1
|
Cho A, Jin W, Lee J, Shin N, Lee MS, Li L, Yang SH, Park KS, Yang CW, Kim DK, Oh YK, Lim CS, Lee JP. Periostin deficiency attenuates kidney fibrosis in diabetic nephropathy by improving pancreatic β-cell dysfunction and reducing kidney EMT. Sci Rep 2023; 13:17599. [PMID: 37845302 PMCID: PMC10579313 DOI: 10.1038/s41598-023-44177-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023] Open
Abstract
Diabetic nephropathy (DN) is associated with kidney fibrosis. A previous study revealed that periostin (POSTN) contributes to kidney fibrosis. This study examined the role of POSTN in DN. The urinary concentrations of POSTN and TNC increased according to the severity of DN in human samples. Streptozotocin (STZ) was administered after unilateral nephrectomy (UNXSTZ) to induce DN in wild-type and Postn-null mice. Four experimental groups were generated: wild-typeham (WT Sham), wild-type UNXSTZ (WT STZ), Postn-null Sham (KO Sham), and Postn-null UNXSTZ (KO STZ). After 20 weeks, the KO STZ group had lower levels of urine albumin excretion, glomerular sclerosis, and interstitial fibrosis than those of the WT STZ group. Additionally, the KO STZ group had lower expression of fibrosis markers, including TNC. The KO STZ group showed better glucose regulation than the WT STZ model. Furthermore, the KO STZ group exhibited significantly preserved pancreatic islet integrity and insulin expression. HK-2 cells were used to observe the aggravation of fibrosis caused by POSTN under TGF-β conditions. We stimulated INS-1 cells with streptozotocin and evaluated the viability of these cells. The anti-POSTN antibody treatment of INS-1 cells with streptozotocin resulted in higher cell viability than that with treatment with streptozotocin alone. The absence of POSTN in DN contributes to renal fibrosis alleviation by improving pancreatic β-cell function. Additionally, there is an association between POSTN and TNC.
Collapse
Affiliation(s)
- Ara Cho
- Translational Medicine Major, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wencheng Jin
- Department of Internal Medicine, Seoul National University Boramae Medical Center, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeonghwan Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Nayeon Shin
- Department of Internal Medicine, Seoul National University Boramae Medical Center, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Republic of Korea
| | - Myoung Seok Lee
- Department of Radiology, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Lilin Li
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Critical Care Medicine, Yanbian University Hospital, Yanji, Jilin, China
| | - Seung Hee Yang
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Kidney Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul Woo Yang
- Transplantation Research Center, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University Boramae Medical Center, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Pyo Lee
- Translational Medicine Major, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul National University Boramae Medical Center, 20, Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Republic of Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Talukdar A, Basumatary M. Rodent models to study type 1 and type 2 diabetes induced human diabetic nephropathy. Mol Biol Rep 2023; 50:7759-7782. [PMID: 37458869 DOI: 10.1007/s11033-023-08621-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/21/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Diabetic nephropathy (DN), an outcome of prolonged diabetes, has affected millions of people worldwide and every year the incidence and prevalence increase substantially. The symptoms may start with mild manifestations of the disease such as increased albuminuria, serum creatinine levels, thickening of glomerular basement membrane, expansion of mesangial matrix to severe pathological symptoms such as glomerular lesions and tubulointerstitial fibrosis which may further proceed to cardiovascular dysfunction or end-stage renal disease. PERSPECTIVE Numerous therapeutic interventions are being explored for the management of DN, however, these interventions do not completely halt the progression of this disease and hence animal models are being explored to identify critical genetic and molecular parameters which could help in tackling the disease. Rodent models which mostly include mice and rats are commonly used experimental animals which provide a wide range of advantages in understanding the onset and progression of disease in humans and also their response to a wide range of interventions helps in the development of effective therapeutics. Rodent models of type 1 and type 2 diabetes induced DN have been developed utilizing different platforms and interventions during the last few decades some of which mimic various stages of diabetes ranging from early to later stages. However, a rodent model which replicates all the features of human DN is still lacking. This review tries to evaluate the rodent models that are currently available and understand their features and limitations which may help in further development of more robust models of human DN. CONCLUSION Using these rodent models can help to understand different aspects of human DN although further research is required to develop more robust models utilizing diverse genetic platforms which may, in turn, assist in developing effective interventions to target the disease at different levels.
Collapse
Affiliation(s)
- Amit Talukdar
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam, 784028, India.
| | - Mandira Basumatary
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam, 784028, India
| |
Collapse
|
3
|
Ma XL, Shi QY, Zhao QG, Xu Q, Yan SS, Han BX, Fang C, Zhang L, Pei YF. Causal associations between type 1 diabetes and COVID-19 infection and prognosis: a two-sample Mendelian randomization study. BMJ Open Diabetes Res Care 2023; 11:e003167. [PMID: 37311601 DOI: 10.1136/bmjdrc-2022-003167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 05/20/2023] [Indexed: 06/15/2023] Open
Abstract
INTRODUCTION It has been suggested that type 1 diabetes was associated with increased COVID-19 morbidity and mortality. However, their causal relationship is still unclear. Herein, we performed a two-sample Mendelian randomization (MR) to investigate the causal effect of type 1 diabetes on COVID-19 infection and prognosis. RESEARCH DESIGN AND METHODS The summary statistics of type 1 diabetes were obtained from two published genome-wide association studies of European population, one as a discovery sample including 15 573 cases and 158 408 controls, and the other data as a replication sample consisting of 5913 cases and 8828 controls. We first performed a two-sample MR analysis to evaluate the causal effect of type 1 diabetes on COVID-19 infection and prognosis. Then, reverse MR analysis was conducted to determine whether reverse causality exists. RESULTS MR analysis results showed that the genetically predicted type 1 diabetes was associated with higher risk of severe COVID-19 (OR=1.073, 95% CI: 1.034 to 1.114, pFDR=1.15×10-3) and COVID-19 death (OR=1.075, 95% CI: 1.033 to 1.119, pFDR=1.15×10-3). Analysis of replication dataset showed similar results, namely a positive association between type 1 diabetes and severe COVID-19 (OR=1.055, 95% CI: 1.029 to 1.081, pFDR=1.59×10-4), and a positively correlated association with COVID-19 death (OR=1.053, 95% CI: 1.026 to 1.081, pFDR=3.50×10-4). No causal association was observed between type 1 diabetes and COVID-19 positive, hospitalized COVID-19, the time to the end of COVID-19 symptoms in the colchicine treatment group and placebo treatment group. Reverse MR analysis showed no reverse causality. CONCLUSIONS Type 1 diabetes had a causal effect on severe COVID-19 and death after COVID-19 infection. Further mechanistic studies are needed to explore the relationship between type 1 diabetes and COVID-19 infection and prognosis.
Collapse
Affiliation(s)
- Xin-Ling Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Qi-Yun Shi
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi-Gang Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Qian Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Shan-Shan Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Bai-Xue Han
- Department of Epidemiology and Biostatistics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Chen Fang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yu-Fang Pei
- Department of Epidemiology and Biostatistics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Dos Anjos AA, de Paiva IT, Simões Lima GL, da Silva Filha R, Fróes BPE, Brant Pinheiro SV, Silva ACSE. Nephrotic Syndrome and Renin-angiotensin System: Pathophysiological Role and Therapeutic Potential. Curr Mol Pharmacol 2023; 16:465-474. [PMID: 35713131 DOI: 10.2174/1874467215666220616152312] [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: 02/02/2022] [Revised: 05/14/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022]
Abstract
Idiopathic Nephrotic Syndrome (INS) is the most frequent etiology of glomerulopathy in pediatric patients and one of the most common causes of chronic kidney disease (CKD) and end-stage renal disease (ESRD) in this population. In this review, we aimed to summarize evidence on the pathophysiological role and therapeutic potential of the Renin-Angiotensin System (RAS) molecules for the control of proteinuria and for delaying the onset of CKD in patients with INS. This is a narrative review in which the databases PubMed, Web of Science, and Sci- ELO were searched for articles about INS and RAS. We selected articles that evaluated the pathophysiological role of RAS and the effects of the alternative RAS axis as a potential therapy for INS. Several studies using rodent models of nephropathies showed that the treatment with activators of the Angiotensin-Converting Enzyme 2 (ACE2) and with Mas receptor agonists reduces proteinuria and improves kidney tissue damage. Another recent paper showed that the reduction of urinary ACE2 levels in children with INS correlates with proteinuria and higher concentrations of inflammatory cytokines, although data with pediatric patients are still limited. The molecules of the alternative RAS axis comprise a wide spectrum, not yet fully explored, of potential pharmacological targets for kidney diseases. The effects of ACE2 activators and receptor Mas agonists show promising results that can be useful for nephropathies including INS.
Collapse
Affiliation(s)
- Alessandra Aguiar Dos Anjos
- Departamento de Pediatria, Faculdade de Medicina, Unidade de Nefrologia Pediátrica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Isadora Tucci de Paiva
- Departamento de Pediatria, Faculdade de Medicina, Unidade de Nefrologia Pediátrica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Giovanna Letícia Simões Lima
- Faculdade de Medicina, Laboratório Interdisciplinar de Investigação Médica, UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Roberta da Silva Filha
- Faculdade de Medicina, Laboratório Interdisciplinar de Investigação Médica, UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Brunna Pinto E Fróes
- Departamento de Pediatria, Faculdade de Medicina, Unidade de Nefrologia Pediátrica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Sérgio Veloso Brant Pinheiro
- Departamento de Pediatria, Faculdade de Medicina, Unidade de Nefrologia Pediátrica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Ana Cristina Simões E Silva
- Departamento de Pediatria, Faculdade de Medicina, Unidade de Nefrologia Pediátrica, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Faculdade de Medicina, Laboratório Interdisciplinar de Investigação Médica, UFMG, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
5
|
Palau V, Villanueva S, Jarrín J, Benito D, Márquez E, Rodríguez E, Soler MJ, Oliveras A, Gimeno J, Sans L, Crespo M, Pascual J, Barrios C, Riera M. Redefining the Role of ADAM17 in Renal Proximal Tubular Cells and Its Implications in an Obese Mouse Model of Pre-Diabetes. Int J Mol Sci 2021; 22:ijms222313093. [PMID: 34884897 PMCID: PMC8657896 DOI: 10.3390/ijms222313093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022] Open
Abstract
Acute and chronic kidney lesions induce an increase in A Disintegrin And Metalloproteinase domain 17 (ADAM17) that cleaves several transmembrane proteins related to inflammatory and fibrotic pathways. Our group has demonstrated that renal ADAM17 is upregulated in diabetic mice and its inhibition decreases renal inflammation and fibrosis. The purpose of the present study was to analyze how Adam17 deletion in proximal tubules affects different renal structures in an obese mice model. Tubular Adam17 knockout male mice and their controls were fed a high-fat diet (HFD) for 22 weeks. Glucose tolerance, urinary albumin-to-creatinine ratio, renal histology, and pro-inflammatory and pro-fibrotic markers were evaluated. Results showed that wild-type mice fed an HFD became obese with glucose intolerance and renal histological alterations mimicking a pre-diabetic condition; consequently, greater glomerular size and mesangial expansion were observed. Adam17 tubular deletion improved glucose tolerance and protected animals against glomerular injury and prevented podocyte loss in HFD mice. In addition, HFD mice showed more glomerular macrophages and collagen accumulation, which was prevented by Adam17 deletion. Galectin-3 expression increased in the proximal tubules and glomeruli of HFD mice and ameliorated with Adam17 deletion. In conclusion, Adam17 in proximal tubules influences glucose tolerance and participates in the kidney injury in an obese pre-diabetic murine model. The role of ADAM17 in the tubule impacts on glomerular inflammation and fibrosis.
Collapse
Affiliation(s)
- Vanesa Palau
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Sofia Villanueva
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Josué Jarrín
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - David Benito
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Eva Márquez
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Eva Rodríguez
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - María José Soler
- Nephrology Research Group, Vall d’Hebron Research Institute (VHIR), Nephrology Department, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Anna Oliveras
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Javier Gimeno
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain;
| | - Laia Sans
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Clara Barrios
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
- Correspondence: (C.B.); (M.R.); Tel.: +34-65-004-2149 (C.B.); +34-93-316-0626 (M.R.)
| | - Marta Riera
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
- Correspondence: (C.B.); (M.R.); Tel.: +34-65-004-2149 (C.B.); +34-93-316-0626 (M.R.)
| |
Collapse
|
6
|
Wang T, Wen X, Zhang Z, Xie M, Zhou J. Phillyrin ameliorates diabetic nephropathy through the PI3K/Akt/GSK-3β signalling pathway in streptozotocin-induced diabetic mice. Hum Exp Toxicol 2021; 40:S487-S496. [PMID: 34649470 DOI: 10.1177/09603271211051598] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Diabetic nephropathy is a progressive kidney disease resulting from long-term hyperglycaemia in diabetic patients, and the underlying mechanism is complex and lacks effective treatments. Various active ingredients in Chinese herbs have been shown to alleviate renal injury and improve DN in recent years. Phillyrin, a natural medicinal active compound extracted from the Oleaceae family, has various pharmacological effects, including antioxidative, antiapoptotic and antiobesity effects. However, the role of phillyrin and its underlying mechanism in DN have not yet been explored. To investigate the effects of phillyrin on DN and its potential mechanisms of action, we performed experiments using streptozotocin (STZ)-induced DN mice as models. Phillyrin significantly reduced the levels of fasting blood glucose (FBG) and glycosylated haemoglobin A1c (HbA1c), downregulated the levels of serum blood urea nitrogen (BUN), serum creatinine (Scr), serum and urine β2-microglobulins (β2-MG) and improved the pathological changes of the kidney in a DN mouse model. Phillyrin also increased the level of antioxidants and attenuated oxidative damage in DN model mice. In addition, phillyrin inhibited Glycogen synthase kinase-3β (GSK-3β) activity by activating the PI3K/Akt signalling pathway, increased the Bcl-2/Bax ratio, reduced the release of cytochrome c from the mitochondria to the cytoplasm, subsequently inhibited the activation of caspase-3 and ultimately suppressed renal cell apoptosis. These findings suggested that phillyrin could be a new promising therapeutic strategy for DN, and this protective effect might be related to suppressing oxidative stress and apoptosis via the PI3K/Akt/GSK-3β pathway.
Collapse
Affiliation(s)
- Tianyang Wang
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Xuejiao Wen
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Ziwen Zhang
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Minjuan Xie
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| | - Jie Zhou
- School of Medicine, 117771Yichun University, Yichun, P.R. China
| |
Collapse
|
7
|
Both Specific Endothelial and Proximal Tubular Adam17 Deletion Protect against Diabetic Nephropathy. Int J Mol Sci 2021; 22:ijms22115520. [PMID: 34073747 PMCID: PMC8197223 DOI: 10.3390/ijms22115520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
ADAM17 is a disintegrin and metalloproteinase capable of cleaving the ectodomains of a diverse variety of molecules including TNF-α, TGF-α, L-selectin, and ACE2. We have previously demonstrated that renal ADAM17 is upregulated in diabetic mice. The role of endothelial (eAdam17) and proximal tubular (tAdam17) Adam17 deletion in renal histology, modulation of the renin angiotensin system (RAS), renal inflammation, and fibrosis was studied in a mouse model of type 1 Diabetes Mellitus. Moreover, the effect of Adam17 deletion in an in vitro 3D cell culture from human proximal tubular cells under high glucose conditions was evaluated. eAdam17 deletion attenuates renal fibrosis and inflammation, whereas tAdam17 deletion decreases podocyte loss, attenuates the RAS, and decreases macrophage infiltration, α-SMA and collagen accumulation. The 3D in vitro cell culture reinforced the findings obtained in tAdam17KO mice with decreased fibrosis in the Adam17 knockout spheroids. In conclusion, Adam17 deletion either in the endothelial or the tubular cells mitigates kidney injury in the diabetic mice by targeting different pathways. The manipulation of Adam17 should be considered as a therapeutic strategy for treating DN.
Collapse
|
8
|
Pathangey G, Fadadu PP, Hospodar AR, Abbas AE. Angiotensin-converting enzyme 2 and COVID-19: patients, comorbidities, and therapies. Am J Physiol Lung Cell Mol Physiol 2021; 320:L301-L330. [PMID: 33237815 PMCID: PMC7938645 DOI: 10.1152/ajplung.00259.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
On March 11, 2020, the World Health Organization declared coronavirus disease 2019 (COVID-19) a pandemic, and the reality of the situation has finally caught up to the widespread reach of the disease. The presentation of the disease is highly variable, ranging from asymptomatic carriers to critical COVID-19. The availability of angiotensin-converting enzyme 2 (ACE2) receptors may reportedly increase the susceptibility and/or disease progression of COVID-19. Comorbidities and risk factors have also been noted to increase COVID-19 susceptibility. In this paper, we hereby review the evidence pertaining to ACE2's relationship to common comorbidities, risk factors, and therapies associated with the susceptibility and severity of COVID-19. We also highlight gaps of knowledge that require further investigation. The primary comorbidities of respiratory disease, cardiovascular disease, renal disease, diabetes, obesity, and hypertension had strong evidence. The secondary risk factors of age, sex, and race/genetics had limited-to-moderate evidence. The tertiary factors of ACE inhibitors and angiotensin II receptor blockers had limited-to-moderate evidence. Ibuprofen and thiazolidinediones had limited evidence.
Collapse
Affiliation(s)
- Girish Pathangey
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
| | | | | | - Amr E Abbas
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
- Department of Cardiovascular Medicine, Beaumont Hospital Royal Oak, Royal Oak, Michigan
| |
Collapse
|
9
|
Wijnant SRA, Jacobs M, Van Eeckhoutte HP, Lapauw B, Joos GF, Bracke KR, Brusselle GG. Expression of ACE2, the SARS-CoV-2 Receptor, in Lung Tissue of Patients With Type 2 Diabetes. Diabetes 2020; 69:2691-2699. [PMID: 33024003 DOI: 10.2337/db20-0669] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/30/2020] [Indexed: 01/01/2023]
Abstract
Increased expression of pulmonary ACE2, the SARS-CoV-2 receptor, could contribute to increased infectivity of COVID-19 in patients with diabetes, but ACE2 expression has not been studied in lung tissue of subjects with diabetes. We therefore studied ACE2 mRNA and protein expression in lung tissue samples of subjects with and without diabetes that were collected between 2002 and 2020 from patients undergoing lobectomy for lung tumors. For RT-PCR analyses, samples from 15 subjects with diabetes were compared with 91 randomly chosen control samples. For immunohistochemical staining, samples from 26 subjects with diabetes were compared with 66 randomly chosen control samples. mRNA expression of ACE2 was measured by quantitative RT-PCR. Protein levels of ACE2 were visualized by immunohistochemistry on paraffin-embedded lung tissue samples and quantified in alveolar and bronchial epithelium. Pulmonary ACE2 mRNA expression was not different between subjects with or without diabetes. In contrast, protein levels of ACE2 were significantly increased in both alveolar tissue and bronchial epithelium of patients with diabetes compared with control subjects, independent of smoking, chronic obstructive pulmonary disease, BMI, renin-angiotensin-aldosterone system inhibitor use, and other potential confounders. To conclude, we show increased bronchial and alveolar ACE2 protein expression in patients with diabetes. Further research is needed to elucidate whether upregulation of ACE2 expression in airways and lungs has consequences on infectivity and clinical outcomes of COVID-19.
Collapse
Affiliation(s)
- Sara R A Wijnant
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Department of Respiratory Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Merel Jacobs
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Hannelore P Van Eeckhoutte
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bruno Lapauw
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Guy F Joos
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Guy G Brusselle
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Respiratory Diseases, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
10
|
Rao S, Ali K, Rivas M, Nugent K. Diabetic Ketoacidosis in Patients With COVID-19. Am J Med Sci 2020; 361:668-669. [PMID: 34024356 PMCID: PMC7686758 DOI: 10.1016/j.amjms.2020.11.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 10/31/2020] [Accepted: 11/23/2020] [Indexed: 11/17/2022]
Affiliation(s)
- Sanjana Rao
- Deaprtment of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, United States
| | - Kiran Ali
- Deaprtment of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, United States
| | - Marcella Rivas
- CoxHealth Adult Medicine & Endocrinology Specialists, Springfield, MO, United States
| | - Kenneth Nugent
- Deaprtment of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, United States.
| |
Collapse
|
11
|
Kaur U, Acharya K, Mondal R, Singh A, Saso L, Chakrabarti S, Chakrabarti SS. Should ACE2 be given a chance in COVID-19 therapeutics: A semi-systematic review of strategies enhancing ACE2. Eur J Pharmacol 2020; 887:173545. [PMID: 32926917 PMCID: PMC7485553 DOI: 10.1016/j.ejphar.2020.173545] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
The severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) has resulted in almost 28 million cases of COVID-19 (Corona virus disease-2019) and more than 900000 deaths worldwide since December 2019. In the absence of effective antiviral therapy and vaccine, treatment of COVID-19 is largely symptomatic. By making use of its spike (S) protein, the virus binds to its primary human cell receptor, angiotensin converting enzyme 2 (ACE2) which is present in the pulmonary epithelial cells as well as other organs. SARS-CoV-2 may cause a downregulation of ACE2. ACE2 plays a protective role in the pulmonary system through its Mas-receptor and alamandine-MrgD-TGR7 pathways. Loss of this protective effect could be a major component of COVID-19 pathogenesis. An attractive strategy in SARS-CoV-2 therapeutics would be to augment ACE2 either directly by supplementation or indirectly through drugs which increase its levels or stimulate its downstream players. In this semi-systematic review, we have analysed the pathophysiological interplay between ACE and ACE2 in the cardiopulmonary system, the modulation of these two proteins by SARS-CoV-2, and potential therapeutic avenues targeting ACE-Ang II and ACE2-Ang (1-7) axes, that can be utilized against COVID-19 disease progression.
Collapse
Affiliation(s)
- Upinder Kaur
- Department of Pharmacology, All India Institute of Medical Sciences, Gorakhpur, UP, India
| | - Kumudini Acharya
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, UP, India
| | - Ritwick Mondal
- Department of Internal Medicine, Institute of Post Graduate Medical Education and Research, Kolkata, WB, India
| | - Amit Singh
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, UP, India
| | - Luciano Saso
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar (deemed to be) University, Mullana, Ambala, Haryana, India.
| | | |
Collapse
|
12
|
SARS-CoV-2 receptor networks in diabetic and COVID-19-associated kidney disease. Kidney Int 2020; 98:1502-1518. [PMID: 33038424 PMCID: PMC7543950 DOI: 10.1016/j.kint.2020.09.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022]
Abstract
COVID-19 morbidity and mortality are increased via unknown mechanisms in patients with diabetes and kidney disease. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) for entry into host cells. Because ACE2 is a susceptibility factor for infection, we investigated how diabetic kidney disease and medications alter ACE2 receptor expression in kidneys. Single cell RNA profiling of kidney biopsies from healthy living donors and patients with diabetic kidney disease revealed ACE2 expression primarily in proximal tubular epithelial cells. This cell-specific localization was confirmed by in situ hybridization. ACE2 expression levels were unaltered by exposures to renin-angiotensin-aldosterone system inhibitors in diabetic kidney disease. Bayesian integrative analysis of a large compendium of public -omics datasets identified molecular network modules induced in ACE2-expressing proximal tubular epithelial cells in diabetic kidney disease (searchable at hb.flatironinstitute.org/covid-kidney) that were linked to viral entry, immune activation, endomembrane reorganization, and RNA processing. The diabetic kidney disease ACE2-positive proximal tubular epithelial cell module overlapped with expression patterns seen in SARS-CoV-2–infected cells. Similar cellular programs were seen in ACE2-positive proximal tubular epithelial cells obtained from urine samples of 13 hospitalized patients with COVID-19, suggesting a consistent ACE2-coregulated proximal tubular epithelial cell expression program that may interact with the SARS-CoV-2 infection processes. Thus SARS-CoV-2 receptor networks can seed further research into risk stratification and therapeutic strategies for COVID-19–related kidney damage.
Collapse
|
13
|
Dambha-Miller H, Albasri A, Hodgson S, Wilcox CR, Khan S, Islam N, Little P, Griffin SJ. Currently prescribed drugs in the UK that could upregulate or downregulate ACE2 in COVID-19 disease: a systematic review. BMJ Open 2020; 10:e040644. [PMID: 32928868 PMCID: PMC7490921 DOI: 10.1136/bmjopen-2020-040644] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/02/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To review evidence on routinely prescribed drugs in the UK that could upregulate or downregulate ACE2 and potentially affect COVID-19 disease. DESIGN Systematic review. DATA SOURCE MEDLINE, EMBASE, CINAHL, the Cochrane Library and Web of Science. STUDY SELECTION Any design with animal or human models examining a currently prescribed UK drug compared with a control, placebo or sham group, and reporting an effect on ACE2 level, activity or gene expression. DATA EXTRACTION AND SYNTHESIS MEDLINE, EMBASE, CINAHL, the Cochrane Library, Web of Science and OpenGrey from inception to 1 April 2020. Methodological quality was assessed using the SYstematic Review Centre for Laboratory animal Experimentation (SYRCLE) risk-of-bias tool for animal studies and Cochrane risk-of-bias tool for human studies. RESULTS We screened 3360 titles and included 112 studies with 21 different drug classes identified as influencing ACE2 activity. Ten studies were in humans and one hundred and two were in animal models None examined ACE2 in human lungs. The most frequently examined drugs were angiotensin receptor blockers (ARBs) (n=55) and ACE inhibitors (ACE-I) (n=22). More studies reported upregulation than downregulation with ACE-I (n=22), ARBs (n=55), insulin (n=8), thiazolidinedione (n=7) aldosterone agonists (n=3), statins (n=5), oestrogens (n=5) calcium channel blockers (n=3) glucagon-like peptide 1 (GLP-1) agonists (n=2) and Non-steroidal anti-inflammatory drugs (NSAIDs) (n=2). CONCLUSIONS There is an abundance of the academic literature and media reports on the potential of drugs that could attenuate or exacerbate COVID-19 disease. This is leading to trials of repurposed drugs and uncertainty among patients and clinicians concerning continuation or cessation of prescribed medications. Our review indicates that the impact of currently prescribed drugs on ACE2 has been poorly studied in vivo, particularly in human lungs where the SARS-CoV-2 virus appears to enact its pathogenic effects. We found no convincing evidence to justify starting or stopping currently prescribed drugs to influence outcomes of COVID-19 disease.
Collapse
Affiliation(s)
- Hajira Dambha-Miller
- Department of Primary Care, University of Southampton, Southampton, UK
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Ali Albasri
- Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Sam Hodgson
- Department of Primary Care, University of Southampton, Southampton, UK
| | | | - Shareen Khan
- Oxford University Hospitals NHS Trust, Oxford, UK
| | - Nazrul Islam
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Department of Population Health, University of Oxford, Oxford, UK
| | - Paul Little
- Department of Primary Care, University of Southampton, Southampton, UK
| | - Simon J Griffin
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Angiotensin-converting enzyme 2 influences pancreatic and renal function in diabetic mice. J Transl Med 2020; 100:1169-1183. [PMID: 32472097 DOI: 10.1038/s41374-020-0440-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes is a T-cell mediated autoimmune disease characterized by pancreatic beta cells destruction. Angiotensin-converting enzyme 2 (ACE2), a component of renin-angiotensin system (RAS) has been identified in pancreas from type 2 diabetic mice and its overexpression prevents beta cell dysfunction. We studied the effect of ACE2 deletion on pancreatic and renal function in the nonobese diabetic mice, a model that mimics type 1 diabetes. ACE2-deficient NOD mice and the respective controls were generated. Pancreas function and immunohistochemistry studies were performed. Renal function and RAS gene expression were also analyzed. Renal proximal tubular cells were obtained from these animals to dissect the effect of ACE2 deficiency in these cells. In NOD mice, ACE2 deletion significantly worsened glucose homeostasis, decreased islet insulin content, increased beta cell oxidative stress, and RIPK1-positive islets as compared with control mice. Angiotensin-converting enzyme and angiotensin II type 1 receptor (AT1R) were also increased in ACE2-deficient mice. In kidneys of 30-day diabetic mice, ACE2 deletion decreased podocyte number within the glomeruli, and altered renal RAS gene expression in tubules. ACE2 deletion influenced the expression of fibrosis-related genes in isolated primary renal proximal tubular cells before diabetes onset in NOD mice. Our findings suggest that ACE2 deletion may have a deleterious impact on beta cell and renal function, by promoting oxidative stress and increasing necroptosis mediators. In addition, this effect is accompanied by RAS alterations in both pancreas and renal proximal tubular cells, indicating that ACE2 may exert a renopancreatic protective effect on type 1 diabetes, which is activated before diabetes starts.
Collapse
|
15
|
SARS-CoV-2 receptor networks in diabetic and COVID-19 associated kidney disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 32511461 PMCID: PMC7241118 DOI: 10.1101/2020.05.09.20096511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
COVID-19 morbidity and mortality is increased in patients with diabetes and kidney disease via unknown mechanisms. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) for entry into host cells. Since ACE2 is a susceptibility factor for infection, we investigated how diabetic kidney disease (DKD) and medications alter ACE2 receptor expression in kidneys. Single cell RNA profiling of healthy living donor (LD) and DKD kidney biopsies revealed ACE2 expression primarily in proximal tubular epithelial cells (PTEC). This cell specific localization was confirmed by in situ hybridization. ACE2 expression levels were unaltered by exposures to renin angiotensin aldosterone system inhibitors in DKD. Bayesian integrative analysis of a large compendium of public -omics datasets identified molecular network modules induced in ACE2-expressing PTEC in DKD (searchable at hb.flatironinstitute.org/covid-kidney) that were linked to viral entry, immune activation, endomembrane reorganization, and RNA processing. The DKD ACE2-positive PTEC module overlapped with expression patterns seen in SARS-CoV-2 infected cells. Similar cellular programs were seen in ACE2-positive PTEC obtained from urine samples of 13 COVID-19 patients who were hospitalized, suggesting a consistent ACE2-coregulated PTEC expression program that may interact with the SARS-CoV-2 infection processes. Thus SARS-CoV-2 receptor networks can seed further research into risk stratification and therapeutic strategies for COVID-19 related kidney damage.
Collapse
|
16
|
Akhtar S, Benter IF, Danjuma MI, Doi SAR, Hasan SS, Habib AM. Pharmacotherapy in COVID-19 patients: a review of ACE2-raising drugs and their clinical safety. J Drug Target 2020; 28:683-699. [PMID: 32700580 DOI: 10.1080/1061186x.2020.1797754] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic is caused by the severe acute-respiratory-syndrome-coronavirus-2 that uses ACE2 as its receptor. Drugs that raise serum/tissue ACE2 levels include ACE inhibitors (ACEIs) and angiotensin-II receptor blockers (ARBs) that are commonly used in patients with hypertension, cardiovascular disease and/or diabetes. These comorbidities have adverse outcomes in COVID-19 patients that might result from pharmacotherapy. Increasing ACE2 could potentially increase the risk of infection, severity or mortality in COVID-19 or it might be protective as it forms angiotensin-(1-7) which exhibits anti-inflammatory/anti-oxidative effects and prevents diabetes- and/or hypertension-induced end-organ damage. Thus, there existed clinical uncertainty. Here, we review studies implicating 15 classes of drugs in increasing ACE2 levels in vivo and the available literature on the clinical safety of these drugs in COVID-19 patients. Further, in a re-analysis of clinical data from a meta-analysis of 9 studies, we show that ACEIs/ARBs usage was not associated with an increased risk of all-cause mortality. Literature suggests that ACEIs/ARBs usage generally appears to be clinically safe though their use in severe COVID-19 patients might increase the risk of acute renal injury. For definitive clarity, further clinical and mechanistic studies are needed in assessing the safety of all classes of ACE2 raising medications.
Collapse
Affiliation(s)
- Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Mohammed I Danjuma
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Division of Internal Medicine, Hamad Medical Corporation Hospital, Doha, Qatar
| | - Suhail A R Doi
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Syed S Hasan
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
17
|
Kim NY, Ha E, Moon JS, Lee YH, Choi EY. Response: Acute Hyperglycemic Crises with Coronavirus Disease-19: Case Reports (Diabetes Metab J 2020;44:349-53). Diabetes Metab J 2020; 44:484-485. [PMID: 32613781 PMCID: PMC7332326 DOI: 10.4093/dmj.2020.0129] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Na Young Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Eunyeong Ha
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jun Sung Moon
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Yong Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea.
| | - Eun Young Choi
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea.
| |
Collapse
|
18
|
D’Marco L, Puchades MJ, Romero-Parra M, Gimenez-Civera E, Soler MJ, Ortiz A, Gorriz JL. Coronavirus disease 2019 in chronic kidney disease. Clin Kidney J 2020; 13:297-306. [PMID: 32699615 PMCID: PMC7367105 DOI: 10.1093/ckj/sfaa104] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Indexed: 01/04/2023] Open
Abstract
The clinical spectrum of coronavirus disease 2019 (COVID-19) infection ranges from asymptomatic infection to severe pneumonia with respiratory failure and even death. More severe cases with higher mortality have been reported in older patients and in those with chronic illness such as hypertension, diabetes or cardiovascular diseases. In this regard, patients with chronic kidney disease (CKD) have a higher rate of all-type infections and cardiovascular disease than the general population. A markedly altered immune system and immunosuppressed state may predispose CKD patients to infectious complications. Likewise, they have a state of chronic systemic inflammation that may increase their morbidity and mortality. In this review we discuss the chronic immunologic changes observed in CKD patients, the risk of COVID-19 infections and the clinical implications for and specific COVID-19 therapy in CKD patients. Indeed, the risk for severe COVID-19 is 3-fold higher in CKD than in non-CKD patients; CKD is 12-fold more frequent in intensive care unit than in non-hospitalized COVID-19 patients, and this ratio is higher than for diabetes or cardiovascular disease; and acute COVID-19 mortality is 15-25% for haemodialysis patients even when not developing pneumonia.
Collapse
Affiliation(s)
- Luis D’Marco
- Nephrology Department, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - María Jesús Puchades
- Nephrology Department, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - María Romero-Parra
- Nephrology Department, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Elena Gimenez-Civera
- Nephrology Department, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - María José Soler
- Nephrology Department, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Ortiz
- IIS-Fundación Jiménez Diaz UAM and School of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - José Luis Gorriz
- Nephrology Department, Hospital Clínico Universitario, INCLIVA, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
19
|
Giralt-López A, Molina-Van den Bosch M, Vergara A, García-Carro C, Seron D, Jacobs-Cachá C, Soler MJ. Revisiting Experimental Models of Diabetic Nephropathy. Int J Mol Sci 2020; 21:ijms21103587. [PMID: 32438732 PMCID: PMC7278948 DOI: 10.3390/ijms21103587] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetes prevalence is constantly increasing and, nowadays, it affects more than 350 million people worldwide. Therefore, the prevalence of diabetic nephropathy (DN) has also increased, becoming the main cause of end-stage renal disease (ESRD) in the developed world. DN is characterized by albuminuria, a decline in glomerular filtration rate (GFR), hypertension, mesangial matrix expansion, glomerular basement membrane thickening, and tubulointerstitial fibrosis. The therapeutic advances in the last years have been able to modify and delay the natural course of diabetic kidney disease (DKD). Nevertheless, there is still an urgent need to characterize the pathways that are involved in DN, identify risk biomarkers and prevent kidney failure in diabetic patients. Rodent models provide valuable information regarding how DN is set and its progression through time. Despite the utility of these models, kidney disease progression depends on the diabetes induction method and susceptibility to diabetes of each experimental strain. The classical DN murine models (Streptozotocin-induced, Akita, or obese type 2 models) do not develop all of the typical DN features. For this reason, many models have been crossed to a susceptible genetic background. Knockout and transgenic strains have also been created to generate more robust models. In this review, we will focus on the description of the new DN rodent models and, additionally, we will provide an overview of the available methods for renal phenotyping.
Collapse
Affiliation(s)
- Anna Giralt-López
- Nephrology Research Group, Vall d’Hebrón Institut de Recerca, 08035 Barcelona, Spain; (A.G.-L.); (M.M.-V.d.B.); (A.V.); (C.G.-C.); (D.S.)
| | - Mireia Molina-Van den Bosch
- Nephrology Research Group, Vall d’Hebrón Institut de Recerca, 08035 Barcelona, Spain; (A.G.-L.); (M.M.-V.d.B.); (A.V.); (C.G.-C.); (D.S.)
| | - Ander Vergara
- Nephrology Research Group, Vall d’Hebrón Institut de Recerca, 08035 Barcelona, Spain; (A.G.-L.); (M.M.-V.d.B.); (A.V.); (C.G.-C.); (D.S.)
- Nephrology Department, Vall d’Hebrón Hospital, 08035 Barcelona, Spain
| | - Clara García-Carro
- Nephrology Research Group, Vall d’Hebrón Institut de Recerca, 08035 Barcelona, Spain; (A.G.-L.); (M.M.-V.d.B.); (A.V.); (C.G.-C.); (D.S.)
- Nephrology Department, Vall d’Hebrón Hospital, 08035 Barcelona, Spain
| | - Daniel Seron
- Nephrology Research Group, Vall d’Hebrón Institut de Recerca, 08035 Barcelona, Spain; (A.G.-L.); (M.M.-V.d.B.); (A.V.); (C.G.-C.); (D.S.)
- Nephrology Department, Vall d’Hebrón Hospital, 08035 Barcelona, Spain
| | - Conxita Jacobs-Cachá
- Nephrology Research Group, Vall d’Hebrón Institut de Recerca, 08035 Barcelona, Spain; (A.G.-L.); (M.M.-V.d.B.); (A.V.); (C.G.-C.); (D.S.)
- Correspondence: (C.J.-C.); (M.J.S.)
| | - Maria José Soler
- Nephrology Research Group, Vall d’Hebrón Institut de Recerca, 08035 Barcelona, Spain; (A.G.-L.); (M.M.-V.d.B.); (A.V.); (C.G.-C.); (D.S.)
- Nephrology Department, Vall d’Hebrón Hospital, 08035 Barcelona, Spain
- Correspondence: (C.J.-C.); (M.J.S.)
| |
Collapse
|
20
|
Finucane FM, Davenport C. Coronavirus and Obesity: Could Insulin Resistance Mediate the Severity of Covid-19 Infection? Front Public Health 2020; 8:184. [PMID: 32574288 PMCID: PMC7247836 DOI: 10.3389/fpubh.2020.00184] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/24/2020] [Indexed: 01/08/2023] Open
Affiliation(s)
- Francis M Finucane
- HRB Clinical Research Facility, National University of Ireland Galway, Galway, Ireland.,Bariatric Medicine Service, Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Galway, Ireland
| | - Colin Davenport
- HRB Clinical Research Facility, National University of Ireland Galway, Galway, Ireland.,Bariatric Medicine Service, Centre for Diabetes, Endocrinology and Metabolism, Galway University Hospitals, Galway, Ireland
| |
Collapse
|
21
|
Fang Y, Gao F, Liu Z. Angiotensin-converting enzyme 2 attenuates inflammatory response and oxidative stress in hyperoxic lung injury by regulating NF-κB and Nrf2 pathways. QJM 2019; 112:914-924. [PMID: 31393582 DOI: 10.1093/qjmed/hcz206] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/25/2019] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To investigate the role of angiotensin-converting enzyme 2 (ACE2) in hyperoxic lung injury. METHODS Adult mice were exposed to 95% O2 for 72 h to induce hyperoxic lung injury, and simultaneously treated with ACE2 agonist diminazene aceturate (DIZE) or inhibitor MLN-4760. ACE2 expression/activity in lung tissue and angiotensin (Ang)-(1-7)/Ang II in bronchoalveolar lavage fluid (BALF), and the severity of hyperoxic lung injury were evaluated. The levels of inflammatory factors in BALF and lung tissue and the expression levels of phospho-p65, p65 and IkBα were measured. Oxidative parameter and antioxidant enzyme levels in lung tissue were measured to assess oxidative stress. Finally, the expression levels of nuclear factor-erythroid-2-related factor (Nrf2), NAD(P)H quinine oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1) were measured using Western blotting. RESULTS Hyperoxia treatment significantly decreased lung ACE2 expression/activity and increased the Ang II/Ang-(1-7) ratio, while co-treatment with hyperoxia and DIZE significantly increased lung ACE2 expression/activity and decreased the Ang II/Ang-(1-7) ratio. By contrast, co-treatment with hyperoxia and MLN-4760 significantly decreased lung ACE2 expression/activity and increased the Ang II/Ang-(1-7) ratio. Hyperoxia treatment induced significant lung injury, inflammatory response and oxidative stress, which were attenuated by DIZE but aggravated by MLN-4760. The NF-κB pathways were activated by hyperoxia and MLN-4760 but inhibited by DIZE. The Nrf2 pathway and its downstream proteins NQO1 and HO-1 were activated by DIZE but inhibited by MLN-4760. CONCLUSION Activation of ACE2 can reduce the severity of hyperoxic lung injury by inhibiting inflammatory response and oxidative stress. ACE2 can inhibit the NF-κB pathway and activate the Nrf2/HO-1/NQO1 pathway, which may be involved in the underlying mechanism.
Collapse
Affiliation(s)
- Y Fang
- Department of Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University, 85 WuJin Road, Shanghai, China
| | - F Gao
- Department of Respiratory Medicine, Shanghai Construction Group Hospital, No. 666, Zhongshan North 1st Road, Shanghai, China
| | - Z Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, 85 WuJing Road, Shanghai, China
| |
Collapse
|
22
|
Soler MJ, Batlle D. Single-cell RNA profiling of glomerular cells in diabetic kidney: a step forward for understanding diabetic nephropathy. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S340. [PMID: 32016058 PMCID: PMC6976463 DOI: 10.21037/atm.2019.09.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 09/18/2019] [Indexed: 12/25/2022]
Affiliation(s)
- María José Soler
- Nephrology Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Nephrology Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Red de Investigación Renal (REDINREN), Instituto Carlos IIIFEDER, Madrid, Spain
| | - Daniel Batlle
- Division of Nephrology & Hypertension, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Division of Nephrology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
23
|
Anguiano Gómez L, Lei Y, Kumar Devarapu S, Anders HJ. The diabetes pandemic suggests unmet needs for 'CKD with diabetes' in addition to 'diabetic nephropathy'-implications for pre-clinical research and drug testing. Nephrol Dial Transplant 2019; 33:1292-1304. [PMID: 28992221 DOI: 10.1093/ndt/gfx219] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/21/2017] [Indexed: 12/12/2022] Open
Abstract
Curing 'diabetic nephropathy' is considered an unmet medical need of high priority. We propose to question the concept of 'diabetic nephropathy' that implies diabetes as the predominant cause of kidney disease, which may not apply to the majority of type 2 diabetics approaching end-stage kidney disease. With the onset of diabetes, hyperglycaemia/sodium-glucose co-transporter-2-driven glomerular hyperfiltration promotes nephron hypertrophy, which, however, on its own, causes proteinuria not before a decade later, probably because podocyte hypertrophy can usually accommodate an increase in the filtration surface. In contrast, precedent chronic kidney disease (CKD), that is, few nephrons per body mass, e.g. due to poor nephron endowment from birth, obesity, pregnancy, or renal ageing or injury-related nephron loss, usually precedes the onset of type 2 diabetes. This applies in particular in older adults, and each on its own, but especially in combination, further aggravates single nephron hyperfiltration and glomerular hypertrophy. Whenever this additional hyperglycaemia-driven enlargement of the glomerular filtration surface exceeds the capacity of podocytes for hypertrophy, podocytes detachment leads to glomerulosclerosis and nephron loss, i.e. CKD progression. Animal models of 'diabetic nephropathy' based only on hyperglycaemia do not mimic this aspect and therefore poorly predict outcomes of clinical trials usually performed on elderly CKD patients with type 2 diabetes. Thus, we advocate the use of renal mass (nephron) ablation in type 2 diabetic animals to better mimic the pathophysiology of 'CKD with diabetes' in the target patient population and the use of the glomerular filtration rate as a primary endpoint to more reliably predict trial outcomes.
Collapse
Affiliation(s)
- Lidia Anguiano Gómez
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.,Department of Nephrology, Hospital del Mar-IMIM, Barcelona, Spain
| | - Yutian Lei
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Satish Kumar Devarapu
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
24
|
Guo H, Wang B, Li H, Ling L, Niu J, Gu Y. Glucagon-like peptide-1 analog prevents obesity-related glomerulopathy by inhibiting excessive autophagy in podocytes. Am J Physiol Renal Physiol 2017; 314:F181-F189. [PMID: 29070572 DOI: 10.1152/ajprenal.00302.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To investigate the role of glucagon-like peptide-1 analog (GLP-1) in high-fat diet-induced obesity-related glomerulopathy (ORG). Male C57BL/6 mice fed a high-fat diet for 12 wk were treated with GLP-1 (200 μg/kg) or 0.9% saline for 4 wk. Fasting blood glucose and insulin and the expression of podocin, nephrin, phosphoinositide 3-kinase (PI3K), glucose transporter type (Glut4), and microtubule-associated protein 1A/1B-light chain 3 (LC3) were assayed. Glomerular morphology and podocyte foot structure were evaluated by periodic acid-Schiff staining and electron microscopy. Podocytes were treated with 150 nM GLP-1 and incubated with 400 μM palmitic acid (PA) for 12 h. The effect on autophagy was assessed by podocyte-specific Glut4 siRNA. Insulin resistance and autophagy were assayed by immunofluorescence and Western blotting. The high-fat diet resulted in weight gain, ectopic glomerular lipid accumulation, increased insulin resistance, and fusion of podophyte foot processes. The decreased translocation of Glut4 to the plasma membrane and excess autophagy seen in mice fed a high-fat diet and in PA-treated cultured podocytes were attenuated by GLP-1. Podocyte-specific Glut4 siRNA promoted autophagy, and rapamycin-enhanced autophagy worsened the podocyte injury caused by PA. Excess autophagy in podocytes was induced by inhibition of Glut4 translocation to the plasma membrane and was involved in the pathology of ORG. GLP-1 restored insulin sensitivity and ameliorated renal injury by decreasing the level of autophagy.
Collapse
Affiliation(s)
- Honglei Guo
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China.,Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, Jiangsu Province, China
| | - Bin Wang
- Department of Nephrology, Zhongda Hospital Southeast University, Nanjing, Jiangsu Province, China
| | - Hongmei Li
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Lilu Ling
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Jianying Niu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China
| | - Yong Gu
- Division of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University , Shanghai , China.,Division of Nephrology, Huashan Hospital and Institute of Nephrology, Fudan University , Shanghai , China
| |
Collapse
|
25
|
Gonadectomy prevents the increase in blood pressure and glomerular injury in angiotensin-converting enzyme 2 knockout diabetic male mice. Effects on renin-angiotensin system. J Hypertens 2017; 34:1752-65. [PMID: 27379538 DOI: 10.1097/hjh.0000000000001015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Angiotensin-converting enzyme 2 (ACE2) deletion worsens kidney injury, and its amplification ameliorates diabetic nephropathy. Male sex increases the incidence, prevalence, and progression of chronic kidney disease in our environment. METHOD Here, we studied the effect of ACE2 deficiency and gonadectomy (GDX) on diabetic nephropathy and its relationship with fibrosis, protein kinase B (Akt) activation, and the expression of several components of the renin-angiotensin system (RAS).Mice were injected with streptozotocin to induce diabetes and followed for 19 weeks. Physiological and renal parameters were studied in wild-type and ACE2 knockout (ACE2KO) male mice with and without GDX. RESULTS Diabetic ACE2KO showed increased blood pressure (BP), glomerular injury, and renal fibrosis compared with diabetic wild-type. Gonadectomized diabetic ACE2KO presented a decrease in BP. In the absence of ACE2, GDX attenuated albuminuria and renal lesions, such as mesangial matrix expansion and podocyte loss. Both, α-smooth muscle actin accumulation and collagen deposition were significantly decreased in renal cortex of gonadectomized diabetic ACE2KO but not diabetic wild-type mice. GDX also reduced circulating ACE activity in ACE2KO mice. Loss of ACE2 modified the effect of GDX on cortical gene expression of RAS in diabetic mice. Akt phosphorylation in renal cortex was increased by diabetes and loss of ACE2 and decreased by GDX in control and diabetic ACE2KO but not in wild-type mice. CONCLUSIONS Our results suggest that GDX may exert a protective effect within the kidney under pathological conditions of diabetes and ACE2 deficiency. This renoprotection may be ascribed to different mechanisms such as decrease in BP, modulation of RAS, and downregulation of Akt-related pathways.
Collapse
|
26
|
Wysocki J, Goodling A, Burgaya M, Whitlock K, Ruzinski J, Batlle D, Afkarian M. Urine RAS components in mice and people with type 1 diabetes and chronic kidney disease. Am J Physiol Renal Physiol 2017; 313:F487-F494. [PMID: 28468961 DOI: 10.1152/ajprenal.00074.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/14/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
The pathways implicated in diabetic kidney disease (DKD) are largely derived from animal models. To examine if alterations in renin-angiotensin system (RAS) in humans are concordant with those in rodent models, we measured concentration of angiotensinogen (AOG), cathepsin D (CTSD), angiotensin-converting enzyme (ACE), and ACE2 and enzymatic activities of ACE, ACE2, and aminopeptidase-A in FVB mice 13-20 wk after treatment with streptozotocin (n = 9) or vehicle (n = 15) and people with long-standing type 1 diabetes, with (n = 37) or without (n = 81) DKD. In streptozotocin-treated mice, urine AOG and CTSD were 10.4- and 3.0-fold higher than in controls, respectively (P < 0.001). Enzymatic activities of ACE, ACE2, and APA were 6.2-, 3.2-, and 18.8-fold higher, respectively, in diabetic animals (P < 0.001). Angiotensin II was 2.4-fold higher in diabetic animals (P = 0.017). Compared with people without DKD, those with DKD had higher urine AOG (170 vs. 15 μg/g) and CTSD (147 vs. 31 μg/g). In people with DKD, urine ACE concentration was 1.8-fold higher (1.4 vs. 0.8 μg/g in those without DKD), while its enzymatic activity was 0.6-fold lower (1.0 vs. 1.6 × 109 RFU/g in those without DKD). Lower ACE activity, but not ACE protein concentration, was associated with ACE inhibitor (ACEI) treatment. After adjustment for clinical covariates, AOG, CTSD, ACE concentration, and ACE activity remained associated with DKD. In conclusion, in mice with streptozotocin-induced diabetes and in humans with DKD, urine concentrations and enzymatic activities of several RAS components are concordantly increased, consistent with enhanced RAS activity and greater angiotensin II formation. ACEI use was associated with a specific reduction in urine ACE activity, not ACE protein concentration, suggesting that it may be a marker of exposure to this widely-used therapy.
Collapse
Affiliation(s)
- Jan Wysocki
- Division of Nephrology and Hypertension, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Anne Goodling
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Mar Burgaya
- Division of Nephrology and Hypertension, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kathryn Whitlock
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, Seattle, Washington; and
| | - John Ruzinski
- Kidney Research Institute and Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Daniel Batlle
- Division of Nephrology and Hypertension, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois;
| | - Maryam Afkarian
- Division of Nephrology, Department of Medicine, University of California, Davis, California
| |
Collapse
|
27
|
Roca-Ho H, Riera M, Palau V, Pascual J, Soler MJ. Characterization of ACE and ACE2 Expression within Different Organs of the NOD Mouse. Int J Mol Sci 2017; 18:ijms18030563. [PMID: 28273875 PMCID: PMC5372579 DOI: 10.3390/ijms18030563] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Renin angiotensin system (RAS) is known to play a key role in several diseases such as diabetes, and renal and cardiovascular pathologies. Its blockade has been demonstrated to delay chronic kidney disease progression and cardiovascular damage in diabetic patients. In this sense, since local RAS has been described, the aim of this study is to characterize angiotensin converting enzyme (ACE) and ACE2 activities, as well as protein expression, in several tissues of the non-obese diabetic (NOD) mice model. After 21 or 40 days of diabetes onset, mouse serums and tissues were analyzed for ACE and ACE2 enzyme activities and protein expression. ACE and ACE2 enzyme activities were detected in different tissues. Their expressions vary depending on the studied tissue. Thus, whereas ACE activity was highly expressed in lungs, ACE2 activity was highly expressed in pancreas among the studied tissues. Interestingly, we also observed that diabetes up-regulates ACE mainly in serum, lung, heart, and liver, and ACE2 mainly in serum, liver, and pancreas. In conclusion, we found a marked serum and pulmonary alteration in ACE activity of diabetic mice, suggesting a common regulation. The increase of ACE2 activity within the circulation in diabetic mice may be ascribed to a compensatory mechanism of RAS.
Collapse
Affiliation(s)
- Heleia Roca-Ho
- Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain.
| | - Marta Riera
- Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain.
| | - Vanesa Palau
- Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain.
| | - Julio Pascual
- Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain.
- Nephrology Department-Hospital del Mar and Institut Hospital del Mar d'Investigacions Mèdiques-IMIM, 08003 Barcelona, Spain.
| | - Maria Jose Soler
- Institut Hospital del Mar d'Investigacions Mèdiques, 08003 Barcelona, Spain.
- Nephrology Department-Hospital del Mar and Institut Hospital del Mar d'Investigacions Mèdiques-IMIM, 08003 Barcelona, Spain.
| |
Collapse
|
28
|
Wolke C, Teumer A, Endlich K, Endlich N, Rettig R, Stracke S, Fiene B, Aymanns S, Felix SB, Hannemann A, Lendeckel U. Serum protease activity in chronic kidney disease patients: The GANI_MED renal cohort. Exp Biol Med (Maywood) 2016; 242:554-563. [PMID: 28038565 DOI: 10.1177/1535370216684040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Serum or plasma proteases have been associated with various diseases including cancer, inflammation, or reno-cardiovascular diseases. We aimed to investigate whether the enzymatic activities of serum proteases are associated with the estimated glomerular filtration rate (eGFR) in patients with different stages of chronic kidney disease (CKD). Our study population comprised 268 participants of the "Greifswald Approach to Individualized Medicine" (GANI_MED) cohort. Enzymatic activity of aminopeptidase A, aminopeptidase B, alanyl (membrane) aminopeptidase, insulin-regulated aminopeptidase, puromycin-sensitive aminopeptidase, leucine aminopeptidase 3, prolyl-endopeptidase (PEP), dipeptidyl peptidase 4 (DPP4), angiotensin I-converting enzyme, and angiotensin I-converting enzyme 2 (ACE2) proteases was measured in serum. Linear regression of the respective protease was performed on kidney function adjusted for age and sex. Kidney function was modeled either by the continuous Modification of Diet in Renal Disease (MDRD)-based eGFR or dichotomized by eGFR < 15 mL/min/1.73 m2 or <45 mL/min/1.73 m2, respectively. Results with a false discovery rate below 0.05 were deemed statistically significant. Among the 10 proteases investigated, only the activities of ACE2 and DPP4 were correlated with eGFR. Patients with lowest eGFR exhibited highest DPP4 and ACE2 activities. DPP4 and PEP were correlated with age, but all other serum protease activities showed no associations with age or sex. Our data indicate that ACE2 and DPP4 enzymatic activity are associated with the eGFR in patients with CKD. This finding distinguishes ACE2 and DPP4 from other serum peptidases analyzed and clearly indicates that further analyses are warranted to identify the precise role of these serum ectopeptidases in the pathogenesis of CKD and to fully elucidate underlying molecular mechanisms. Impact statement • Renal and cardiac diseases are very common and often occur concomitantly, resulting in increased morbidity and mortality. Understanding of molecular mechanisms linking both diseases is limited, available fragmentary data point to a role of the renin-angiotensin system (RAS) and, in particular, Ras-related peptidases. • Here, a comprehensive analysis of serum peptidase activities in patients with different stages of chronic kidney disease (CKD) is presented, with special emphasis given to RAS peptidases • The serum activities of the peptidases angiotensin I-converting enzyme 2 and dipeptidyl peptidase 4 were identified as closely associated with kidney function, specifically with the estimated glomerular filtration rate. The findings are discussed in the context of available data suggesting protective roles for both enzymes in reno-cardiac diseases. • The data add to our understanding of pathomechanisms underlying development and progression of CKD and indicate that both enzymes might represent potential pharmacological targets for the preservation of renal function.
Collapse
Affiliation(s)
- Carmen Wolke
- 1 Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Alexander Teumer
- 2 Dept. SHIP/KEF, Institute of Community Medicine, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Karlhans Endlich
- 3 Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Nicole Endlich
- 3 Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Rainer Rettig
- 4 Institute of Physiology, University Medicine Greifswald, Karlsburg D-17495, Germany
| | - Sylvia Stracke
- 5 Department of Internal Medicine A, Nephrology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Beate Fiene
- 5 Department of Internal Medicine A, Nephrology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Simone Aymanns
- 5 Department of Internal Medicine A, Nephrology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Stephan B Felix
- 6 Department of Internal Medicine B, Cardiology, Angiology, Pneumology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Anke Hannemann
- 7 Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Uwe Lendeckel
- 1 Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| |
Collapse
|
29
|
Gnudi L, Coward RJM, Long DA. Diabetic Nephropathy: Perspective on Novel Molecular Mechanisms. Trends Endocrinol Metab 2016; 27:820-830. [PMID: 27470431 DOI: 10.1016/j.tem.2016.07.002] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/04/2016] [Accepted: 07/07/2016] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus (DM) is the major cause of end-stage renal disease (ESRD) globally, and novel treatments are urgently needed. Current therapeutic approaches for diabetic nephropathy (DN) are focussing on blood pressure control with inhibitors of the renin-angiotensin-aldosterone system, on glycaemic and lipid control, and life-style changes. In this review, we highlight new molecular insights aiding our understanding of the initiation and progression of DN, including glomerular insulin resistance, dysregulation of cellular substrate utilisation, podocyte-endothelial communication, and inhibition of tubular sodium coupled glucose reabsorption. We believe that these mechanisms offer new therapeutic targets that can be exploited to develop important renoprotective treatments for DN over the next decade.
Collapse
Affiliation(s)
- Luigi Gnudi
- Cardiovascular Division, King's College London, London, SE1 9NH, UK.
| | - Richard J M Coward
- Academic Renal Unit, Dorothy Hodgkin Building, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - David A Long
- Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, WC1N 1EH, UK.
| |
Collapse
|
30
|
Anguiano L, Riera M, Pascual J, Valdivielso JM, Barrios C, Betriu A, Clotet S, Mojal S, Fernández E, Soler MJ. Circulating angiotensin converting enzyme 2 activity as a biomarker of silent atherosclerosis in patients with chronic kidney disease. Atherosclerosis 2016; 253:135-143. [PMID: 27615597 DOI: 10.1016/j.atherosclerosis.2016.08.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/21/2016] [Accepted: 08/23/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIMS Circulating Angiotensin Converting Enzyme 2 (ACE2) activity in chronic kidney disease (CKD) patients without previous history of cardiovascular disease (CVD) has been associated with classical risk factors (older age, diabetes and male gender). Furthermore, silent atherosclerosis has been described as a pathological link between CKD and CVD. We analyzed baseline ACE2 activity in non-dialysis CKD stages 3-5 (CKD3-5) patients as a biomarker of renal progression, silent atherosclerosis and CV events after 2 years of follow-up. METHODS Prospective study of 1458 CKD3-5 subjects without any previous CV event included in the Spanish multicenter NEFRONA study. Association between baseline circulating ACE2 activity and renal parameters, carotid/femoral echography, atheromatous disease, ankle-brachial index, intima-media thickness, need of renal replacement therapy, cardiovascular events and mortality at 24 months of follow-up were analyzed. RESULTS Patients with an increase in the number of territories with plaques at 24 months showed significantly higher levels of baseline ACE2 activity as compared to stable patients (29.6 (20.6-47.6)RFU/μL/h versus 35.7 (24.5-56), p < 0.001). Multivariate linear regression analysis showed that male gender, pathological ankle-brachial index and progressive silent atherosclerosis defined as an increased number of territories with plaques at 24 months were associated with increased baseline ACE2 activity. Male gender, older age, diabetes, smoking and increased baseline circulating ACE2 were independent predictors of atherosclerosis at 24 months of follow-up. CONCLUSIONS In CKD3-5 patients, higher circulating ACE2 activity at baseline is associated with higher risk for silent atherosclerosis, suggesting that ACE2 may serve as a biomarker to predict CV risk before CVD is established.
Collapse
Affiliation(s)
- Lidia Anguiano
- Department of Nephrology, Hospital del Mar-IMIM, Barcelona, Spain
| | - Marta Riera
- Department of Nephrology, Hospital del Mar-IMIM, Barcelona, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar-IMIM, Barcelona, Spain
| | | | - Clara Barrios
- Department of Nephrology, Hospital del Mar-IMIM, Barcelona, Spain
| | - Angels Betriu
- Nephrology Research Laboratory, Institute for Biomedical Research, IRB, Lleida, Spain; Department of Nephrology and UDETMA, University Hospital Arnau de Vilanova, Lleida, Spain
| | - Sergi Clotet
- Department of Nephrology, Hospital del Mar-IMIM, Barcelona, Spain
| | - Sergi Mojal
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Elvira Fernández
- Nephrology Research Laboratory, Institute for Biomedical Research, IRB, Lleida, Spain; Department of Nephrology and UDETMA, University Hospital Arnau de Vilanova, Lleida, Spain
| | - María José Soler
- Department of Nephrology, Hospital del Mar-IMIM, Barcelona, Spain.
| | | |
Collapse
|
31
|
Lin M, Gao P, Zhao T, He L, Li M, Li Y, Shui H, Wu X. Calcitriol regulates angiotensin-converting enzyme and angiotensin converting-enzyme 2 in diabetic kidney disease. Mol Biol Rep 2016; 43:397-406. [PMID: 26968558 DOI: 10.1007/s11033-016-3971-5] [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: 09/03/2015] [Accepted: 03/05/2016] [Indexed: 10/22/2022]
Abstract
To investigate the effects of calcitriol on angiotensin-converting enzyme (ACE) and ACE2 in diabetic nephropathy. Streptozotocin (STZ) induced diabetic rats were treated with calcitriol for 16 weeks. ACE/ACE2 and mitogen activated protein kinase (MAPK) enzymes were measured in the kidneys of diabetic rats and rat renal tubular epithelial cells exposed to high glucose. Calcitriol reduced proteinuria in diabetic rats without affecting calcium-phosphorus metabolism. ACE and ACE2 levels were significantly elevated in diabetic rats compared to those in control rats. The increase in ACE levels was greater than that of ACE2, leading to an elevated ACE/ACE2 ratio. Calcitriol reduced ACE levels and ACE/ACE2 ratio and increased ACE2 levels in diabetic rats. Similarly, high glucose up-regulated ACE expression in NRK-52E cells, which was blocked by the p38 MAPK inhibitor SB203580, but not the extracellular signal-regulated kinase (ERK) inhibitor FR180204 or the c-Jun N-terminal kinase (JNK) inhibitor SP600125. High glucose down-regulated ACE2 expression, which was blocked by FR180204, but not SB203580 or SP600125. Incubation of cells with calcitriol significantly inhibited p38 MAPK and ERK phosphorylation, but not JNK phosphorylation, and effectively attenuated ACE up-regulation and ACE2 down-regulation in high glucose conditions. The renoprotective effects of calcitriol in diabetic nephropathy were related to the regulation of tubular levels of ACE and ACE2, possibly by p38 MAPK or ERK, but not JNK pathways.
Collapse
Affiliation(s)
- Mei Lin
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Ping Gao
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| | - Tianya Zhao
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Lei He
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Mengshi Li
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yaoyao Li
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Hua Shui
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiaoyan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| |
Collapse
|
32
|
Riera M, Anguiano L, Clotet S, Roca-Ho H, Rebull M, Pascual J, Soler MJ. Paricalcitol modulates ACE2 shedding and renal ADAM17 in NOD mice beyond proteinuria. Am J Physiol Renal Physiol 2015; 310:F534-46. [PMID: 26697977 DOI: 10.1152/ajprenal.00082.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 12/17/2015] [Indexed: 02/07/2023] Open
Abstract
Circulating and renal activity of angiotensin-converting enzyme 2 (ACE2) is increased in non-obese diabetic (NOD) mice. Because paricalcitol has been reported to protect against diabetic nephropathy, we investigated the role of paricalcitol in modulating ACE2 in these mice. In addition, renal ADAM17, a metalloprotease implied in ACE2 shedding, was assessed. NOD female and non-diabetic control mice were studied for 21 days after diabetes onset and divided into various treatment groups. Diabetic animals received either vehicle; 0.4 or 0.8 μg/kg paricalcitol, aliskiren, or a combination of paricalcitol and aliskiren. We then studied the effect of paricalcitol on ACE2 expression in proximal tubular epithelial cells. Paricalcitol alone or in combination with aliskiren resulted in significantly reduced circulating ACE2 activity in NOD mice but there were no changes in urinary albumin excretion. Serum renin activity was significantly decreased in mice that received aliskiren but no effect was found when paricalcitol was used alone. Renal content of ADAM17 was significantly decreased in animals that received a high dose of paricalcitol. Renal and circulating oxidative stress (quantified by plasma H2O2 levels and immunolocalization of nitrotyrosine) were reduced in high-dose paricalcitol-treated mice compared with non-treated diabetic mice. In culture, paricalcitol incubation resulted in a significant increase in ACE2 expression compared with nontreated cells. In NOD mice with type 1 diabetes, paricalcitol modulates ACE2 activity, ADAM17, and oxidative stress renal content independently from the glycemic profile and urinary albumin excretion. In tubular cells, paricalcitol may modulate ACE2 by blocking its shedding. In the early stage of diabetic nephropathy, paricalcitol treatment counterbalances the effect of diabetes on circulating ACE2 activity. Our results suggest that additional use of paricalcitol may be beneficial in treating patients with diabetes under standard therapeutic strategies.
Collapse
Affiliation(s)
- Marta Riera
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III-FEDER, Madrid, Spain
| | - Lidia Anguiano
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; and
| | - Sergi Clotet
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; and
| | - Heleia Roca-Ho
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; and
| | - Marta Rebull
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; and
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III-FEDER, Madrid, Spain
| | - Maria Jose Soler
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III-FEDER, Madrid, Spain
| |
Collapse
|
33
|
Anguiano L, Riera M, Pascual J, Valdivielso JM, Barrios C, Betriu A, Mojal S, Fernández E, Soler MJ. Circulating angiotensin-converting enzyme 2 activity in patients with chronic kidney disease without previous history of cardiovascular disease. Nephrol Dial Transplant 2015; 30:1176-85. [PMID: 25813276 PMCID: PMC7107869 DOI: 10.1093/ndt/gfv025] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/10/2015] [Indexed: 01/01/2023] Open
Abstract
Background Patients with cardiovascular (CV) disease have an increased circulating angiotensin-converting enzyme 2 (ACE2) activity, but there is little information about changes in ACE2 in chronic kidney disease (CKD) patients without history of CV disease. We examined circulating ACE2 activity in CKD patients at stages 3–5 (CKD3-5) and in dialysis (CKD5D) without any history of CV disease. Methods Circulating ACE2 activity was measured in human ethylenediamine-tetraacetic acid (EDTA)-plasma samples from the NEFRONA study (n = 2572): control group (CONT) (n = 568), CKD3-5 (n = 1458) and CKD5D (n = 546). Different clinical and analytical variables such as gender; age; history of diabetes mellitus (DM), dyslipidemia and hypertension; glycaemic, renal, lipid and anaemia profiles; vitamin D analogues treatment and antihypertensive treatments (angiotensin-converting enzyme inhibitor and angiotensin receptor blockade) were analysed. Circulating ACE2 and ACE activities were measured using modified fluorimetric assay for EDTA-plasma samples, where zinc chloride was added to recover enzymatic activity. Results In CKD3-5 and CKD5D, significant decrease in circulating ACE2 activity was observed when compared with CONT, but no differences were found between CKD3-5 and CKD5 when performing paired case-control studies. By multivariate linear regression analysis, male gender and advanced age were identified as independent predictors of ACE2 activity in all groups. Diabetes was identified as independent predictor of ACE2 activity in CKD3-5. Significant increase in the activity of circulating ACE was found in CKD3-5 and CKD5D when compared with CONT and in CKD5D when compared with CKD3-5. By multiple regression analysis, female gender and younger age were identified as independent predictors of ACE activity in CONT and CKD3-5. Diabetes was also identified as an independent predictor of ACE activity in CKD3-5 patients. Conclusions Circulating ACE2 and ACE activities can be measured in human EDTA-plasma samples with zinc added to recover enzymatic activity. In a CKD population without previous history of CV disease, ACE2 activity from human EDTA-plasma samples directly correlated with the classical CV risk factors namely older age, diabetes and male gender. Our data suggest that circulating ACE2 is altered in CKD patients at risk for CV event.
Collapse
Affiliation(s)
- Lidia Anguiano
- Department of Nephrology, Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Marta Riera
- Department of Nephrology, Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | - Clara Barrios
- Department of Nephrology, Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Angels Betriu
- Nephrology Research Laboratory, Institute for Biomedical Research, IRB Lleida, Spain Department of Nephrology and UDETMA, University Hospital Arnau de Vilanova, Lleida, Spain
| | | | - Elvira Fernández
- Nephrology Research Laboratory, Institute for Biomedical Research, IRB Lleida, Spain Department of Nephrology and UDETMA, University Hospital Arnau de Vilanova, Lleida, Spain
| | - María José Soler
- Department of Nephrology, Hospital del Mar-IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | |
Collapse
|
34
|
Liang Y, Deng H, Bi S, Cui Z, A L, Zheng D, Wang Y. Urinary angiotensin converting enzyme 2 increases in patients with type 2 diabetic mellitus. Kidney Blood Press Res 2015; 40:101-10. [PMID: 25791940 DOI: 10.1159/000368486] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Angiotensin converting enzyme 2 (ACE2) is highly expressed in the kidney and recognized to be renoprotective by degrading Angiotensin II to Angiotensin (1-7) in diabetic nephropathy. However, little is known about the role of urinary ACE2 (UACE2) in diabetes. The present study was performed to evaluate UACE2 levels in type 2 diabetic patients with various degrees of albuminuria and its associations with metabolic parameters. The effect of RAS inhibitors on UACE2 excretion was also assessed. METHODS A total of 132 type 2 diabetic patients with different degrees of albuminuria and 34 healthy volunteers were studied. UACE2 levels and activity were measured. RESULTS Compared to healthy controls, UACE2 to creatinine (UACE2/Cr) levels were significantly increased in both albuminuric and non-albuminuric diabetic patients. UACE2/Cr levels were much higher in hypertensive diabetic patients compared with their normotensive counterparts and treatment with RAS inhibitors markedly attenuated the augmentation. Furthermore, UACE2/Cr was positively correlated with fasting blood glucose, hemoglobin A1C (HbA1C), triglyceride, and total cholesterol. In multiple regression analysis, UACE2/Cr was independently predicted by HbA1C and RAS inhibitors treatment. CONCLUSIONS UACE2 increased in type 2 diabetic patients with various degrees of albuminuria and RAS inhibitors suppresses UACE2 excretion. UACE2 might potentially function as a marker for monitoring the metabolic status and therapeutic response of RAS inhibitors in diabetes.
Collapse
Affiliation(s)
- Yaoxian Liang
- Department of Nephrology, Peking University Third Hospital, Beijing 100191, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Márquez E, Riera M, Pascual J, Soler MJ. Renin-angiotensin system within the diabetic podocyte. Am J Physiol Renal Physiol 2014; 308:F1-10. [PMID: 25339703 DOI: 10.1152/ajprenal.00531.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetic kidney disease is the leading cause of end-stage renal disease. Podocytes are differentiated cells necessary for the development and maintenance of the glomerular basement membrane and the capillary tufts, as well as the function of the glomerular filtration barrier. The epithelial glomerular cells express a local renin-angiotensin system (RAS) that varies in different pathological situations such as hyperglycemia or mechanical stress. RAS components have been shown to be altered in diabetic podocytopathy, and their modulation may modify diabetic nephropathy progression. Podocytes are a direct target for angiotensin II-mediated injury by altered expression and distribution of podocyte proteins. Furthermore, angiotensin II promotes podocyte injury indirectly by inducing cellular hypertrophy, increased apoptosis, and changes in the anionic charge of the glomerular basement membrane, among other effects. RAS blockade has been shown to decrease the level of proteinuria and delay the progression of chronic kidney disease. This review summarizes the local intraglomerular RAS and its imbalance in diabetic podocytopathy. A better understanding of the intrapodocyte RAS might provide a new approach for diabetic kidney disease treatment.
Collapse
Affiliation(s)
- Eva Márquez
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and
| | - Marta Riera
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| | - María José Soler
- Department of Nephrology, Hospital del Mar, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| |
Collapse
|
36
|
Santos SHS, Andrade JMO. Angiotensin 1-7: a peptide for preventing and treating metabolic syndrome. Peptides 2014; 59:34-41. [PMID: 25017239 DOI: 10.1016/j.peptides.2014.07.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/02/2014] [Accepted: 07/02/2014] [Indexed: 12/14/2022]
Abstract
Angiotensin-(1-7) is one of the most important active peptides of the renin-angiotensin system (RAS) with recognized cardiovascular relevance; however several studies have shown the potential therapeutic role of Ang-(1-7) on treating and preventing metabolic disorders as well. This peptide achieves a special importance considering that in the last few decades obesity and metabolic syndrome (MS) have become a growing worldwide health problem. Angiotensin (Ang) II is the most studied component of RAS and is increased during obesity, diabetes and dyslipidemia (MS); some experimental evidence has shown that Ang II modulates appetite and metabolism as well as mechanisms that induce adipose tissue growth and metabolism in peripheral organs. Recent articles demonstrated that Ang-(1-7)/Mas axis modulates lipid and glucose metabolism and counterregulates the effects of Ang II. Based on these data, angiotensin-converting enzyme 2 (ACE2)/Ang-(1-7)/Mas pathway activation have been advocated as a new tool for treating metabolic diseases. This review summarizes the new evidence from animal and human experiments indicating the use of Ang-(1-7) in prevention and treatment of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Sérgio Henrique Sousa Santos
- Pharmacology Department, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil; Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil.
| | - João Marcus Oliveira Andrade
- Laboratory of Health Science, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Minas Gerais, Brazil
| |
Collapse
|
37
|
Márquez E, Riera M, Pascual J, Soler MJ. Albumin inhibits the insulin-mediated ACE2 increase in cultured podocytes. Am J Physiol Renal Physiol 2014; 306:F1327-34. [DOI: 10.1152/ajprenal.00594.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Podocytes are key cells in the glomerular filtration barrier with a major role in the development of diabetic nephropathy. Podocytes are insulin-sensitive cells and have a functionally active local renin-angiotensin system. The presence and activity of angiotensin-converting enzyme 2 (ACE2), the main role of which is cleaving profibrotic and proinflammatory angiotensin-II into angiotensin-(1–7), have been demonstrated in podocytes. Conditionally immortalized mouse podocytes were cultured with insulin in the presence and absence of albumin. We found that insulin increases ACE2 gene and protein expression, by real-time PCR and Western blotting, respectively, and enzymatic activity within the podocyte and these increases were maintained over time. Furthermore, insulin favored an “anti-angiotensin II” regarding ACE/ACE2 gene expression balance and decreased fibronectin gene expression as a marker of fibrosis in the podocytes, all studied by real-time PCR. Similarly, insulin incubation seemed to protect podocytes from cell death, studied by a terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay. However, all these effects disappeared in the presence of albumin, which may mimic albuminuria, a main feature of DN pathophysiology. Our results suggest that modulation of renin-angiotensin system balance, fibrosis, and apoptosis by insulin in the podocyte may be an important factor in preventing the development and progression of diabetic kidney disease, but the presence of albuminuria seems to block these beneficial effects.
Collapse
Affiliation(s)
- Eva Márquez
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and
| | - Marta Riera
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and
- Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and
- Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| | - María José Soler
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; and
- Red de Investigación Renal (REDINREN), Instituto Carlos III, Madrid, Spain
| |
Collapse
|
38
|
Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol 2014; 11:413-26. [PMID: 24776703 PMCID: PMC7097196 DOI: 10.1038/nrcardio.2014.59] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) 2 and its product angiotensin 1–7 are thought to have effects that counteract the adverse actions of other, better-known renin–angiotensin system (RAS) components Numerous experimental studies have suggested that ACE2 and angiotensin 1–7 have notable protective effects in the heart and blood vessels ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the functional importance and signalling mechanisms of angiotensin-1–7-induced actions remain unclear New pharmacological interventions targeting ACE2 are expected to be useful in clinical treatment of cardiovascular disease, especially those associated with overactivation of the conventional RAS More studies, especially randomized controlled clinical trials, are needed to clearly delineate the benefits of therapies targeting angiotensin 1–7 actions
Angiotensin-converting enzyme 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of the better-known members of the renin–angiotensin system and might, therefore, be useful therapeutic targets in patients with cardiovascular disease. Professor Jiang and colleagues review the evidence for the potential roles of these proteins in various cardiovascular conditions, including hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes. The renin–angiotensin system (RAS) has pivotal roles in the regulation of normal physiology and the pathogenesis of cardiovascular disease. Angiotensin-converting enzyme (ACE) 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of other, better known and understood, members of the RAS. The physiological and pathological importance of ACE2 and angiotensin 1–7 in the cardiovascular system are not completely understood, but numerous experimental studies have indicated that these components have protective effects in the heart and blood vessels. Here, we provide an overview on the basic properties of ACE2 and angiotensin 1–7 and a summary of the evidence from experimental and clinical studies of various pathological conditions, such as hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes mellitus. ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the relevant functions and signalling mechanisms of actions induced by angiotensin 1–7 have not been conclusively determined. The ACE2–angiotensin 1–7 pathway, however, might provide a useful therapeutic target for the treatment of cardiovascular disease, especially in patients with overactive RAS.
Collapse
Affiliation(s)
- Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Jianmin Yang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Yongtao Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Mei Dong
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Fang Fang Liu
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Kai Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| |
Collapse
|