetes continues to be the leading cause of ESRD in the United States. The United Kingdom Prospective Diabetes Study data and other population-level studies support that lowering blood pressure reduces kidney disease and cardiovascular disease in patients with type 2 diabetes. Furthermore, strategies targeting renin-angiotensin-aldosterone system interruption have shown to improve DKD outcomes to a greater extent than other classes of antihypertensive regimens.
Collapse
|
29
|
Intracrine action of angiotensin II in mesangial cells: subcellular distribution of angiotensin II receptor subtypes AT 1 and AT 2. Mol Cell Biochem 2018; 448:265-274. [PMID: 29455433 DOI: 10.1007/s11010-018-3331-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/07/2018] [Indexed: 10/18/2022]
Abstract
Biological effects of angiotensin II (AngII) such as regulation of AngII target genes may be triggered by interaction of AngII with intracellular AngII receptor types 1 and 2 (AT1 and AT2), defined as intracrine response. The aim of this study was to examine the presence of AT1 and AT2 receptors in nuclear membrane of human mesangial cells (HMCs) and evaluate the possible biological effects mediated by intracellular AT1 through an intracrine mechanism. Subcellular distribution of AT1 and AT2 was evaluated by immunofluorescence and by western blot in isolated nuclear extract. Endogenous intracellular synthesis of AngII was stimulated by high glucose (HG). Effects of HG were analyzed in the presence of candesartan, which prevents AngII internalization. Both receptors were found in nuclear membrane. Fluorescein isothiocyanate (FITC)-labeled AngII added to isolated nuclei produced a fluorescence that was reduced in the presence of losartan or PD-123319 and quenched in the presence of both inhibitors simultaneously. HG induced overexpression of fibronectin and increased cell proliferation in the presence of candesartan, indicating an intracrine action of AngII induced by HG. Results showed the presence of nuclear receptors in HMCs that can be activated by AngII through an intracrine response independent of cytoplasmic membrane AngII receptors.
Collapse
|
30
|
Lewko B, Maryn A, Latawiec E, Daca A, Rybczynska A. Angiotensin II Modulates Podocyte Glucose Transport. Front Endocrinol (Lausanne) 2018; 9:418. [PMID: 30087656 PMCID: PMC6066665 DOI: 10.3389/fendo.2018.00418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/02/2018] [Indexed: 11/27/2022] Open
Abstract
Podocytes play a central role in the maintenance of the glomerular filtration barrier and are cellular targets of angiotensin II (AngII). Non-hemodynamic pathways of AngII signaling regulate cellular function and mediate podocyte abnormalities that are associated with various glomerulopathies, including diabetic kidney disease. In this study we investigated the capacity of AngII to modulate glucose uptake in mouse podocytes expressing the human AT1 receptor (AT1R+) after 5 days of exposure to normal (NG, 5.6 mmol/L) or to high (HG, 30 mmol/L) glucose. Short (30 min) as well as long-term (24 h) incubations with AngII markedly enhanced glucose transport in both NG and HG cells. In podocytes cultured under NG conditions, AngII inhibited insulin-stimulated glucose uptake. Regardless of the presence or absence of AngII, no effect of insulin on glucose uptake was observed in HG cells. Stimulation of glucose transport by AngII was mediated by protein kinase C and by phosphoinositide 3-kinase. Glucose dependent surface expression of the glucose transporters GLUT1, GLUT2, and GLUT4 was modulated by AngII in a time and glucose concentration dependent manner. Furthermore, despite its inhibitory effect on insulin's action, AngII elevated the number of podocyte insulin receptors in both NG and HG cultured cells. These findings demonstrate that AngII modulates podocyte basal, as well as insulin-dependent glucose uptake by regulating glucose transporters and insulin signaling.
Collapse
Affiliation(s)
- Barbara Lewko
- Department of Pathophysiology Faculty of Pharmacy, Medical University of Gdansk, Gdańsk, Poland
- *Correspondence: Barbara Lewko
| | - Anna Maryn
- Department of Pathophysiology Faculty of Pharmacy, Medical University of Gdansk, Gdańsk, Poland
| | - Elzbieta Latawiec
- Department of Pathophysiology Faculty of Pharmacy, Medical University of Gdansk, Gdańsk, Poland
| | - Agnieszka Daca
- Department of Pathology and Experimental Rheumatology, Faculty of Medicine, Medical University of Gdansk, Gdańsk, Poland
| | - Apolonia Rybczynska
- Department of Pathophysiology Faculty of Pharmacy, Medical University of Gdansk, Gdańsk, Poland
| |
Collapse
|
31
|
Griffin TP, Wall D, Browne GA, Dennedy MC, O'Shea PM. Associations between glycaemic control and activation of the renin-angiotensin-aldosterone system in participants with type 2 diabetes mellitus and hypertension. Ann Clin Biochem 2017; 55:373-384. [DOI: 10.1177/0004563217728964] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction Hyperglycaemia increases succinate concentrations and succinate receptor activation in the kidney resulting in renin release. The aim of our study was to determine if there is an association between glycaemic control as evidenced by glycated haemoglobin values and activation of the renin-angiotensin-aldosterone system in patients with type 2 diabetes mellitus and hypertension. Methods A cross-sectional study was conducted at Galway University Hospitals between December 2014 and March 2015. Participants ( n = 66) were identified following interrogation of the electronic database for patients with type 2 diabetes mellitus. Baseline clinical demographics, aldosterone, plasma renin activity, direct renin concentration, urea and electrolytes, glycated haemoglobin, cholesterol, urine sodium and albumin creatinine ratio were recorded. Results There was a significant positive linear correlation between glycated haemoglobin and renin (both plasma renin activity [ P = 0.002] and direct renin concentration [ P = 0.008]) and between serum creatinine and aldosterone measured using both radioimmunoassay ( P = 0.008) and immunochemiluminometric assay ( P = 0.008). A significant negative linear correlation was demonstrated between serum sodium and plasma renin activity ( P = 0.005) and direct renin concentration ( P = 0.015) and between estimated glomerular filtration rate and aldosterone measured using radioimmunoassay ( P = 0.02) and immunochemiluminometric assay ( P = 0.016). A significant negative linear correlation existed between urine sodium and plasma renin activity ( P = 0.04) and aldosterone measured using radioimmunoassay ( P = 0.045). Conclusions There is a direct positive association between glycaemic control and renin. We advocate for renin measurement to be part of the diabetologist's armamentarium to assess, guide and optimize therapeutic strategies in patients with diabetes.
Collapse
Affiliation(s)
- TP Griffin
- Centre for Endocrinology, Diabetes and Metabolism, Galway University Hospitals, Galway, Ireland
| | - D Wall
- School of Mathematics, Statistics and Applied Mathematics, National University of Ireland, Galway, Ireland
| | - GA Browne
- Discipline of Pharmacology & Therapeutics, Lambe Institute/Translational Research Facility, School of Medicine, National University of Ireland, Galway, Ireland
| | - MC Dennedy
- Centre for Endocrinology, Diabetes and Metabolism, Galway University Hospitals, Galway, Ireland
- Discipline of Pharmacology & Therapeutics, Lambe Institute/Translational Research Facility, School of Medicine, National University of Ireland, Galway, Ireland
| | - PM O'Shea
- Department of Clinical Biochemistry, Galway University Hospitals, Galway, Ireland
| |
Collapse
|
32
|
Satirapoj B. Sodium-Glucose Cotransporter 2 Inhibitors with Renoprotective Effects. KIDNEY DISEASES (BASEL, SWITZERLAND) 2017; 3:24-32. [PMID: 28785561 PMCID: PMC5527177 DOI: 10.1159/000471765] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/17/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Diabetes is the leading cause of end-stage renal disease (ESRD) and accounts for 40-50% of patients requiring renal replacement therapy. The main pathophysiology of diabetic nephropathy comprises glucose-dependent pathways, hemodynamic pathways, and genetic factors. SUMMARY Glucose-dependent pathways, known as advanced glycation, polyols, and protein kinase C activation have been implicated in the pathogenesis of diabetic nephropathy. Current studies have indicated that intensified glycemic control retards the rate of development of albuminuria and impairs renal function in both patients with type 1 and 2 diabetes. However, therapeutic options have substantially increased over the last decade, but have not yet been translated to remarkably reduce the incidence of ESRD from diabetic nephropathy. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a novel class of glucose-lowering agents with potential renoprotective effects. KEY MESSAGE SGLT2 inhibitors represent a promising therapeutic approach to prevent and improve nephropathy among patients with type 2 diabetes. The current data strongly support that SGLT2 inhibitors have renoprotective properties not only by improving glycemic control but also through hemodynamic and nonhemodynamic renal effects. This review focuses on the latest published data dealing with hypoglycemic agents and SGLT2 inhibitors regarding the progression of kidney disease.
Collapse
Affiliation(s)
- Bancha Satirapoj
- Division of Nephrology, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| |
Collapse
|
33
|
Mesenchymal Stem Cell-Based Therapies against Podocyte Damage in Diabetic Nephropathy. Tissue Eng Regen Med 2017; 14:201-210. [PMID: 30603477 DOI: 10.1007/s13770-017-0026-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 05/25/2016] [Accepted: 06/13/2016] [Indexed: 01/02/2023] Open
Abstract
Injury to podocytes is an early event in diabetic nephropathy leading to proteinuria with possible progression to end-stage renal failure. The podocytes are unique and highly specialized cells that cover the outer layer of kidney ultra-filtration barrier and play an important role in glomerular function. In the past few decades, adult stem cells, such as mesenchymal stem cells (MSCs) with a regenerative and differentiative capacity have been extensively used in cell-based therapies. In addition to their capability for regeneration and differentiation, MSCs contributes to their milieu by paracrine action of a series of growth factors via antiapoptotic, mitogenic and other cytokine actions that actively participate in treatment of podocyte damage through prevention of podocyte effacement, detachment and apoptosis. It is hoped that novel stem cell-based therapies will be developed in the future to prevent podocyte injury, thereby reducing the burden of kidney disease.
Collapse
|
34
|
Velez JCQ, Arif E, Rodgers J, Hicks MP, Arthur JM, Nihalani D, Bruner ET, Budisavljevic MN, Atkinson C, Fitzgibbon WR, Janech MG. Deficiency of the Angiotensinase Aminopeptidase A Increases Susceptibility to Glomerular Injury. J Am Soc Nephrol 2017; 28:2119-2132. [PMID: 28202497 DOI: 10.1681/asn.2016111166] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/04/2017] [Indexed: 01/13/2023] Open
Abstract
Aminopeptidase A (APA) is expressed in glomerular podocytes and tubular epithelia and metabolizes angiotensin II (AngII), a peptide known to promote glomerulosclerosis. In this study, we tested whether APA expression changes in response to progressive nephron loss or whether APA exerts a protective role against glomerular damage and during AngII-mediated hypertensive kidney injury. At advanced stages of FSGS, fawn-hooded hypertensive rat kidneys exhibited distinctly increased APA staining in areas of intact glomerular capillary loops. Moreover, BALB/c APA-knockout (KO) mice injected with a nephrotoxic serum showed persistent glomerular hyalinosis and albuminuria 96 hours after injection, whereas wild-type controls achieved virtually full recovery. We then tested the effect of 4-week infusion of AngII (400 ng/kg per minute) in APA-KO and wild-type mice. Although we observed no significant difference in achieved systolic BP, AngII-treated APA-KO mice developed a significant rise in albuminuria not observed in AngII-treated wild-type mice along with increased segmental and global sclerosis and/or collapse of juxtamedullary glomeruli, microcystic tubular dilation, and tubulointerstitial fibrosis. In parallel, AngII treatment significantly increased the kidney AngII content and attenuated the expression of podocyte nephrin in APA-KO mice but not in wild-type controls. These data show that deficiency of APA increases susceptibility to glomerular injury in BALB/c mice. The augmented AngII-mediated kidney injury observed in association with increased intrarenal AngII accumulation in the absence of APA suggests a protective metabolizing role of APA in AngII-mediated glomerular diseases.
Collapse
Affiliation(s)
- Juan Carlos Q Velez
- Department of Nephrology, Ochsner Clinic Foundation, New Orleans, Louisiana;
| | | | | | - Megan P Hicks
- Institute of Public and Preventative Health, Augusta University, Augusta, Georgia; and
| | - John M Arthur
- Division of Nephrology, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | | | | | - Carl Atkinson
- Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | | | | |
Collapse
|
35
|
Yamazaki M, Fukusumi Y, Kayaba M, Kitazawa Y, Takamura S, Narita I, Kawachi H. Possible role for glomerular-derived angiotensinogen in nephrotic syndrome. J Renin Angiotensin Aldosterone Syst 2016; 17:17/4/1470320316681223. [PMID: 27932705 PMCID: PMC5843942 DOI: 10.1177/1470320316681223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/28/2016] [Indexed: 01/13/2023] Open
Abstract
Background and objective: Renin–angiotensin system (RAS) inhibitors reduce glomerular injury and proteinuria, indicating that angiotensin II (Ang II) is involved in glomerular diseases. Although the local RAS is reported to play an essential role in maintaining local tissue functions, the role of the local RAS in regulating glomerular function is not well evaluated. In this study, we analyzed the glomerular expression of RAS components in nephrotic models and the effect of Ang II receptor blockers (ARB) on the expression of angiotensinogen (AGT). Methods: The levels of glomerular expression of RAS components were analyzed in two nephrotic models: anti-nephrin antibody-induced nephropathy and PAN nephropathy, a mimic of human minimal change nephrotic syndrome. The effect of the ARB irbesartan on the expression of AGT in the nephrotic model was analyzed. Results: Glomerular expression of AGT and the receptors for Ang II was clearly increased in the nephrotic models, while the expression levels of renin, ACE and ACE2 were decreased. ARB treatment suppressed the increase of glomerular expression of AGT in the nephrotic model. Conclusion: It is conceivable that the promoted local RAS action participated in the glomerular dysfunction, and that ARB treatment ameliorated slit diaphragm injury by inhibiting the positive feedback loop of the activated local Ang II action.
Collapse
Affiliation(s)
- Mihoko Yamazaki
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan.,Department of Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | - Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | - Mutsumi Kayaba
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | - Yukina Kitazawa
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | - Sayuri Takamura
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | - Ichiei Narita
- Department of Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | - Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Japan
| |
Collapse
|
36
|
Sonneveld R, Hoenderop JG, Isidori AM, Henique C, Dijkman HB, Berden JH, Tharaux PL, van der Vlag J, Nijenhuis T. Sildenafil Prevents Podocyte Injury via PPAR- γ-Mediated TRPC6 Inhibition. J Am Soc Nephrol 2016; 28:1491-1505. [PMID: 27895156 DOI: 10.1681/asn.2015080885] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/02/2016] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential channel C6 (TRPC6) gain-of-function mutations and increased TRPC6 expression in podocytes induce glomerular injury and proteinuria. Sildenafil reduces TRPC6 expression and activity in nonrenal cell types, although the mechanism is unknown. Peroxisome proliferator-activated receptor γ (PPAR-γ) is a downstream target of sildenafil in the cyclic guanosine monophosphate (cGMP)-activated protein kinase G (PKG) axis. PPAR-γ agonists, like pioglitazone, appear antiproteinuric. We hypothesized that sildenafil inhibits TRPC6 expression in podocytes through PPAR-γ-dependent mechanisms, thereby counteracting podocyte injury and proteinuria. Treatment with sildenafil, the cGMP derivative 8-bromoguanosine 3',5'-cyclic monophosphate sodium salt (8-Br-cGMP), or pioglitazone dose-dependently downregulated podocyte injury-induced TRPC6 expression in vitro Knockdown or application of antagonists of PKG or PPAR-γ enhanced TRPC6 expression in podocytes and counteracted effects of sildenafil and 8-Br-cGMP. We observed similar effects on TRPC6 promoter activity and TRPC6-dependent calcium influx. Chromatin immunoprecipitation showed PPAR-γ binding to the TRPC6 promoter. Sildenafil or pioglitazone treatment prevented proteinuria and the increased TRPC6 expression in rats with adriamycin-induced nephropathy and mice with hyperglycemia-induced renal injury. Rats receiving PPAR-γ antagonists displayed proteinuria and increased podocyte TRPC6 expression, as did podocyte-specific PPAR-γ knockout mice, which were more sensitive to adriamycin and not protected by sildenafil. Thus, sildenafil ameliorates podocyte injury and prevents proteinuria through cGMP- and PKG-dependent binding of PPAR-γ to the TRPC6 promoter, which inhibits TRPC6 promoter activity, expression, and activity. Because sildenafil is approved for clinical use, our results suggest that additional clinical study of its antiproteinuric effect in glomerular disease is warranted.
Collapse
Affiliation(s)
| | | | - Andrea M Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Carole Henique
- Paris Cardiovascular Centre, Institut National de la Santé et de la Recherche Médicale, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France; and
| | - Henry B Dijkman
- Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Pierre-Louis Tharaux
- Paris Cardiovascular Centre, Institut National de la Santé et de la Recherche Médicale, Paris, France.,Sorbonne Paris Cité, Université Paris Descartes, Paris, France; and.,Service de Néphrologie, Hôpital Européen Georges Pompidou, Paris, France
| | | | | |
Collapse
|
37
|
Dugbartey GJ. Diabetic nephropathy: A potential savior with 'rotten-egg' smell. Pharmacol Rep 2016; 69:331-339. [PMID: 28183033 DOI: 10.1016/j.pharep.2016.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/20/2016] [Accepted: 11/09/2016] [Indexed: 02/06/2023]
Abstract
Diabetic nephropathy (DN) is currently the leading cause of end-stage renal disease. Despite optimal management, DN is still a major contributor to morbidity and mortality of diabetic patients worldwide. The major pathological alterations in DN include excessive accumulation and deposition of extracellular matrix, leading to expansion of mesangial matrix, thickening of glomerular basement membrane and tubulointerstitial fibrosis. At the molecular level, accumulating evidence suggests that hyperglycemia or high glucose mediates renal injury in DN via multiple molecular mechanisms such as induction of oxidative stress, upregulation of renal transforming growth factor beta-1 expression, production of proinflammatory cytokines, activation of fibroblasts and renin angiotensin system, and depletion of adenosine triphosphate. Also worrying is the fact that existing therapies only retard the disease progression but do not prevent it. Therefore, there is urgent need to identify novel therapies to target additional disease mechanisms. Hydrogen sulfide (H2S), the third member of the gasotransmitter family, has recently been identified and demonstrated to possess important therapeutic characteristics that prevent the development and progression of DN in experimental animals by targeting several important molecular pathways, and therefore may represent an alternative or additional therapeutic approach for DN. This review discusses recent experimental findings on the molecular mechanisms underlying the therapeutic effects of H2S against the development and progression of DN and its clinical application in the future.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
38
|
Shin SJ, Chung S, Kim SJ, Lee EM, Yoo YH, Kim JW, Ahn YB, Kim ES, Moon SD, Kim MJ, Ko SH. Effect of Sodium-Glucose Co-Transporter 2 Inhibitor, Dapagliflozin, on Renal Renin-Angiotensin System in an Animal Model of Type 2 Diabetes. PLoS One 2016; 11:e0165703. [PMID: 27802313 PMCID: PMC5089752 DOI: 10.1371/journal.pone.0165703] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
Background Renal renin-angiotensin system (RAS) activation is one of the important pathogenic mechanisms in the development of diabetic nephropathy in type 2 diabetes. The aim of this study was to investigate the effects of a sodium-glucose co-transporter 2 (SGLT-2) inhibitor, dapagliflozin, on renal RAS in an animal model with type 2 diabetes. Methods Dapagliflozin (1.0 mg/kg, OL-DA) or voglibose (0.6 mg/kg, OL-VO, diabetic control) (n = 10 each) was administered to Otsuka Long-Evans Tokushima Fatty (OLETF) rats for 12 weeks. We used voglibose, an alpha-glucosidase inhibitor, as a comparable counterpart to SGLT2 inhibitor because of its postprandial glucose-lowering effect without proven renoprotective effects. Control Long-Evans Tokushima Otsuka (LT) and OLETF (OL-C) rats received saline (n = 10, each). Changes in blood glucose, urine albumin, creatinine clearance, and oxidative stress were measured. Inflammatory cell infiltration, mesangial widening, and interstitial fibrosis in the kidney were evaluated by histological analysis. The effects of dapagliflozin on renal expression of the RAS components were evaluated by quantitative RT-PCR in renal tissue. Results After treatment, hyperglycemia and urine microalbumin levels were attenuated in both OL-DA and OL-VO rather than in the OL-C group (P < 0.05). The urine angiotensin II (Ang II) and angiotensinogen levels were significantly decreased following treatment with dapagliflozin or voglibose, but suppression of urine Ang II level was more prominent in the OL-DA than the OL-VO group (P < 0.05). The expressions of angiotensin type 1 receptor and tissue oxidative stress markers were markedly increased in OL-C rats, which were reversed by dapagliflozin or voglibose (P < 0.05, both). Inflammatory cell infiltration, mesangial widening, interstitial fibrosis, and total collagen content were significantly increased in OL-C rats, which were attenuated in OL-DA group (P < 0.05). Conclusion Dapagliflozin treatment showed beneficial effects on diabetic nephropathy, which might be via suppression of renal RAS component expression, oxidative stress and interstitial fibrosis in OLETF rats. We suggest that, in addition to control of hyperglycemia, partial suppression of renal RAS with an SGLT2 inhibitor would be a promising strategy for the prevention of treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Seok Joon Shin
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sungjin Chung
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Soo Jung Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun-Mi Lee
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young-Hye Yoo
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji-Won Kim
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yu-Bae Ahn
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun-Sook Kim
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung-Dae Moon
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Myung-Jun Kim
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Hyun Ko
- Division of Endocrinology & Metabolism, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- * E-mail:
| |
Collapse
|
39
|
Jha JC, Banal C, Chow BSM, Cooper ME, Jandeleit-Dahm K. Diabetes and Kidney Disease: Role of Oxidative Stress. Antioxid Redox Signal 2016; 25:657-684. [PMID: 26906673 PMCID: PMC5069735 DOI: 10.1089/ars.2016.6664] [Citation(s) in RCA: 449] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Intrarenal oxidative stress plays a critical role in the initiation and progression of diabetic kidney disease (DKD). Enhanced oxidative stress results from overproduction of reactive oxygen species (ROS) in the context of concomitant, insufficient antioxidant pathways. Renal ROS production in diabetes is predominantly mediated by various NADPH oxidases (NOXs), but a defective antioxidant system as well as mitochondrial dysfunction may also contribute. Recent Advances: Effective agents targeting the source of ROS generation hold the promise to rescue the kidney from oxidative damage and prevent subsequent progression of DKD. Critical Issues and Future Directions: In the present review, we summarize and critically analyze molecular and cellular mechanisms that have been demonstrated to be involved in NOX-induced renal injury in diabetes, with particular focus on the role of increased glomerular injury, the development of albuminuria, and tubulointerstitial fibrosis, as well as mitochondrial dysfunction. Furthermore, novel agents targeting NOX isoforms are discussed. Antioxid. Redox Signal. 25, 657-684.
Collapse
Affiliation(s)
- Jay C Jha
- 1 Diabetic Complications Division, JDRF Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart and Diabetes Institute , Melbourne, Australia
| | - Claudine Banal
- 1 Diabetic Complications Division, JDRF Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart and Diabetes Institute , Melbourne, Australia
| | - Bryna S M Chow
- 1 Diabetic Complications Division, JDRF Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart and Diabetes Institute , Melbourne, Australia
| | - Mark E Cooper
- 1 Diabetic Complications Division, JDRF Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart and Diabetes Institute , Melbourne, Australia .,2 Department of Medicine, Monash University , Melbourne, Australia
| | - Karin Jandeleit-Dahm
- 1 Diabetic Complications Division, JDRF Danielle Alberti Memorial Centre for Diabetic Complications, Baker IDI Heart and Diabetes Institute , Melbourne, Australia .,2 Department of Medicine, Monash University , Melbourne, Australia
| |
Collapse
|
40
|
Simão S, Santos DF, Silva GA. Aliskiren inhibits the renin-angiotensin system in retinal pigment epithelium cells. Eur J Pharm Sci 2016; 92:22-7. [DOI: 10.1016/j.ejps.2016.06.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 01/06/2023]
|
41
|
Katsoulieris EN, Drossopoulou GI, Kotsopoulou ES, Vlahakos DV, Lianos EA, Tsilibary EC. High Glucose Impairs Insulin Signaling in the Glomerulus: An In Vitro and Ex Vivo Approach. PLoS One 2016; 11:e0158873. [PMID: 27434075 PMCID: PMC4951020 DOI: 10.1371/journal.pone.0158873] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/23/2016] [Indexed: 01/14/2023] Open
Abstract
Objective Chronic hyperglycaemia, as seen in type II diabetes, results in both morphological and functional impairments of podocytes in the kidney. We investigated the effects of high glucose (HG) on the insulin signaling pathway, focusing on cell survival and apoptotic markers, in immortalized human glomerular cells (HGEC; podocytes) and isolated glomeruli from healthy rats. Methods and Findings HGEC and isolated glomeruli were cultured for various time intervals under HG concentrations in the presence or absence of insulin. Our findings indicated that exposure of HGEC to HG led to downregulation of all insulin signaling markers tested (IR, p-IR, IRS-1, p-Akt, p-Fox01,03), as well as to increased sensitivity to apoptosis (as seen by increased PARP cleavage, Casp3 activation and DNA fragmentation). Short insulin pulse caused upregulation of insulin signaling markers (IR, p-IR, p-Akt, p-Fox01,03) in a greater extent in normoglycaemic cells compared to hyperglycaemic cells and for the case of p-Akt, in a PI3K-dependent manner. IRS-1 phosphorylation of HG-treated podocytes was negatively regulated, favoring serine versus tyrosine residues. Prolonged insulin treatment caused a significant decrease of IR levels, while alterations in glucose concentrations for various time intervals demonstrated changes of IR, p-IR and p-Akt levels, suggesting that the IR signaling pathway is regulated by glucose levels. Finally, HG exerted similar effects in isolated glomeruli. Conclusions These results suggest that HG compromises the insulin signaling pathway in the glomerulus, promoting a proapoptotic environment, with a possible critical step for this malfunction lying at the level of IRS-1 phosphorylation; thus we herein demonstrate glomerular insulin signaling as another target for investigation for the prevention and/ or treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Elias N. Katsoulieris
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
| | - Garyfalia I. Drossopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
- * E-mail: (GID); (ECT)
| | - Eleni S. Kotsopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
| | - Dimitrios V. Vlahakos
- 2nd Department of Propaedeutic Medicine, Attikon University Hospital, Athens, Greece
| | - Elias A. Lianos
- Department of Pathology, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Effie C. Tsilibary
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
- * E-mail: (GID); (ECT)
| |
Collapse
|
42
|
Mao N, Tan RZ, Wang SQ, Wei C, Shi XL, Fan JM, Wang L. Ginsenoside Rg1 inhibits angiotensin II-induced podocyte autophagy via AMPK/mTOR/PI3K pathway. Cell Biol Int 2016; 40:917-25. [PMID: 27296076 DOI: 10.1002/cbin.10634] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/09/2016] [Indexed: 01/07/2023]
Abstract
Recent researches have reported the extensive pharmacological activities of Ginsenoside Rg1 including antioxidant, anti-inflammatory, and anticancer properties. Furthermore Rg1 was also shown to protect various kinds of cells from self-digestion by its anti-autophagy activity. In previous studies, angiotensin II (Ang II), a key mediator of renin-angiotensin system, has been demonstrated to contribute to the progression of renal injury including abnormal autophagy. However, whether Rg1 can relieve Ang II-induced autophagy in podocyte as well as the underlying molecular mechanism remains to be elucidated. Here, we employed Ang II-treated podocyte as a model to investigate the effect of Rg1 on autophagy and the involved signal pathways. In the present study, we found that Ang II strongly promoted autophagy in immortalized mouse podocyte cells by observing the formation of autophagosomes and detecting the expression of autophagic marker, for example, LC3-II. Notably, compared to the Ang II-treated cells, treatment with Rg1 significantly inhibited the formation of autophagosomes and expression of autophagy-related proteins in Ang II pre-treated podocyte. Meanwhile, Rg1 downregulated the activity of AMPK and GSK-3β and upregulated the activity of P70S6K in Ang II-treated podocyte. In conclusion, these findings demonstrate that Ang II promotes autophagy in podocyte, and Rg1 effectively attenuates this process through AMPK/mTOR/PI3K pathway, suggesting that Rg1 may be beneficial to alleviate podocyte injury.
Collapse
Affiliation(s)
- Nan Mao
- Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Rui-Zhi Tan
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shao-Qing Wang
- Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Cong Wei
- Clinical Laboratory, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin-Li Shi
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Hebei, Shijiazhuang, 050200, China
| | - Jun-Ming Fan
- Department of Nephrology, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Li Wang
- Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| |
Collapse
|
43
|
Mariana CP, Ramona PA, Ioana BC, Diana M, Claudia RC, Stefan VD, Maria KI. Urinary angiotensin converting enzyme 2 is strongly related to urinary nephrin in type 2 diabetes patients. Int Urol Nephrol 2016; 48:1491-7. [PMID: 27312782 DOI: 10.1007/s11255-016-1334-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/23/2016] [Indexed: 01/27/2023]
Abstract
PURPOSE Podocyte lesion is recently recognized as an early event in diabetic kidney disease (DKD) and is reflected by urinary (u) nephrin (Neph) shedding. Angiotensin II plays an important role in podocyte dysfunction of diabetes. Angiotensin converting enzyme 2 (ACE2) is the main ACE variant in podocytes and counteracts deleterious angiotensin II effects. We assessed for the first time the relation of uACE2 and uNeph in type 2 diabetes subjects. MATERIAL AND METHOD Seventy-five type 2 diabetes patients were included in a transversal study. History, clinical and laboratory data, urinary albumin-to-creatinine ratio (uACR), and ELISA determination of uNeph and uACE2 were obtained. RESULTS uNeph was 349.00 ± 133.42 pg/ml, and uACE2 was 45.50 (36.35-62.60) pg/ml. uNeph correlated to uACE2 (r = 0.44, p < 0.001) and to uACR (r = 0.25, p = 0.032). In multivariate regression, introducing parameters that are known to be related to DKD, uACE2 (p < 0.0001), LDL cholesterol (p = 0.02) and glycated hemoglobin (p = 0.03) remained significant predictors of uNeph. Normoalbuminuric patients had lower uNeph than patients with uACR > 30 mg/g (325.50 ± 135.45 vs 391.03 ± 121.40 pg/ml, p = 0.04); they also had a tendency versus lower uACE2 [41.40 (34.30-60.65) vs 52.57 (37.95-69.85) pg/ml, p = 0.06]. A cutoff for uNeph of 451.6 pg/ml was derived from the ROC curve analysis; uACE2 was the main determinant for uNeph being above or below this cutoff-OR = 1.09; 95 %CI (1.04-1.15), p = 0.001. Patients taking blockers of the renin angiotensin system had similar uNeph and uACE2. uNeph and uACE2 were not influenced by renal function. CONCLUSION uNeph is significantly correlated to uACE2 and uACR in type 2 diabetes patients.
Collapse
Affiliation(s)
- Ciorba Pop Mariana
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj, 3-5 Clinicilor Street, 400006, Cluj-Napoca, Romania
| | - Potra Alina Ramona
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj, 3-5 Clinicilor Street, 400006, Cluj-Napoca, Romania.
| | - Bondor Cosmina Ioana
- Department of Informatics and Biostatistics, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj, 6 Pasteur Street, 400349, Cluj-Napoca, Romania
| | - Moldovan Diana
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj, 3-5 Clinicilor Street, 400006, Cluj-Napoca, Romania
| | - Rusu Crina Claudia
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj, 3-5 Clinicilor Street, 400006, Cluj-Napoca, Romania
| | - Vladutiu Dan Stefan
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj, 3-5 Clinicilor Street, 400006, Cluj-Napoca, Romania
| | - Kacso Ina Maria
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj, 3-5 Clinicilor Street, 400006, Cluj-Napoca, Romania
| |
Collapse
|
44
|
Kidokoro K, Satoh M, Itano S, Kuwabara A, Sasaki T, Kashihara N. Feasibility of fluorescence energy transfer system for imaging the renoprotective effects of aliskiren in diabetic mice. J Renin Angiotensin Aldosterone Syst 2016; 17:1470320315625704. [PMID: 27216079 PMCID: PMC5841571 DOI: 10.1177/1470320315625704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 11/21/2015] [Indexed: 01/13/2023] Open
Abstract
Introduction: We investigated the feasibility of using a fluorescence resonance energy transfer system to image enzymatic activity in order to evaluate the effects of aliskiren (a direct renin inhibitor) on diabetic nephropathy. Materials and methods: First, we induced diabetes in C57BL/6J mice using streptozotocin, then treated them with either aliskiren (25 mg/kg/day) or the angiotensin type 1 receptor blocker valsartan (15 mg/kg/day) for four weeks. Finally, we utilized renin fluorescence resonance energy transfer substrate to assess renin activity. Results: Renin activity was much higher in the kidneys of diabetic mice compared to those of the non-diabetic control mice. While aliskiren inhibited this activity, valsartan did not. We noted that production of reactive oxygen species intensified and the bioavailability of nitric oxide diminished in the glomeruli of diabetic mice. Aliskiren and valsartan significantly ameliorated these effects. They suppressed glomerular production of reactive oxygen species and urinary albumin excretion. In fact, urinary albumin excretion in diabetic mice treated with aliskiren or valsartan was lower than that in untreated diabetic mice. Furthermore, aliskiren and valsartan significantly reduced glomerular permeability by maintaining the glomerular endothelial surface layer. Conclusion: Fluorescence resonance energy transfer could provide a new tool for evaluating tissue and plasma enzymatic activity.
Collapse
Affiliation(s)
- Kengo Kidokoro
- Department of Nephrology and Hypertension, Kawasaki Medical School, Japan
| | - Minoru Satoh
- Department of Nephrology and Hypertension, Kawasaki Medical School, Japan
| | - Seiji Itano
- Department of Nephrology and Hypertension, Kawasaki Medical School, Japan
| | - Atsunori Kuwabara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Japan
| | - Tamaki Sasaki
- Department of Nephrology and Hypertension, Kawasaki Medical School, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Japan
| |
Collapse
|
45
|
JI ZHENZHONG, XU YANCHENG. Melatonin protects podocytes from angiotensin II-induced injury in an in vitro diabetic nephropathy model. Mol Med Rep 2016; 14:920-6. [DOI: 10.3892/mmr.2016.5313] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 05/06/2016] [Indexed: 11/06/2022] Open
|
46
|
Tomilin V, Mamenko M, Zaika O, Pochynyuk O. Role of renal TRP channels in physiology and pathology. Semin Immunopathol 2016; 38:371-383. [PMID: 26385481 PMCID: PMC4798925 DOI: 10.1007/s00281-015-0527-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 09/04/2015] [Indexed: 01/05/2023]
Abstract
Kidneys critically contribute to the maintenance of whole-body homeostasis by governing water and electrolyte balance, controlling extracellular fluid volume, plasma osmolality, and blood pressure. Renal function is regulated by numerous systemic endocrine and local mechanical stimuli. Kidneys possess a complex network of membrane receptors, transporters, and ion channels which allows responding to this wide array of signaling inputs in an integrative manner. Transient receptor potential (TRP) channel family members with diverse modes of activation, varied permeation properties, and capability to integrate multiple downstream signals are pivotal molecular determinants of renal function all along the nephron. This review summarizes experimental data on the role of TRP channels in a healthy mammalian kidney and discusses their involvement in renal pathologies.
Collapse
Affiliation(s)
- Viktor Tomilin
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX, 77030, USA
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russian Federation
| | - Mykola Mamenko
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX, 77030, USA
| | - Oleg Zaika
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX, 77030, USA
| | - Oleh Pochynyuk
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, 6431 Fannin, Houston, TX, 77030, USA.
| |
Collapse
|
47
|
Huang F, Wang Q, Ma X, Wu L, Guo F, Qin G. Valsartan inhibits amylin-induced podocyte damage. Microvasc Res 2016; 106:101-9. [PMID: 27102209 DOI: 10.1016/j.mvr.2016.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 03/19/2016] [Accepted: 04/16/2016] [Indexed: 01/01/2023]
Abstract
Previous studies have described the deposition of amylin in the kidney of patients with type 2 diabetes mellitus (T2DM). These deposits play a critical role in the pathogenesis of diabetic nephropathy (DN), although the mechanism underlying this effect is unknown. Thus, this study was undertaken to investigate whether amylin aggregation stimulates the local angiotensin II type 1 receptor (AT1R) in podocytes, and to examine its role in podocyte apoptosis. Amylin-induced apoptosis was investigated in vitro in differentiated, conditionally immortalized mouse podocytes and in vivo in KM mice. Expression of genes including nephrin, podocin, AT1R and desmin was measured through quantitative real time PCR, western blot and immunohistochemistry. Apoptosis was determined by flow cytometry, while the cellular distribution of podocin and nephrin was investigated by immunofluorescence. The ultra-structure of glomeruli was examined by transmission electron microscopy (TEM). Amylin enhanced apoptosis in a dose-dependent manner in vitro. The peptide also suppressed podocin and nephrin expression, but enhanced that of AT1R and desmin. Both effects were significantly blocked by valsartan, which inhibits angiotensin II type 1 receptor. These findings suggest that amylin activates a local intracellular RAS in podocytes and induces damage and apoptosis.
Collapse
Affiliation(s)
- Fengjuan Huang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qingzhu Wang
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaojun Ma
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lina Wu
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Feng Guo
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Guijun Qin
- Division of Endocrinology, Department of Internal Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
48
|
Podocyte injury in diabetic nephropathy: implications of angiotensin II – dependent activation of TRPC channels. Sci Rep 2015. [DOI: 10.1038/srep17637 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
AbstractInjury to podocytes is considered a major contributor to diabetic kidney disease: their loss causes proteinuria and progressive glomerulosclerosis. Podocyte depletion may result from improper calcium handling due to abnormal activation of the calcium permeant TRPC (Transient Receptor Potential Canonical) channels. Angiotensin II (Ang II) levels are found to be elevated in diabetes; furthermore, it was reported that Ang II causes activation of TRPC6 in podocytes. We hypothesized here that Ang II-mediated calcium influx is aggravated in the podocytes under the conditions of type 1 diabetic nephropathy (DN). Diabetes was induced in the Dahl Salt-Sensitive rats by an injection of streptozotocin (STZ-SS). Eleven weeks post treatment was sufficient for the animals to develop hyperglycemia, excessive urination, weight loss, microalbuminuria, nephrinuria and display renal histological lesions typical for patients with DN. Patch-clamp electrophysiology performed on podocytes of the freshly isolated glomeruli showed enhanced basal TRPC channel activity in the STZ-SS rats and increased response to Ang II; total calcium influx triggered by Ang II application was also augmented in podocytes of these rats. Our studies have a strong potential for advancing the understanding of TRPC-mediated effects on podocytopenia in DN initiation.
Collapse
|
49
|
Ilatovskaya DV, Levchenko V, Lowing A, Shuyskiy LS, Palygin O, Staruschenko A. Podocyte injury in diabetic nephropathy: implications of angiotensin II – dependent activation of TRPC channels. Sci Rep 2015. [DOI: 10.1038/srep17637 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
AbstractInjury to podocytes is considered a major contributor to diabetic kidney disease: their loss causes proteinuria and progressive glomerulosclerosis. Podocyte depletion may result from improper calcium handling due to abnormal activation of the calcium permeant TRPC (Transient Receptor Potential Canonical) channels. Angiotensin II (Ang II) levels are found to be elevated in diabetes; furthermore, it was reported that Ang II causes activation of TRPC6 in podocytes. We hypothesized here that Ang II-mediated calcium influx is aggravated in the podocytes under the conditions of type 1 diabetic nephropathy (DN). Diabetes was induced in the Dahl Salt-Sensitive rats by an injection of streptozotocin (STZ-SS). Eleven weeks post treatment was sufficient for the animals to develop hyperglycemia, excessive urination, weight loss, microalbuminuria, nephrinuria and display renal histological lesions typical for patients with DN. Patch-clamp electrophysiology performed on podocytes of the freshly isolated glomeruli showed enhanced basal TRPC channel activity in the STZ-SS rats and increased response to Ang II; total calcium influx triggered by Ang II application was also augmented in podocytes of these rats. Our studies have a strong potential for advancing the understanding of TRPC-mediated effects on podocytopenia in DN initiation.
Collapse
|
50
|
Ilatovskaya DV, Levchenko V, Lowing A, Shuyskiy LS, Palygin O, Staruschenko A. Podocyte injury in diabetic nephropathy: implications of angiotensin II – dependent activation of TRPC channels. Sci Rep 2015. [DOI: 10.1038/srep17637 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
AbstractInjury to podocytes is considered a major contributor to diabetic kidney disease: their loss causes proteinuria and progressive glomerulosclerosis. Podocyte depletion may result from improper calcium handling due to abnormal activation of the calcium permeant TRPC (Transient Receptor Potential Canonical) channels. Angiotensin II (Ang II) levels are found to be elevated in diabetes; furthermore, it was reported that Ang II causes activation of TRPC6 in podocytes. We hypothesized here that Ang II-mediated calcium influx is aggravated in the podocytes under the conditions of type 1 diabetic nephropathy (DN). Diabetes was induced in the Dahl Salt-Sensitive rats by an injection of streptozotocin (STZ-SS). Eleven weeks post treatment was sufficient for the animals to develop hyperglycemia, excessive urination, weight loss, microalbuminuria, nephrinuria and display renal histological lesions typical for patients with DN. Patch-clamp electrophysiology performed on podocytes of the freshly isolated glomeruli showed enhanced basal TRPC channel activity in the STZ-SS rats and increased response to Ang II; total calcium influx triggered by Ang II application was also augmented in podocytes of these rats. Our studies have a strong potential for advancing the understanding of TRPC-mediated effects on podocytopenia in DN initiation.
Collapse
|