1
|
Salman O, Zhao L, Cohen JB, Dib MJ, Azzo JD, Gan S, Richards AM, Pourmussa B, Doughty R, Javaheri A, Mann DL, Rietzschel E, Zhao M, Wang Z, Ebert C, van Empel V, Kammerhoff K, Maranville J, Gogain J, Dennis J, Schafer PH, Seiffert D, Gordon DA, Ramirez-Valle F, Cappola TP, Chirinos JA. Proteomic Correlates and Prognostic Significance of Kidney Injury in Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2024; 13:e033660. [PMID: 39206761 DOI: 10.1161/jaha.123.033660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/15/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Kidney disease is common in heart failure with preserved ejection fraction (HFpEF). However, the biologic correlates and prognostic significance of kidney injury (KI), in HFpEF, beyond the estimated glomerular filtration rate (eGFR), are unclear. METHODS AND RESULTS Using baseline plasma samples from the TOPCAT (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist) trial, we measured the following KI biomarkers: cystatin-C, fatty acid-binding protein-3, Beta-2 microglobulin, neutrophil gelatinase-associated lipocalin, and kidney-injury molecule-1. Factor analysis was used to extract the common variability underlying these biomarkers. We assessed the relationship between the KI-factor score and the risk of death or HF-related hospital admission in models adjusted for the Meta-Analysis Global Group in Chronic Heart Failure risk score and eGFR. We also assessed the relationship between the KI factor score and ~5000 plasma proteins, followed by pathway analysis. We validated our findings among HFpEF participants in the Penn Heart Failure Study. KI was associated with the risk of death or HF-related hospital admission independent of the Meta-Analysis Global Group in Chronic Heart Failure risk score and eGFR. Both the risk score and eGFR were no longer associated with death or HF-related hospital admission after adjusting for the KI factor score. KI was predominantly associated with proteins and biologic pathways related to complement activation, inflammation, fibrosis, and cholesterol homeostasis. KI was associated with 140 proteins, which reproduced across cohorts. Findings regarding biologic associations and the prognostic significance of KI were also reproduced in the validation cohort. CONCLUSIONS KI is associated with adverse outcomes in HFpEF independent of baseline eGFR. Patients with HFpEF and KI exhibit a plasma proteomic signature indicative of complement activation, inflammation, fibrosis, and impaired cholesterol homeostasis.
Collapse
Affiliation(s)
- Oday Salman
- Hospital of the University of Pennsylvania Philadelphia PA USA
| | - Lei Zhao
- Bristol Myers Squibb Company Princeton NJ USA
| | - Jordana B Cohen
- Hospital of the University of Pennsylvania Philadelphia PA USA
- University of Pennsylvania Perelman School of Medicine Philadelphia PA USA
| | - Marie Joe Dib
- Hospital of the University of Pennsylvania Philadelphia PA USA
| | - Joe David Azzo
- Hospital of the University of Pennsylvania Philadelphia PA USA
| | - Sushrima Gan
- Hospital of the University of Pennsylvania Philadelphia PA USA
| | - A Mark Richards
- Cardiovascular Research Institute National University of Singapore Singapore
- Christchurch Heart Institute University of Otago New Zealand
| | - Bianca Pourmussa
- University of Pennsylvania Perelman School of Medicine Philadelphia PA USA
| | | | - Ali Javaheri
- Washington University School of Medicine St. Louis MO USA
| | - Douglas L Mann
- Washington University School of Medicine St. Louis MO USA
| | - Ernst Rietzschel
- Department of Cardiovascular Diseases Ghent University and Ghent University Hospital Ghent Belgium
| | - Manyun Zhao
- Hospital of the University of Pennsylvania Philadelphia PA USA
| | | | | | - Vanessa van Empel
- Department of Cardiology Maastricht University Medical Center Maastricht The Netherlands
| | | | | | | | | | | | | | | | | | - Thomas P Cappola
- Hospital of the University of Pennsylvania Philadelphia PA USA
- University of Pennsylvania Perelman School of Medicine Philadelphia PA USA
| | - Julio A Chirinos
- Hospital of the University of Pennsylvania Philadelphia PA USA
- University of Pennsylvania Perelman School of Medicine Philadelphia PA USA
| |
Collapse
|
2
|
Xu Y, Xu C, Huang J, Xu C, Xiong Y. Astragalus polysaccharide attenuates diabetic nephropathy by reducing apoptosis and enhancing autophagy through activation of Sirt1/FoxO1 pathway. Int Urol Nephrol 2024; 56:3067-3078. [PMID: 38653852 DOI: 10.1007/s11255-024-04038-0] [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: 06/25/2023] [Accepted: 03/16/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVE Diabetic nephropathy (DN) is the leading cause of end-stage renal disease in diabetic patients. Astragalus polysaccharide (APS) is a natural active ingredient in Astragalus membranaceus with anti-hypertensive and anti-oxidative properties. This study aimed to explore the protective roles of APS and its underlying mechanisms in DN. METHODS After the establishment of a rat model of DN by a high-fat diet and treatment with 30 mg/kg streptozotocin (STZ), the effects of 100 mg/kg APS on the levels of serum creatinine, blood urea nitrogen, blood glucose, and urinary albumin-to-creatinine ratio were measured. Histopathological alterations in renal tissues, renal cell apoptosis, renal inflammation, and oxidative stress were examined. The impacts of 0-200 μg/mL APS on the viability and apoptosis in high glucose (HG)-stimulated podocytes were measured by Cell Counting Kit-8 assays and flow cytometry, respectively. The expression of genes was tested by immunoblotting, quantitative real-time PCR, and immunofluorescence staining. RESULTS APS enhanced the expression of podocin and nephrin, increased viability, and reduced apoptosis in HG-induced podocytes. APS treatment abrogated high glucose-mediate suppression of autophagy in podocytes by activating the Sirt1/FoxO1 pathway. The Sirt1 inhibitor EX-527 eliminated the ameliorative effects of APS on renal dysfunction and renal tissue damage, as well as the inhibitory effects of APS on oxidative stress, inflammation, and apoptosis in DN rats. Moreover, EX-527 inhibited APS-induced autophagy activation in DN rats. CONCLUSION APS mitigated DN under hyperglycemic conditions by activating the Sirt1/FoxO1 autophagy pathway, suggesting that APS is a promising agent for DN treatment.
Collapse
Affiliation(s)
- Yanmei Xu
- Department of Nephrology, Wuhan Fourth Hospital, 473 Hanzheng Street, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Chen Xu
- Department of Nephrology, Wuhan Fourth Hospital, 473 Hanzheng Street, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Jie Huang
- Department of Nephrology, Wuhan Fourth Hospital, 473 Hanzheng Street, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Chuanwen Xu
- Department of Nephrology, Wuhan Fourth Hospital, 473 Hanzheng Street, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Yan Xiong
- Department of Nephrology, Wuhan Fourth Hospital, 473 Hanzheng Street, Qiaokou District, Wuhan, 430030, Hubei, China.
| |
Collapse
|
3
|
Yun D, Bae S, Gao Y, Lopez L, Han D, Nicora CD, Kim TY, Moon KC, Kim DK, Fillmore TL, Kim YS, Rosenberg AZ, Wang W, Sarder P, Qian WJ, Afkarian M, Han SS. Urine complement proteins are associated with kidney disease progression of type 2 diabetes in Korean and American cohorts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.15.24312080. [PMID: 39211860 PMCID: PMC11361241 DOI: 10.1101/2024.08.15.24312080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Mechanisms of progression of diabetic kidney disease (DKD) are not completely understood. This study uses untargeted and targeted mass spectrometry-based proteomics in two independent cohorts on two continents to decipher the mechanisms of DKD in patients with type 2 diabetes. Methods We conducted untargeted mass spectrometry on urine samples collected at the time of kidney biopsy from Korean patients with type 2 diabetes and biopsy-proven diabetic nephropathy at Seoul National University Hospital (SNUH-DN cohort; n = 64). These findings were validated using targeted mass spectrometry in urine samples from a Chronic Renal Insufficiency Cohort subgroup with type 2 diabetes and DKD (CRIC-T2D; n = 282). Urinary biomarkers/pathways associated with kidney disease progression (doubling of serum creatinine, ≥50% decrease in estimated glomerular filtration rates, or the development of end-stage kidney disease) were identified. Results SNUH-DN patients had an estimated glomerular filtration rate (eGFR) of 55 mL/min/1.73 m 2 (interquartile range [IQR], 44-75) and random urine protein-to-creatinine ratio of 3.1 g/g (IQR, 1.7-7.0). Urine proteins clustered into two groups, with cluster 2 having a 4.6-fold greater hazard (95% confidence interval [CI], 1.9-11.5) of disease progression than cluster 1 in multivariable-adjusted, time-to-event analyses. Proteins in cluster 2 mapped to 10 pathways, four of the top five of which were complement or complement-related. A high complement score, constructed from urine complement protein abundance, was strongly correlated to 4 of 5 histopathologic DN features and was associated with a 2.4-fold greater hazard (95% CI, 1.0-5.4) of disease progression than a low complement score. Targeted mass spectrometry of the CRIC-T2D participants, who had an eGFR of 42 mL/min/1.73 m 2 (IQR, 37-49) and 24-hr urine protein of 0.48 g (IQR, 0.10-1.87), showed that the complement score similarly segregated them into rapid and slow DKD progression groups. In both cohorts, the complement score had a linear association with disease progression. Conclusions Urinary proteomic profiling confirms the association between the complement pathway and rapid DKD progression in two independent cohorts. These results suggest a need to further investigate complement pathway inhibition as a novel treatment for DKD.
Collapse
|
4
|
Charlotte Brinck H, Lene H, Tom D, Wilhelm S, Troels Krarup H, Steffen T, Jakob Appel Ø. MASP-2 deficiency does not prevent the progression of diabetic kidney disease in a mouse model of type 1 diabetes. Scand J Immunol 2024; 99:e13348. [PMID: 39008346 DOI: 10.1111/sji.13348] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 07/16/2024]
Abstract
Mannan-binding lectin (MBL) initiates the lectin pathway of complement and has been linked to albuminuria and mortality in diabetes. We hypothesize that MBL-associated serine protease 2 (MASP-2) deficiency will protect against diabetes-induced kidney damage. Male C57BL/6J MASP-2 knockout (Masp2-/-) mice and wildtype (WT) mice were divided into a diabetic group and a non-diabetic group. Renal hypertrophy, albumin excretion, mesangial area and specific mRNA expressions in the renal cortex were measured after 8 and 12 weeks of diabetes. By two-way ANOVA it was tested if MASP-2 modulated the renal effects of diabetes, that is interaction. After 12 weeks of diabetes Masp2-/- diabetic mice had a smaller mesangium at 21.1% of the glomerular area (95% CI 19.7, 22.6) compared with WT diabetic mice, 25.2% (23.2, 27.2), p(interaction) = 0.001. After 8 weeks of diabetes, plasma cystatin C was 261.5 ng/mL (229.6, 297.8) in the WT diabetic group compared to 459.9 ng/mL (385.7, 548.3) in non-diabetic WT mice, p < 0.001. By contrast, no difference in plasma cystatin C levels was found between the Masp2-/- diabetic mice, 288.2 ng/mL (260.6, 318.6) and Masp2-/- non-diabetic mice, 293.5 ng/mL (221.0, 389.7), p = 0.86 and p(interaction) = 0.001. We demonstrated a protective effect of MASP-2 deficiency on mesangial hypertrophy after 12 weeks of diabetes and an effect on plasma cystatin C level. MASP-2 deficiency did, however, fail to protect against diabetic-induced alterations of kidney weight, albuminuria and renal mRNA expression of fibrotic- and oxidative stress markers.
Collapse
Affiliation(s)
- Holt Charlotte Brinck
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Halkjær Lene
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Dudler Tom
- Omeros Corporation, Seattle, Washington, USA
| | - Schwaeble Wilhelm
- Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | | | - Thiel Steffen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Østergaard Jakob Appel
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
5
|
Rajamanickam A, Dasan B, Munisankar S, Nott S, Menon PA, Ahamed Shaik F, Chinnaiyan P, Nutman TB, Babu S. Impact of Strongyloides stercoralis infection on complement activation in Type 2 diabetes mellitus: Insights from a clinical and anthelmintic intervention study. PLoS Negl Trop Dis 2024; 18:e0012048. [PMID: 38564496 PMCID: PMC10986927 DOI: 10.1371/journal.pntd.0012048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Numerous studies indicate a potential protective role of helminths in diabetes mellitus (DM) progression. The complement system, vital for host defense, plays a crucial role in tissue homeostasis and immune surveillance. Dysregulated complement activation is implicated in diabetic complications. We aimed to investigate the influence of the helminth, Strongyloides stercoralis (Ss) on complement activation in individuals with type 2 DM (T2D). METHODOLOGY We assessed circulating levels of complement proteins (C1q, C2, C3, C4, C4b, C5, C5a, and MBL (Lectin)) and their regulatory components (Factor B, Factor D, Factor H, and Factor I) in individuals with T2D with (n = 60) or without concomitant Ss infection (n = 58). Additionally, we evaluated the impact of anthelmintic therapy on these parameters after 6 months in Ss-infected individuals (n = 60). RESULTS Ss+DM+ individuals demonstrated reduced levels of complement proteins (C1q, C4b, MBL (Lectin), C3, C5a, and C3b/iC3b) and complement regulatory proteins (Factor B and Factor D) compared to Ss-DM+ individuals. Following anthelmintic therapy, there was a partial reversal of these levels in Ss+DM+ individuals. CONCLUSION Our findings indicate that Ss infection reduces complement activation, potentially mitigating inflammatory processes in individuals with T2D. The study underscores the complex interplay between helminth infections, complement regulation, and diabetes mellitus, offering insights into potential therapeutic avenues.
Collapse
Affiliation(s)
| | - Bindu Dasan
- NIH-NIAID-International Center for Excellence in Research, Chennai, India
| | | | - Sujatha Nott
- Infectious Diseases, Dignity Health, Chandler, Arizona, United States of America
| | | | - Fayaz Ahamed Shaik
- NIH-NIAID-International Center for Excellence in Research, Chennai, India
| | | | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Subash Babu
- NIH-NIAID-International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
6
|
Tserga A, Saulnier-Blache JS, Palamaris K, Pouloudi D, Gakiopoulou H, Zoidakis J, Schanstra JP, Vlahou A, Makridakis M. Complement Cascade Proteins Correlate with Fibrosis and Inflammation in Early-Stage Type 1 Diabetic Kidney Disease in the Ins2Akita Mouse Model. Int J Mol Sci 2024; 25:1387. [PMID: 38338666 PMCID: PMC10855735 DOI: 10.3390/ijms25031387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
Diabetic kidney disease (DKD) is characterized by histological changes including fibrosis and inflammation. Evidence supports that DKD is mediated by the innate immune system and more specifically by the complement system. Using Ins2Akita T1D diabetic mice, we studied the connection between the complement cascade, inflammation, and fibrosis in early DKD. Data were extracted from a previously published quantitative-mass-spectrometry-based proteomics analysis of kidney glomeruli of 2 (early DKD) and 4 months (moderately advanced DKD)-old Ins2Akita mice and their controls A Spearman rho correlation analysis of complement- versus inflammation- and fibrosis-related protein expression was performed. A cross-omics validation of the correlation analyses' results was performed using public-domain transcriptomics datasets (Nephroseq). Tissue sections from 43 patients with DKD were analyzed using immunofluorescence. Among the differentially expressed proteins, the complement cascade proteins C3, C4B, and IGHM were significantly increased in both early and later stages of DKD. Inflammation-related proteins were mainly upregulated in early DKD, and fibrotic proteins were induced in moderately advanced stages of DKD. The abundance of complement proteins with fibrosis- and inflammation-related proteins was mostly positively correlated in early stages of DKD. This was confirmed in seven additional human and mouse transcriptomics DKD datasets. Moreover, C3 and IGHM mRNA levels were found to be negatively correlated with the estimated glomerular filtration rate (range for C3 rs = -0.58 to -0.842 and range for IGHM rs = -0.6 to -0.74) in these datasets. Immunohistology of human kidney biopsies revealed that C3, C1q, and IGM proteins were induced in patients with DKD and were correlated with fibrosis and inflammation. Our study shows for the first time the potential activation of the complement cascade associated with inflammation-mediated kidney fibrosis in the Ins2Akita T1D mouse model. Our findings could provide new perspectives for the treatment of early DKD as well as support the use of Ins2Akita T1D in pre-clinical studies.
Collapse
Affiliation(s)
- Aggeliki Tserga
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
| | - Jean Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France; (J.S.S.-B.); (J.P.S.)
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Kostantinos Palamaris
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 34400 Athens, Greece; (K.P.); (D.P.); (H.G.)
| | - Despoina Pouloudi
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 34400 Athens, Greece; (K.P.); (D.P.); (H.G.)
| | - Harikleia Gakiopoulou
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 34400 Athens, Greece; (K.P.); (D.P.); (H.G.)
| | - Jerome Zoidakis
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
- Department of Biology, National and Kapodistrian University of Athens, 15701 Zografou, Greece
| | - Joost Peter Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France; (J.S.S.-B.); (J.P.S.)
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Antonia Vlahou
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
| | - Manousos Makridakis
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
| |
Collapse
|
7
|
Yang M, Zhang C. The role of innate immunity in diabetic nephropathy and their therapeutic consequences. J Pharm Anal 2024; 14:39-51. [PMID: 38352948 PMCID: PMC10859537 DOI: 10.1016/j.jpha.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/12/2023] [Accepted: 09/05/2023] [Indexed: 02/16/2024] Open
Abstract
Diabetic nephropathy (DN) is an enduring condition that leads to inflammation and affects a substantial number of individuals with diabetes worldwide. A gradual reduction in glomerular filtration and emergence of proteins in the urine are typical aspects of DN, ultimately resulting in renal failure. Mounting evidence suggests that immunological and inflammatory factors are crucial for the development of DN. Therefore, the activation of innate immunity by resident renal and immune cells is critical for initiating and perpetuating inflammation. Toll-like receptors (TLRs) are an important group of receptors that identify patterns and activate immune responses and inflammation. Meanwhile, inflammatory responses in the liver, pancreatic islets, and kidneys involve inflammasomes and chemokines that generate pro-inflammatory cytokines. Moreover, the activation of the complement cascade can be triggered by glycated proteins. This review highlights recent findings elucidating how the innate immune system contributes to tissue fibrosis and organ dysfunction, ultimately leading to renal failure. This review also discusses innovative approaches that can be utilized to modulate the innate immune responses in DN for therapeutic purposes.
Collapse
Affiliation(s)
- Min Yang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
8
|
Lin J, Weng M, Zheng J, Nie K, Rao S, Zhuo Y, Wan J. Identification and validation of voltage-dependent anion channel 1-related genes and immune cell infiltration in diabetic nephropathy. J Diabetes Investig 2024; 15:87-105. [PMID: 37737517 PMCID: PMC10759719 DOI: 10.1111/jdi.14087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/21/2023] [Accepted: 09/03/2023] [Indexed: 09/23/2023] Open
Abstract
AIMS/INTRODUCTION This study investigated the roles of voltage-dependent anion channel 1-related differentially expressed genes (VRDEGs) in diabetic nephropathy (DN). MATERIALS AND METHODS We downloaded two datasets from patients with DN, namely, GSE30122 and GSE30529, from the Gene Expression Omnibus database. VRDEGs associated with DN were obtained from the intersection of voltage-dependent anion channel 1-related genes from the GeneCards database, and differentially expressed genes were screened according to group (DN/healthy) in the two datasets. The enriched pathways of the VRDEGs were analyzed. Hub genes were selected using a protein-protein interaction network, and their predictive value was verified through receiver operating characteristic curve analysis. The CIBERSORTx software examined hub genes and immune cell infiltration associations. The protein expression of hub genes was verified through immunohistochemistry in 16-week-old db/db mice for experimentation as a model of type 2 DN. Finally, potential drugs targeting hub genes that inhibit DN development were identified. RESULTS A total of 57 VRDEGs were identified. The two datasets showed high expression of the PI3K, Notch, transforming growth factor-β, interleukin-10 and interleukin-17 pathways in DN. Five hub genes (ITGAM, B2M, LYZ, C3 and CASP1) associated with DN were identified and verified. Immunohistochemistry showed that the five hub genes were highly expressed in db/db mice, compared with db/m mice. The infiltration of immune cells was significantly correlated with the five hub genes. CONCLUSIONS Five hub genes were significantly correlated with immune cell infiltration and might be crucial to DN development. This study provides insight into the mechanisms involved in the pathogenesis of DN.
Collapse
Affiliation(s)
- Jiaqun Lin
- Department of Nephrology, Blood Purification Research Center, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Mengjie Weng
- Department of Nephrology, Blood Purification Research Center, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Jing Zheng
- Department of Nephrology, Blood Purification Research Center, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Kun Nie
- Department of Nephrology, Blood Purification Research Center, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Siyi Rao
- Department of Nephrology, Blood Purification Research Center, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yongjie Zhuo
- Department of Nephrology, Blood Purification Research Center, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Fujian Clinical Research Center for Metabolic Chronic Kidney Disease, The First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Nephrology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| |
Collapse
|
9
|
Liu MC, Li JL, Wang YF, Meng Y, Zheng GM, Cai Z, Shen C, Wang MD, Zhu XG, Chen YZ, Wang YL, Zhao WJ, Niu WQ, Wang YX. Association between serum complements and kidney function in patients with diabetic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1195966. [PMID: 38047115 PMCID: PMC10690951 DOI: 10.3389/fendo.2023.1195966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
Objective We aimed to explore the association between serum complements and kidney function of diabetic kidney disease (DKD) in Chinese patients. Methods This is a retrospective study involving 2,441 participants. DKD was diagnosed according to the Kidney Disease: Improving Global Outcomes (KDIGO) categories. Participants were classified as stages G1-G5 by KDIGO glomerular filtration rate (GFR) categories. Effect sizes are expressed as odds ratio (OR) with 95% confidence interval (CI). Results After balancing age, gender, systolic blood pressure (SBP), hemoglobin A1c (HbA1C), serum triglyceride (TG), and urinary albumin-to-creatinine ratio (UACR) between the G2-G5 and control groups, per 0.1 g/L increment in serum complement C3 was significantly associated with a 27.8% reduced risk of DKD at G5 stage (OR, 95% CI, P: 0.722, 0.616-0.847, <0.001) relative to the G1 stage. Conversely, per 0.1 g/L increment in serum complement C4 was associated with an 83.0-177.6% increased risk of G2-G5 stage (P<0.001). Serum complement C1q was not statistically significant compared to controls at all stages prior to or after propensity score matching. Conclusions Our results indicate that high concentrations of serum C4 were associated with the significantly elevated risk of kidney function deterioration across all stages, and reduced serum C3 levels with an increased risk of DKD stage G5.
Collapse
Affiliation(s)
- Meng-chao Liu
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jia-lin Li
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yue-fen Wang
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yuan Meng
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Gui-min Zheng
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zhen Cai
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Cun Shen
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Meng-di Wang
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiang-gang Zhu
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yang-zi Chen
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yu-lin Wang
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wen-jing Zhao
- Department of Nephropathy, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Wen-quan Niu
- Center for Evidence-Based Medicine, Capital Institute of Pediatrics, Beijing, China
| | - Yao-xian Wang
- The First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
- Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
10
|
Ghosh M, Rana S. The anaphylatoxin C5a: Structure, function, signaling, physiology, disease, and therapeutics. Int Immunopharmacol 2023; 118:110081. [PMID: 36989901 DOI: 10.1016/j.intimp.2023.110081] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
The complement system is one of the oldest known tightly regulated host defense systems evolved for efficiently functioning cell-based immune systems and antibodies. Essentially, the complement system acts as a pivot between the innate and adaptive arms of the immune system. The complement system collectively represents a cocktail of ∼50 cell-bound/soluble glycoproteins directly involved in controlling infection and inflammation. Activation of the complement cascade generates complement fragments like C3a, C4a, and C5a as anaphylatoxins. C5a is the most potent proinflammatory anaphylatoxin, which is involved in inflammatory signaling in a myriad of tissues. This review provides a comprehensive overview of human C5a in the context of its structure and signaling under several pathophysiological conditions, including the current and future therapeutic applications targeting C5a.
Collapse
Affiliation(s)
- Manaswini Ghosh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India.
| |
Collapse
|
11
|
Barreiro K, Lay AC, Leparc G, Tran VDT, Rosler M, Dayalan L, Burdet F, Ibberson M, Coward RJM, Huber TB, Krämer BK, Delic D, Holthofer H. An in vitro approach to understand contribution of kidney cells to human urinary extracellular vesicles. J Extracell Vesicles 2023; 12:e12304. [PMID: 36785873 PMCID: PMC9925963 DOI: 10.1002/jev2.12304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/26/2022] [Accepted: 01/05/2023] [Indexed: 02/15/2023] Open
Abstract
Extracellular vesicles (EV) are membranous particles secreted by all cells and found in body fluids. Established EV contents include a variety of RNA species, proteins, lipids and metabolites that are considered to reflect the physiological status of their parental cells. However, to date, little is known about cell-type enriched EV cargo in complex EV mixtures, especially in urine. To test whether EV secretion from distinct human kidney cells in culture differ and can recapitulate findings in normal urine, we comprehensively analysed EV components, (particularly miRNAs, long RNAs and protein) from conditionally immortalised human kidney cell lines (podocyte, glomerular endothelial, mesangial and proximal tubular cells) and compared to EV secreted in human urine. EV from cell culture media derived from immortalised kidney cells were isolated by hydrostatic filtration dialysis (HFD) and characterised by electron microscopy (EM), nanoparticle tracking analysis (NTA) and Western blotting (WB). RNA was isolated from EV and subjected to miRNA and RNA sequencing and proteins were profiled by tandem mass tag proteomics. Representative sets of EV miRNAs, RNAs and proteins were detected in each cell type and compared to human urinary EV isolates (uEV), EV cargo database, kidney biopsy bulk RNA sequencing and proteomics, and single-cell transcriptomics. This revealed that a high proportion of the in vitro EV signatures were also found in in vivo datasets. Thus, highlighting the robustness of our in vitro model and showing that this approach enables the dissection of cell type specific EV cargo in biofluids and the potential identification of cell-type specific EV biomarkers of kidney disease.
Collapse
Affiliation(s)
- Karina Barreiro
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Abigail C. Lay
- Bristol RenalBristol Medical SchoolFaculty of Health SciencesUniversity of BristolBristolUK
| | - German Leparc
- Boehringer Ingelheim Pharma GmbH & Co. KG BiberachBiberachGermany
| | - Van Du T. Tran
- Vital‐IT GroupSIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Marcel Rosler
- Boehringer Ingelheim Pharma GmbH & Co. KG BiberachBiberachGermany
| | - Lusyan Dayalan
- Bristol RenalBristol Medical SchoolFaculty of Health SciencesUniversity of BristolBristolUK
| | - Frederic Burdet
- Vital‐IT GroupSIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Mark Ibberson
- Vital‐IT GroupSIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Richard J. M. Coward
- Bristol RenalBristol Medical SchoolFaculty of Health SciencesUniversity of BristolBristolUK
| | - Tobias B. Huber
- III Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Bernhard K. Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology)University Medical Centre MannheimUniversity of HeidelbergMannheimGermany
| | - Denis Delic
- Boehringer Ingelheim Pharma GmbH & Co. KG BiberachBiberachGermany
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology/Pneumology)University Medical Centre MannheimUniversity of HeidelbergMannheimGermany
| | - Harry Holthofer
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
- III Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
12
|
Steffen BT, Tang W, Lutsey PL, Demmer RT, Selvin E, Matsushita K, Morrison AC, Guan W, Rooney MR, Norby FL, Pankratz N, Couper D, Pankow JS. Proteomic analysis of diabetes genetic risk scores identifies complement C2 and neuropilin-2 as predictors of type 2 diabetes: the Atherosclerosis Risk in Communities (ARIC) Study. Diabetologia 2023; 66:105-115. [PMID: 36194249 PMCID: PMC9742300 DOI: 10.1007/s00125-022-05801-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/15/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Genetic predisposition to type 2 diabetes is well-established, and genetic risk scores (GRS) have been developed that capture heritable liabilities for type 2 diabetes phenotypes. However, the proteins through which these genetic variants influence risk have not been thoroughly investigated. This study aimed to identify proteins and pathways through which type 2 diabetes risk variants may influence pathophysiology. METHODS Using a proteomics data-driven approach in a discovery sample of 7241 White participants in the Atherosclerosis Risk in Communities Study (ARIC) cohort and a replication sample of 1674 Black ARIC participants, we interrogated plasma levels of 4870 proteins and four GRS of specific type 2 diabetes phenotypes related to beta cell function, insulin resistance, lipodystrophy, BMI/blood lipid abnormalities and a composite score of all variants combined. RESULTS Twenty-two plasma proteins were identified in White participants after Bonferroni correction. Of the 22 protein-GRS associations that were statistically significant, 10 were replicated in Black participants and all but one were directionally consistent. In a secondary analysis, 18 of the 22 proteins were found to be associated with prevalent type 2 diabetes and ten proteins were associated with incident type 2 diabetes. Two-sample Mendelian randomisation indicated that complement C2 may be causally related to greater type 2 diabetes risk (inverse variance weighted estimate: OR 1.65 per SD; p=7.0 × 10-3), while neuropilin-2 was inversely associated (OR 0.44 per SD; p=8.0 × 10-3). CONCLUSIONS/INTERPRETATION Identified proteins may represent viable intervention or pharmacological targets to prevent, reverse or slow type 2 diabetes progression, and further research is needed to pursue these targets.
Collapse
Affiliation(s)
- Brian T Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Weihong Tang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Pamela L Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Weihua Guan
- Division of Biostatistics, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Mary R Rooney
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Faye L Norby
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Health System, Los Angeles, CA, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - David Couper
- University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
13
|
Tan SM, Snelson M, Østergaard JA, Coughlan MT. The Complement Pathway: New Insights into Immunometabolic Signaling in Diabetic Kidney Disease. Antioxid Redox Signal 2022; 37:781-801. [PMID: 34806406 PMCID: PMC9587781 DOI: 10.1089/ars.2021.0125] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: The metabolic disorder, diabetes mellitus, results in microvascular complications, including diabetic kidney disease (DKD), which is partly believe to involve disrupted energy generation in the kidney, leading to injury that is characterized by inflammation and fibrosis. An increasing body of evidence indicates that the innate immune complement system is involved in the pathogenesis of DKD; however, the precise mechanisms remain unclear. Recent Advances: Complement, traditionally thought of as the prime line of defense against microbial intrusion, has recently been recognized to regulate immunometabolism. Studies have shown that the complement activation products, Complement C5a and C3a, which are potent pro-inflammatory mediators, can mediate an array of metabolic responses in the kidney in the diabetic setting, including altered fuel utilization, disrupted mitochondrial respiratory function, and reactive oxygen species generation. In diabetes, the lectin pathway is activated via autoreactivity toward altered self-surfaces known as danger-associated molecular patterns, or via sensing altered carbohydrate and acetylation signatures. In addition, endogenous complement inhibitors can be glycated, whereas diet-derived glycated proteins can themselves promote complement activation, worsening DKD, and lending support for environmental influences as an additional avenue for propagating complement-induced inflammation and kidney injury. Critical Issues: Recent evidence indicates that conventional renoprotective agents used in DKD do not target the complement, leaving this web of inflammatory stimuli intact. Future Directions: Future studies should focus on the development of novel pharmacological agents that target the complement pathway to alleviate inflammation, oxidative stress, and kidney fibrosis, thereby reducing the burden of microvascular diseases in diabetes. Antioxid. Redox Signal. 37, 781-801.
Collapse
Affiliation(s)
- Sih Min Tan
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Australia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Australia
| | - Jakob A Østergaard
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Australia.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Australia.,Baker Heart & Diabetes Institute, Melbourne, Australia
| |
Collapse
|
14
|
Mannan-binding lectin serine protease-2 (MASP-2) in human kidney and its relevance for proteolytic activation of the epithelial sodium channel. Sci Rep 2022; 12:15955. [PMID: 36153401 PMCID: PMC9509361 DOI: 10.1038/s41598-022-20213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 09/09/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractProteolytic activation of the renal epithelial sodium channel (ENaC) is increased by aldosterone. The aldosterone-sensitive protease remains unidentified. In humans, elevated circulating aldosterone is associated with increased urinary extracellular vesicle (uEVs) excretion of mannan-binding lectin associated serine protease-2 (MASP-2). We hypothesized that MASP-2 is a physiologically relevant ENaC-activating protease. It was confirmed that MASP2 mRNA is abundantly present in liver but not in human and mouse kidneys. Aldosterone-stimulation of murine cortical colleting duct (mCCD) cells did not induce MASP-2 mRNA. In human kidney collecting duct, MASP-2 protein was detected in AQP2-negative/ATP6VB1-positive intercalated cells suggestive of MASP2 protein uptake. Plasma concentration of full-length MASP-2 and the short splice variant MAp19 were not changed in a cross-over intervention study in healthy humans with low (70 mmol/day) versus high (250 mmol/day) Na+ intake despite changes in aldosterone. The ratio of MAp19/MASP-2 in plasma was significantly increased with a high Na+ diet and the ratio correlated with changes in aldosterone and fractional Na+ excretion. MASP-2 was not detected in crude urine or in uEVs. MASP2 activated an amiloride-sensitive current when co-expressed with ENaC in Xenopus oocytes, but not when added to the bath solution. In monolayers of collecting duct M1 cells, MASP2 expression did not increase amiloride-sensitive current and in HEK293 cells, MASP-2 did not affect γENaC cleavage. MASP-2 is neither expressed nor co-localized and co-regulated with ENaC in the human kidney or in urine after low Na+ intake. MASP-2 does not mediate physiological ENaC cleavage in low salt/high aldosterone settings.
Collapse
|
15
|
Xu Z, Tao L, Su H. The Complement System in Metabolic-Associated Kidney Diseases. Front Immunol 2022; 13:902063. [PMID: 35924242 PMCID: PMC9339597 DOI: 10.3389/fimmu.2022.902063] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolic syndrome (MS) is a group of clinical abnormalities characterized by central or abdominal obesity, hypertension, hyperuricemia, and metabolic disorders of glucose or lipid. Currently, the prevalence of MS is estimated about 25% in general population and is progressively increasing, which has become a challenging public health burden. Long-term metabolic disorders can activate the immune system and trigger a low-grade chronic inflammation named “metaflammation.” As an important organ involved in metabolism, the kidney is inevitably attacked by immunity disequilibrium and “metaflammation.” Recently, accumulating studies have suggested that the complement system, the most important and fundamental component of innate immune responses, is actively involved in the development of metabolic kidney diseases. In this review, we updated and summarized the different pathways through which the complement system is activated in a series of metabolic disturbances and the mechanisms on how complement mediate immune cell activation and infiltration, renal parenchymal cell damage, and the deterioration of renal function provide potential new biomarkers and therapeutic options for metabolic kidney diseases.
Collapse
|
16
|
Jiang S, Di D, Jiao Y, Zou G, Gao H, Li W. Complement Deposition Predicts Worsening Kidney Function and Underlines the Clinical Significance of the 2010 Renal Pathology Society Classification of Diabetic Nephropathy. Front Immunol 2022; 13:868127. [PMID: 35711407 PMCID: PMC9196586 DOI: 10.3389/fimmu.2022.868127] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Converging evidence points towards a role of the complement system in the pathogenesis of diabetic nephropathy (DN). The classification system of diabetic kidney lesions devised by the Renal Pathology Society (RPS) in 2010 are based on the pathogenic process of DN. Therefore, we investigated the correlation between glomerular C3 deposits and RPS DN classification and the combined deleterious effects thereof on kidney function. Methods The study analyzed data from 217 diabetic patients who underwent renal biopsy between 2010 and 2021 and were found to have DN as the only glomerular disease. C3 deposition was considered positive if the glomerular C3 immunofluorescence intensity was at the trace or ≥1+ level. We divided DN into five glomerular lesion classes and separately evaluated the degree of interstitial and vascular involvement. The primary outcome was the composite of a ≥50% decline from the initial estimated glomerular filtration rate, end-stage renal disease, and death. Results None of the patients were classified into class I, and few were classified into classes IIa (7.8%) and IV (9.2%). Most patients were classified as IIb (30.9%) and III (52.1%). C3 deposition was detected in 53.9% of patients. Multivariate logistic regression analysis showed that DN class was significantly correlated with C3 deposits [odds ratio, 1.59; 95% confidence interval (CI), 1.08-2.36; p = 0.02). During a median follow-up of 22 months, 123 (56.7%) patients reached the composite outcome. The endpoints occurred more frequently in patients with C3 deposition (69.2 vs. 42%) compared with those without C3 deposition. Patients with C3 deposition in either class IIb [hazards ratio (HR), 3.9 (95% CI, 1.14-13.17) vs. 2.46 (95% CI, 0.68-8.89)] or III [HR, 4.98 (95% CI, 1.53-16.23) vs. 2.63 (95% CI, 0.77-9.0)] had a higher risk of adverse kidney outcomes than those without C3 deposition. The prognostic accuracy of the combination of DN class and C3 deposits at 1 and 3 years was higher than that for DN class only. Conclusions Complement deposition together with DN class predicts more rapid deterioration of kidney function in DN, which underlines the clinical significance of the DN phenotype according to the RPS classification.
Collapse
Affiliation(s)
- Shimin Jiang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Dingxin Di
- Graduate School of Peking Union Medical College, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanyuan Jiao
- Graduate School of Peking Union Medical College, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guming Zou
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Hongmei Gao
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
17
|
Ebefors K, Bergwall L, Nyström J. The Glomerulus According to the Mesangium. Front Med (Lausanne) 2022; 8:740527. [PMID: 35155460 PMCID: PMC8825785 DOI: 10.3389/fmed.2021.740527] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
The glomerulus is the functional unit for filtration of blood and formation of primary urine. This intricate structure is composed of the endothelium with its glycocalyx facing the blood, the glomerular basement membrane and the podocytes facing the urinary space of Bowman's capsule. The mesangial cells are the central hub connecting and supporting all these structures. The components as a unit ensure a high permselectivity hindering large plasma proteins from passing into the urine while readily filtering water and small solutes. There has been a long-standing interest and discussion regarding the functional contribution of the different cellular components but the mesangial cells have been somewhat overlooked in this context. The mesangium is situated in close proximity to all other cellular components of the glomerulus and should be considered important in pathophysiological events leading to glomerular disease. This review will highlight the role of the mesangium in both glomerular function and intra-glomerular crosstalk. It also aims to explain the role of the mesangium as a central component involved in disease onset and progression as well as signaling to maintain the functions of other glomerular cells to uphold permselectivity and glomerular health.
Collapse
Affiliation(s)
- Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lovisa Bergwall
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Nyström
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
18
|
Zhang Y, Liao H, Shen D, Zhang X, Wang J, Zhang X, Wang X, Li R. Renal Protective Effects of Inonotus obliquus on High-Fat Diet/Streptozotocin-Induced Diabetic Kidney Disease Rats: Biochemical, Color Doppler Ultrasound and Histopathological Evidence. Front Pharmacol 2022; 12:743931. [PMID: 35111043 PMCID: PMC8801815 DOI: 10.3389/fphar.2021.743931] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/03/2021] [Indexed: 11/30/2022] Open
Abstract
Diabetic kidney disease (DKD) is the current leading cause of end-stage renal disease. Inonotus obliquus (chaga), a medicinal fungus, has been used in treatment of diabetes. Here, we aim to identify the renal protective effects of chaga extracts on a DKD rat model which was induced by a high-fat diet and streptozotocin injection. During the total 17-weeks experiment, the biological parameters of serum and urine were examined, and the color Doppler ultrasound of renal artery, the periodic acid-Schiff staining, and electron microscopy of kidney tissue were performed. The compositions of chaga extracts were analyzed and the intervention effects of the extracts were also observed. Compared with the normal control group, the biochemical research showed that insulin resistance was developed, blood glucose and total cholesterol were elevated, urinary protein excretion and serum creatinine levels were significantly increased in the DKD model. The ultrasound examinations confirmed the deteriorated blood flow parameters of the left renal interlobar artery in the rat models. Finally, histopathological data supported renal injury on the thickened glomerular basement membrane and fusion of the foot processes. 8 weeks intervention of chaga improved the above changes significantly, and the 100 mg/kg/d chaga group experienced significant effects compared with the 50 mg/kg/d in some parameters. Our findings suggested that Doppler ultrasound examinations guided with biochemical indicators played important roles in evaluating the renal injury as an effective, noninvasive, and repeatable method in rats. Based on biochemical, ultrasound, and histopathological evidence, we confirmed that chaga had pharmacodynamic effects on diabetes-induced kidney injury and the aforementioned effects may be related to delaying the progression of DKD.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Hui Liao
- Department of Pharmacy, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Dayue Shen
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Xilan Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Jufang Wang
- Department of Ultrasonic Diagnosis, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Xiaohong Zhang
- Department of Ultrasonic Diagnosis, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Xiaocheng Wang
- Department of Statistic and Medical Record, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| | - Rongshan Li
- Department of Nephrology, Fifth Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan, China
| |
Collapse
|
19
|
Babu SN, Govindarajan S, Noor A. Aloe vera and its two bioactive constituents in alleviation of diabetes -proteomic & mechanistic insights. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114445. [PMID: 34303804 DOI: 10.1016/j.jep.2021.114445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aloe barbadensis Miller, commonly known as Aloe vera has been used since time immemorial for treatment of various diseases such as cancer, inflammatory disorders, diabetes, wound healing etc. AIM: Diabetes mellitus is a complex disorder and understanding the molecular mechanisms involved is a key to identify different markers for early diagnosis of the disease. The proteomic approach offers a plethora of opportunities to identify markers and targets involved in pathogenesis of diabetes. The present study was undertaken to understand the mechanism of action of Aloe vera and its two constituents (Carbohydrates and Polypeptides) in the alleviation of diabetes in streptozotocin-induced diabetic rats through a proteomics approach. METHODS Different groups of rats were fed with Aloe vera extract, carbohydrate fraction and peptide/polypeptide fraction for three weeks. The diabetic rats fed with Aloe vera and its two fractions restored the glucose and insulin levels to normal. The plasma of the rats was depleted with IgG and albumin and proteomic analysis was carried out. Apolipoproteins (dyslipidemia), complement factors (inflammatory pathways), zonulin (intestinal permeability), anti-oxidant related proteins were selected in this study as these are involved in the progression of diabetes. RESULTS It was observed that Aloe vera extract is involved in the alleviation of diabetes through these pathways while the carbohydrate fraction alleviates diabetes through an anti-oxidant mechanism and glucose uptake while the polypeptide fraction alleviates diabetes through the restoration of intestinal permeability by reduced zonulin levels. CONCLUSION The constituents of Aloe vera works different pathways involved in diabetes and the synergistic effect of these constituents make Aloe vera extract a prospective candidate, which can alleviate diabetes through regulation of the pathways involved in the progression of diabetes.
Collapse
Affiliation(s)
- Spoorthy N Babu
- Centre for Bio Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore-632 014, Tamil Nadu, India
| | - S Govindarajan
- Centre for Bio Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore-632 014, Tamil Nadu, India
| | - Ayesha Noor
- Centre for Bio Separation Technology (CBST), Vellore Institute of Technology (VIT), Vellore-632 014, Tamil Nadu, India.
| |
Collapse
|
20
|
Chang DY, Li XQ, Chen M, Zhao MH. Dapagliflozin Ameliorates Diabetic Kidney Disease via Upregulating Crry and Alleviating Complement Over-activation in db/db Mice. Front Pharmacol 2021; 12:729334. [PMID: 34712135 PMCID: PMC8546210 DOI: 10.3389/fphar.2021.729334] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/29/2021] [Indexed: 12/30/2022] Open
Abstract
Sodium-glucose cotransporter 2(SGLT2) inhibitors show prominent renal protective effect in diabetic kidney disease (DKD), anti-inflammatory effect being one of its key mechanisms. Over-activation of the complement system, a crucial part of innate immunity, plays an important role in DKD. We aimed to investigate the effect of SGLT2 inhibitors on alleviating complement over-activation in DKD. Db/db mice were randomly divided into two groups, with 7 mice in each group treated with dapagliflozin and vehicle respectively, and 7 mice in m/m mice group. Laboratory and renal pathological parameters were evaluated. Mouse proximal tubular epithelial cells (MPTECs) were cultured and treated with high glucose. Dapagliflozin and dimethyloxallyl glycine (DMOG) were added as conditional treatment. Dapagliflozin-treated db/db mice showed significantly lower urinary albumin than vehicle-treated ones. Besides typical glomerular and tubulointerstitial injury, both C3b and membrane attack complex (MAC) depositions were significantly attenuated in dapagliflozin-treated db/db mice. The expression of complement receptor type 1-related protein y (Crry), a key complement regulator which inhibits complement over-activation, was significantly upregulated by dapagliflozin. Dapagliflozin-mediated Crry upregulation was associated with inhibition of HIF-1α accumulation under high glucose. When HIF-1α expression was stabilized by DMOG, the protective effect of dapagliflozin via upregulating Crry was blocked. In conclusion, dapagliflozin could attenuate complement over-activation in diabetic mice via upregulating Crry, which is associated with the suppression of HIF-1α accumulation in MPTECs.
Collapse
Affiliation(s)
- Dong-Yuan Chang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Qian Li
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China.,Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Zhang Y. MiR-92d-3p suppresses the progression of diabetic nephropathy renal fibrosis by inhibiting the C3/HMGB1/TGF-β1 pathway. Biosci Rep 2021; 41:BSR20203131. [PMID: 33729484 PMCID: PMC8485393 DOI: 10.1042/bsr20203131] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
The pathogenesis of diabetic nephropathy (DN) has not been fully elucidated. MicroRNAs (miRNAs) play an important role in the onset and development of DN renal fibrosis. Thus, the present study aimed to investigate the effect of miR-92d-3p on the progression of DN renal fibrosis. We used qRT-PCR to detect the expression levels of miR-92d-3p in the kidneys of patients with DN. Then, after transfecting lentiviruses containing miR-92d-3p into the kidneys of a DN mouse model and HK-2 cell line, we used qRT-PCR to detect the expression levels of miR-92d-3p, C3, HMGB1, TGF-β1, α-SMA, E-cadherin, and Col I. The expression levels of interleukin (IL) 1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α) in the HK-2 cells were detected through enzyme-linked immunosorbent assay (ELISA), and Western blotting and immunofluorescence were used in detecting the expression levels of fibronectin, α-SMA, E-cadherin, and vimentin. Results showed that the expression levels of miR-92d-3p in the kidney tissues of patients with DN and DN animal model mice decreased, and C3 stimulated HK-2 cells to produce inflammatory cytokines. The C3/HMGB1/TGF-β1 pathway was activated, and epithelial-to-interstitial transition (EMT) was induced in the HK-2 cells after human recombinant C3 and TGF-β1 protein were added. miR-92d-3p inhibited inflammatory factor production by C3 in the HK-2 cells and the activation of the C3/HMGB1/TGF-β1 pathway and EMT by C3 and TGF-β1. miR-92d-3p suppressed the progression of DN renal fibrosis by inhibiting the activation of the C3/HMGB1/TGF-β1 pathway and EMT.
Collapse
Affiliation(s)
- Yuhua Zhang
- College of Medicine, Jiangxi University of Technology, Nanchang 330098, Jiangxi, China
| |
Collapse
|
22
|
Wu J, Lai G, Chen F, Zhang B, Zhao Y. Renal NKCC2 Is Dual Regulated by the Synergy of 20-HETE and High-Fat Diet in CYP4F2 Transgenic Mice. Kidney Blood Press Res 2021; 46:601-612. [PMID: 34320496 DOI: 10.1159/000517382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/22/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION 20-Hydroxyeicosatetraenoic acid (20-HETE) is the metabolite of cytochrome P450, which modulates blood pressure by inhibiting renal sodium transport. However, the molecular mechanisms underlying the role of 20-HETE in the development of obesity-related hypertension remain unclear, necessitating this study. METHODS Cytochrome P450 4F2 (CYP4F2) transgenic mice fed high-fat diet (HFD) were used as research animal models. The expression of renal ion transport molecules targeted by 20-HETE was evaluated by real-time PCR and Western blot (WB). The regulatory effect of 20-HETE and HFD on renal Na+-K+-2Cl- cotransporter, isoform 2 (NKCC2) was explored by immunoprecipitation, WB, and luciferase assay. RESULTS A 2-week HFD feeding dramatically decreased protein abundance but increased renal NKCC2 mRNA expression in CYP4F2 transgenic mice. The decrease in NKCC2 protein was demonstrated to be due to ubiquitination induced by the synergy between 20-HETE and HFD. The increased PPAR-γ protein in CYP4F2 transgenic mice fed HFD and the activation of rosiglitazone on the luciferase reporter construct of the NKCC2 promoter demonstrated that the increase in NKCC2 mRNA in CYP4F2 transgenic mice fed HFD was a consequence of elevated PPAR-γ protein induced by the synergy between 20-HETE and HFD. CONCLUSIONS Our data demonstrated that the synergy between 20-HETE and HFD could decrease NKCC2 protein via posttranslational ubiquitination, which was thought to be the main mechanism underlying the short-term effect in response to HFD and might be responsible for the adaptive modulation of renal NKCC2 to resist sodium retention. Moreover, the increased NKCC2 mRNA expression via PPAR-γ-induced transcriptional regulation was thought to be the main mechanism underlying the long-term effect in response to HFD and plays a pivotal role in the development of obesity-related hypertension.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Medical Genetics, School of Life Sciences, China Medical University, Shenyang, China,
| | - Guangrui Lai
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fangjie Chen
- Department of Medical Genetics, School of Life Sciences, China Medical University, Shenyang, China
| | - Bijun Zhang
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanyan Zhao
- Department of Medical Genetics, School of Life Sciences, China Medical University, Shenyang, China
| |
Collapse
|
23
|
Bumiller-Bini V, de Freitas Oliveira-Toré C, Carvalho TM, Kretzschmar GC, Gonçalves LB, Alencar NDM, Gasparetto MA, Beltrame MH, Winter Boldt AB. MASPs at the crossroad between the complement and the coagulation cascades - the case for COVID-19. Genet Mol Biol 2021; 44:e20200199. [PMID: 33729332 PMCID: PMC7982787 DOI: 10.1590/1678-4685-gmb-2020-0199] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/20/2021] [Indexed: 01/08/2023] Open
Abstract
Components of the complement system and atypical parameters of coagulation were reported in COVID-19 patients, as well as the exacerbation of the inflammation and coagulation activity. Mannose binding lectin (MBL)- associated serine proteases (MASPs) play an important role in viral recognition and subsequent activation of the lectin pathway of the complement system and blood coagulation, connecting both processes. Genetic variants of MASP1 and MASP2 genes are further associated with different levels and functional efficiency of their encoded proteins, modulating susceptibility and severity to diseases. Our review highlights the possible role of MASPs in SARS-COV-2 binding and activation of the lectin pathway and blood coagulation cascades, as well as their associations with comorbidities of COVID-19. MASP-1 and/or MASP-2 present an increased expression in patients with COVID-19 risk factors: diabetes, arterial hypertension and cardiovascular disease, chronic kidney disease, chronic obstructive pulmonary disease, and cerebrovascular disease. Based also on the positive results of COVID-19 patients with anti-MASP-2 antibody, we propose the use of MASPs as a possible biomarker of the progression of COVID-19 and the investigation of new treatment strategies taking into consideration the dual role of MASPs, including MASP inhibitors as promising therapeutic targets against COVID-19.
Collapse
Affiliation(s)
- Valéria Bumiller-Bini
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Laboratório de Genética Molecular Humana, Curitiba, PR, Brazil
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Programa de Pós-Graduação em Genética, Curitiba, PR, Brazil
| | - Camila de Freitas Oliveira-Toré
- Universidade Federal do Paraná (UFPR), Programa de Pós-Graduação em Medicina Interna e Ciências da Saúde, Laboratório de Imunopatologia Molecular, Curitiba, PR, Brazil
| | - Tamyres Mingorance Carvalho
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Programa de Pós-Graduação em Genética, Curitiba, PR, Brazil
- Universidade Federal do Paraná, Departamento de Genética, Laboratório de Citogenética Humana e Oncogenética, Curitiba, PR, Brazil
| | - Gabriela Canalli Kretzschmar
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Laboratório de Genética Molecular Humana, Curitiba, PR, Brazil
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Programa de Pós-Graduação em Genética, Curitiba, PR, Brazil
| | - Letícia Boslooper Gonçalves
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Programa de Pós-Graduação em Genética, Curitiba, PR, Brazil
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Laboratório de Imunogenética e Histocompatibilidade (LIGH), Curitiba, PR, Brazil
| | - Nina de Moura Alencar
- Fundação Oswaldo Cruz (Fiocruz), Instituto Carlos Chagas, Programa de Pós-Graduação em Biociências e Biotecnologia, Laboratório de Virologia Molecular, Curitiba, PR, Brazil
| | - Miguel Angelo Gasparetto
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Laboratório de Genética Molecular Humana, Curitiba, PR, Brazil
| | - Marcia Holsbach Beltrame
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Laboratório de Genética Molecular Humana, Curitiba, PR, Brazil
| | - Angelica Beate Winter Boldt
- Universidade Federal do Paraná (UFPR), Departamento de Genética, Laboratório de Genética Molecular Humana, Curitiba, PR, Brazil
| |
Collapse
|
24
|
Solidified glomerulosclerosis, identified using single glomerular proteomics, predicts end-stage renal disease in Chinese patients with type 2 diabetes. Sci Rep 2021; 11:4658. [PMID: 33633132 PMCID: PMC7907371 DOI: 10.1038/s41598-021-83856-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Few histological prognostic indicators for end-stage renal disease (ESRD) have been validated in diabetic patients. This biopsy-based study aimed to identify nephropathological risk factors for ESRD in Chinese patients with type 2 diabetes. Histological features of 322 Chinese type 2 diabetic patients with biopsy-confirmed diabetic nephropathy (DN) were retrospectively analysed. Cox proportional hazards analysis was used to estimate the hazard ratio (HR) for ESRD. Single glomerular proteomics and immunohistochemistry were used to identify differentially expressed proteins and enriched pathways in glomeruli. During the median follow-up period of 24 months, 144 (45%) patients progressed to ESRD. In multivariable models, the Renal Pathology Society classification failed to predict ESRD, although the solidified glomerulosclerosis (score 1: HR 1.65, 95% confidence interval [CI] 1.04-2.60; score 2: HR 2.48, 95% CI 1.40-4.37) and extracapillary hypercellularity (HR 2.68, 95% CI 1.55-4.62) were identified as independent risk factors. Additionally, single glomerular proteomics, combined with immunohistochemistry, revealed that complement C9 and apolipoprotein E were highly expressed in solidified glomerulosclerosis. Therefore, solidified glomerulosclerosis and extracapillary hypercellularity predict diabetic ESRD in Chinese patients. Single glomerular proteomics identified solidified glomerulosclerosis as a unique pathological change that may be associated with complement overactivation and abnormal lipid metabolism.
Collapse
|
25
|
Budge K, Dellepiane S, Yu SMW, Cravedi P. Complement, a Therapeutic Target in Diabetic Kidney Disease. Front Med (Lausanne) 2021; 7:599236. [PMID: 33553201 PMCID: PMC7858668 DOI: 10.3389/fmed.2020.599236] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/21/2020] [Indexed: 01/15/2023] Open
Abstract
Currently available treatments of diabetic kidney disease (DKD) remain limited despite improved understanding of DKD pathophysiology. The complement system is a central part of innate immunity, but its dysregulated activation is detrimental and results in systemic diseases with overt inflammation. Growing evidence suggests complement activation in DKD. With existent drugs and clinical success of treating other kidney diseases, complement inhibition has emerged as a potential novel therapy to halt the progression of DKD. This article will review DKD, the complement system's role in diabetic and non-diabetic disease, and the potential benefits of complement targeting therapies especially for DKD patients.
Collapse
Affiliation(s)
- Kelly Budge
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sergio Dellepiane
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Samuel Mon-Wei Yu
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Paolo Cravedi
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
26
|
Behl T, Kaur I, Sehgal A, Sharma E, Kumar A, Grover M, Bungau S. Unfolding Nrf2 in diabetes mellitus. Mol Biol Rep 2021; 48:927-939. [PMID: 33389540 DOI: 10.1007/s11033-020-06081-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/10/2020] [Indexed: 11/24/2022]
Abstract
In spite of much awareness, diabetes mellitus continues to remain one of major reasons for mortality and morbidity rate all over the globe. Free radicals cause oxidative stress which is responsible for causing diabetes. The recent advancements in elucidation of ARE/keap1/Nrf2 pathway can help in better understanding of diabetes mellitus. Various clinical trials and animal studies have shown the promising effect of Nrf2 pathway in reversing diabetes by counteracting with the oxidative stress produced. The gene is known to dissociate from Keap1 on coming in contact with such stresses to show preventive and prognosis effect. The Nrf2 gene has been marked as a molecular player in dealing with wide intracellular as well as extracellular cellular interactions in different diseases. The regulation of this gene gives some transcription factor that contain antioxidant response elements (ARE) in their promoter region and thus are responsible for encoding certain proteins involved in regulation of metabolic and detoxifying enzymes.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Eshita Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhuri Grover
- B.S. Anangpuria Institute of Pharmacy, Alampur, Haryana, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
27
|
Lu C, Fan G, Wang D. Akebia Saponin D ameliorated kidney injury and exerted anti-inflammatory and anti-apoptotic effects in diabetic nephropathy by activation of NRF2/HO-1 and inhibition of NF-KB pathway. Int Immunopharmacol 2020; 84:106467. [PMID: 32344355 DOI: 10.1016/j.intimp.2020.106467] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy (DN), a common microvascular complication of type 2 diabetes mellitus (T2DM), causes increasing mortality and morbidity due to its high prevalence and severe consequences. Hence, it is urgent to search for effective agents that provide new insights into novel molecular therapeutic targets for DN. This study was designed to investigate the critical role of Akebia saponin D (ASD) in kidney damage, inflammation and apoptosis of renal tubular cells in DN. To probe the protective effects of ASD on DN in vivo, diabetes mellitus model was established by intraperitoneal (ip) injection of STZ (60 mg/kg) for 5 days consecutively. Besides, HG-induced human renal tubular cells (HK-2) were used to analyze the defined effects and underlying mechanism of ASD on DN in vitro. Blood glucose, insulin, serum creatinine (Scr), blood urea nitrogen (BUN), renal injury, inflammation, oxidative stress and apoptosis of renal tubular cells were respectively measured and evaluated. ASD prevented kidney damage, improved renal function and inflammatory reaction, ameliorated oxidative stress and inhibited apoptosis of renal tubular cells in DN mice via activation of NRF2/HO-1 pathway and inhibition of NF-KB pathway.
Collapse
Affiliation(s)
- Congcong Lu
- Dongying People's Hospital, Dongying City, Shandong Province, China
| | - Guoxia Fan
- Dongying People's Hospital, Dongying City, Shandong Province, China
| | - Dianyun Wang
- Dongying People's Hospital, Dongying City, Shandong Province, China.
| |
Collapse
|
28
|
Shim K, Begum R, Yang C, Wang H. Complement activation in obesity, insulin resistance, and type 2 diabetes mellitus. World J Diabetes 2020; 11:1-12. [PMID: 31938469 PMCID: PMC6927818 DOI: 10.4239/wjd.v11.i1.1] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 11/07/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023] Open
Abstract
Amplified inflammatory reaction has been observed to be involved in cardiometabolic diseases such as obesity, insulin resistance, diabetes, dyslipidemia, and atherosclerosis. The complement system was originally viewed as a supportive first line of defense against microbial invaders, and research over the past decade has come to appreciate that the functions of the complement system extend beyond the defense and elimination of microbes, involving in such diverse processes as clearance of the immune complexes, complementing T and B cell immune functions, tissue regeneration, and metabolism. The focus of this review is to summarize the role of the activation of complement system and the initiation and progression of metabolic disorders including obesity, insulin resistance and diabetes mellitus. In addition, we briefly describe the interaction of the activation of the complement system with diabetic complications such as diabetic retinopathy, nephropathy and neuropathy, highlighting that targeting complement system therapeutics could be one of possible routes to slow down those aforementioned diabetic complications.
Collapse
Affiliation(s)
- Kyumin Shim
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
| | - Rayhana Begum
- Department of Pharmacy, Primeasia University, Dhaka 1213, Bangladesh
| | - Catherine Yang
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
- California Northstate University College of Graduate Studies, Elk Grove, CA 95757, United States
| | - Hongbin Wang
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
- California Northstate University College of Graduate Studies, Elk Grove, CA 95757, United States
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, United States
| |
Collapse
|
29
|
Wang T, Gao Y, Wang X, Shi Y, Xu J, Wu B, He J, Li Y. Calpain-10 drives podocyte apoptosis and renal injury in diabetic nephropathy. Diabetes Metab Syndr Obes 2019; 12:1811-1820. [PMID: 31571956 PMCID: PMC6750010 DOI: 10.2147/dmso.s217924] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a progressive microvascular complication of diabetes mellitus (DM), driven largely by podocyte apoptosis. The cysteine protease Calpain 10 is known to augment apoptosis and necrosis, and is a potential therapeutic target in DN. METHODS Type 2 diabetes was induced in SD rats by high-fat diet (HFD) feeding and streptozotocin (STZ) injections, and simulated in vitro by culturing conditionally immortalized mouse podocytes in hyperlipidemic (PA, 100 μM) conditions. The rate of apoptosis in the renal tissues and cultured podocytes was determined by TUNEL assay. The expression of Calpain 10 and its biological effects were assayed by real-time PCR, Western blotting, immunofluorescence and electron microscopy. RESULTS Calpain 10 was up-regulated in the kidneys of DN rats, as well as immortalized mouse podocytes. High levels of Calpain 10 was associated with renal dysfunction and tissue destruction, and podocyte injury and apoptosis. Knockdown of Calpain 10 protected podocytes by decreasing apoptosis rate, and upregulated nephrin. CONCLUSION Calpain 10 is a pro-apoptotic factor in DN, and can be targeted for treating glomerular diseases.
Collapse
Affiliation(s)
- Tao Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yanbin Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, People’s Republic of China
- Correspondence: Yanbin GaoSchool of Traditional Chinese Medicine, Capital Medical University, No. 10, Youanmenwai, Xitoutiao, Fengtai District, Beijing100069, People’s Republic of ChinaTel +86 108 391 1720Email
| | - Xiaolei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yimin Shi
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Jiayi Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Bingjie Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Jiaxin He
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yimeng Li
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, Beijing, People’s Republic of China
| |
Collapse
|