1
|
Quan W, Wang L. Tripartite motif containing 23 functions as a critical regulator in macrophages to control the pathological feature of diabetic nephropathy. Int Urol Nephrol 2022; 55:1263-1270. [PMID: 36449224 DOI: 10.1007/s11255-022-03419-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a kidney disease resulting from diabetes. Macrophages and macrophage-mediated inflammation contributed to the development of DN. Tripartite motif containing 23 (TRIM23) is E3 ligase and has been involved in inflammation. Until now, the precise roles of TRIM23 in DN are not described yet. Therefore, we evaluated the functions of TRIM23 in DN. METHODS We generated mice with TRIM23 deficiency in macrophages. We also established diabetic mice model. The expression of TRIM23 was measured in diabetic animals. The DN symptoms were compared between diabetic wild-type (WT) mice and TRIM23 conditional knock out mice. The cytokine expression, ubiquitination of TAB2, and interactions between TAB2 and IKK were compared in oxidized low-density lipoprotein (oxLDL) or lipopolysaccharides (LPS)-treated WT and TRIM23-deficient macrophages. RESULTS Upregulation of TRIM23 was observed in diabetic mice and LPS or oxLDL-treated macrophages. Diabetic mice with TRIM23 deficiency in macrophages had attenuated DN symptoms. TRIM23-deficient macrophages had decreased pro-inflammatory cytokines production after oxLDL or LPS stimulation. TRIM23 was predicted to interact with TAB2. The ubiquitination of TAB2 was abolished in oxLDL-treated TRIM23-deficient macrophages, which correlated with decreased activation of IKK. CONCLUSION TRIM23 regulates inflammation in macrophages and plays important role in DN.
Collapse
Affiliation(s)
- Wei Quan
- Department of Endocrinology, Daqing Oilfield General Hospital, No. 9, Zhongkang Street, Sartu District, Daqing, 163000, Heilongjiang, China
| | - Lin Wang
- Department of Endocrinology, Daqing Oilfield General Hospital, No. 9, Zhongkang Street, Sartu District, Daqing, 163000, Heilongjiang, China.
| |
Collapse
|
2
|
Bao T, Liu J, Leng J, Cai L. The cGAS-STING pathway: more than fighting against viruses and cancer. Cell Biosci 2021; 11:209. [PMID: 34906241 PMCID: PMC8670263 DOI: 10.1186/s13578-021-00724-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 12/02/2021] [Indexed: 01/07/2023] Open
Abstract
In the classic Cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway, downstream signals can control the production of type I interferon and nuclear factor kappa-light-chain-enhancer of activated B cells to promote the activation of pro-inflammatory molecules, which are mainly induced during antiviral responses. However, with progress in this area of research, studies focused on autoimmune diseases and chronic inflammatory conditions that may be relevant to cGAS–STING pathways have been conducted. This review mainly highlights the functions of the cGAS–STING pathway in chronic inflammatory diseases. Importantly, the cGAS–STING pathway has a major impact on lipid metabolism. Different research groups have confirmed that the cGAS–STING pathway plays an important role in the chronic inflammatory status in various organs. However, this pathway has not been studied in depth in diabetes and diabetes-related complications. Current research on the cGAS–STING pathway has shown that the targeted therapy of diseases that may be caused by inflammation via the cGAS–STING pathway has promising outcomes.
Collapse
Affiliation(s)
- Terigen Bao
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China.,Department of Pediatrics, The Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Jia Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jiyan Leng
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Lu Cai
- Department of Pediatrics, The Pediatric Research Institute, The University of Louisville School of Medicine, Louisville, KY, 40292, USA.,Departments of Pharmacology and Toxicology, The University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
3
|
Abstract
Epigenetic modifications have been implicated to mediate several complications of diabetes mellitus (DM), especially nephropathy and retinopathy. Our aim was to ascertain whether epigenetic alterations in whole blood discriminate among patients with DM with normal, delayed, and rapid gastric emptying (GE).
Collapse
|
4
|
Mosterd CM, Kanbay M, van den Born BJH, van Raalte DH, Rampanelli E. Intestinal microbiota and diabetic kidney diseases: the Role of microbiota and derived metabolites inmodulation of renal inflammation and disease progression. Best Pract Res Clin Endocrinol Metab 2021; 35:101484. [PMID: 33546983 DOI: 10.1016/j.beem.2021.101484] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetic kidney disease (DKD) represents a growing public health burden and is the leading cause of end-stage kidney diseases. In recent years, host-gut microbiota interactions have emerged as an integral part for host homeostasis. In the context of nephropathies, mounting evidence supports a bidirectional microbiota-kidney crosstalk, which becomes particularly manifest during progressive kidney dysfunction. Indeed, in chronic kidney disease (CKD), the "healthy" microbiota structure is disrupted and intestinal microbes produce large quantities of uremic solutes responsible for renal damage; on the other hand, the uremic state, fueled by reduced renal clearance, causes shifts in microbial metabolism and composition, hence creating a vicious cycle in which dysbiosis and renal dysfunction are progressively worsened. In this review, we will summarize the evidence from clinical/experimental studies concerning the occurrence of gut dysbiosis in diabetic and non-diabetic CKD, discuss the functional consequences of dysbiosis for CKD progression and debate putative therapeutic interventions targeting the intestinal microbiome.
Collapse
Affiliation(s)
- C M Mosterd
- Department of Internal and Vascular Medicine, Amsterdam UMC, Location VUmc, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| | - M Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - B J H van den Born
- Department of Nephrology and Vascular Medicine, Amsterdam UMC, Location AMC, the Netherlands
| | - D H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam UMC, Location VUmc, Diabetes Center, Amsterdam, the Netherlands
| | - E Rampanelli
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, Amsterdam Cardiovascular Sciences, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands.
| |
Collapse
|
5
|
A novel salviadione derivative, compound 15a, attenuates diabetes-induced renal injury by inhibiting NF-κB-mediated inflammatory responses. Toxicol Appl Pharmacol 2020; 409:115322. [PMID: 33171189 DOI: 10.1016/j.taap.2020.115322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 01/01/2023]
Abstract
Diabetic nephropathy is the leading cause of renal failure worldwide. Elevated inflammatory signaling has been shown to lead to deterioration of renal function in human and experimental diabetes. We recently developed a salviadione derivative (compound 15a) that prevented microbial lipopolysaccharide-induced inflammatory responses, which are largely driven by nuclear factor-κB (NF-κB). In the present study, we have tested the hypothesis that 15a will protect kidneys from diabetes-induced dysfunction by suppressing NF-κB activation and inflammatory signaling. Treatment of diabetic mice with 15a inhibited diabetes-induced renal fibrosis, NF-κB activation, and upregulation of proinflammatory cytokines. Histologically, kidney specimens from diabetic mice treated with 15a were indistinguishable from non-diabetic controls. We confirmed our findings in cultured renal tubular epithelial cells exposed to high levels of glucose. In these cultured cells, 15a pretreatment prevented high glucose-induced NF-κB activation and expression of inflammatory cytokines. These protective effects were also reflected in reduced levels of proteins involved in matrix expansion. Overall, our studies show that a salviadione derivative, 15a, is effective in suppressing diabetes-induced NF-κB activation and inflammatory signaling.
Collapse
|
6
|
Zheng ZC, Zhu W, Lei L, Liu XQ, Wu YG. Wogonin Ameliorates Renal Inflammation and Fibrosis by Inhibiting NF-κB and TGF-β1/Smad3 Signaling Pathways in Diabetic Nephropathy. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4135-4148. [PMID: 33116403 PMCID: PMC7549498 DOI: 10.2147/dddt.s274256] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022]
Abstract
Introduction Diabetic nephropathy (DN) has become an increasing threat to health, and inflammation and fibrosis play important roles in its progression. Wogonin, a flavonoid, has been proven to suppress inflammation and fibrosis in various diseases, including acute kidney injury. This study aimed at investigating the effect of wogonin on diabetes-induced renal inflammation and fibrosis. Materials and Methods Streptozotocin (STZ)-induced diabetic mouse models received gavage doses of wogonin (10, 20, and 40 mg/kg) for 12 weeks. Metabolic indices from blood and urine and pathological damage of glomerulus in the diabetic model were assessed. Glomerular mesangial cells SV40 were cultured in high glucose (HG) medium containing wogonin at concentrations of 1.5825, 3.125, and 6.25 μg/mL for 24 h. Inflammation and fibrosis indices were evaluated by histopathological, Western blotting, and PCR analyses. Results Wogonin treatment ameliorated albuminuria and histopathological lesions in diabetic mice. Inflammatory cytokines, such as monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and related signaling pathway NF-κB were downregulated after the administration of wogonin in vivo and in vitro. Furthermore, wogonin reduced the expression of extracellular matrix (ECM), including fibronectin (FN), collagen IV (Col-IV), α-smooth muscle actin (α-SMA), and transforming growth factor-β1 (TGF-β1) in the kidneys of diabetic mice and HG-induced mesangial cells. Moreover, the inhibition of TGF-β1/Smad3 pathway might be responsible for these changes. Conclusion Wogonin may ameliorate renal inflammation and fibrosis in diabetic nephropathy by inhibiting the NF-κB and TGF-β1/Smad3 signaling pathways.
Collapse
Affiliation(s)
- Zhi-Chao Zheng
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Wei Zhu
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Lei Lei
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xue-Qi Liu
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yong-Gui Wu
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| |
Collapse
|
7
|
Rayego-Mateos S, Morgado-Pascual JL, Valdivielso JM, Sanz AB, Bosch-Panadero E, Rodrigues-Díez RR, Egido J, Ortiz A, González-Parra E, Ruiz-Ortega M. TRAF3 Modulation: Novel Mechanism for the Anti-inflammatory Effects of the Vitamin D Receptor Agonist Paricalcitol in Renal Disease. J Am Soc Nephrol 2020; 31:2026-2042. [PMID: 32631974 DOI: 10.1681/asn.2019111206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND CKD leads to vitamin D deficiency. Treatment with vitamin D receptor agonists (VDRAs) may have nephroprotective and anti-inflammatory actions, but their mechanisms of action are poorly understood. METHODS Modulation of the noncanonical NF-κB2 pathway and its component TNF receptor-associated factor 3 (TRAF3) by the VDRA paricalcitol was studied in PBMCs from patients with ESKD, cytokine-stimulated cells, and preclinical kidney injury models. RESULTS In PBMCs isolated from patients with ESKD, TRAF3 protein levels were lower than in healthy controls. This finding was associated with evidence of noncanonical NF-κB2 activation and a proinflammatory state. However, PBMCs from patients with ESKD treated with paricalcitol did not exhibit these features. Experiments in cultured cells confirmed the link between TRAF3 and NF-κB2/inflammation. Decreased TRAF3 ubiquitination in K48-linked chains and cIAP1-TRAF3 interaction mediated the mechanisms of paricalcitol action.TRAF3 overexpression by CRISPR/Cas9 technology mimicked VDRA's effects. In a preclinical model of kidney injury, paricalcitol inhibited renal NF-κB2 activation and decreased renal inflammation. In VDR knockout mice with renal injury, paricalcitol prevented TRAF3 downregulation and NF-κB2-dependent gene upregulation, suggesting a VDR-independent anti-inflammatory effect of paricalcitol. CONCLUSIONS These data suggest the anti-inflammatory actions of paricalcitol depend on TRAF3 modulation and subsequent inhibition of the noncanonical NF-κB2 pathway, identifying a novel mechanism for VDRA's effects. Circulating TRAF3 levels could be a biomarker of renal damage associated with the inflammatory state.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain
| | - Jose Luis Morgado-Pascual
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - José Manuel Valdivielso
- Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - Ana Belén Sanz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Enrique Bosch-Panadero
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Raúl R Rodrigues-Díez
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz.Universidad Autónoma. 28040 Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM). 28029 Madrid, Spain
| | - Alberto Ortiz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Emilio González-Parra
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain .,REDinREN (Red de Investigación Renal), Madrid, Spain
| |
Collapse
|
8
|
Pathogenic Pathways and Therapeutic Approaches Targeting Inflammation in Diabetic Nephropathy. Int J Mol Sci 2020; 21:ijms21113798. [PMID: 32471207 PMCID: PMC7312633 DOI: 10.3390/ijms21113798] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy (DN) is associated with an increased morbidity and mortality, resulting in elevated cost for public health systems. DN is the main cause of chronic kidney disease (CKD) and its incidence increases the number of patients that develop the end-stage renal disease (ESRD). There are growing epidemiological and preclinical evidence about the close relationship between inflammatory response and the occurrence and progression of DN. Several anti-inflammatory strategies targeting specific inflammatory mediators (cell adhesion molecules, chemokines and cytokines) and intracellular signaling pathways have shown beneficial effects in experimental models of DN, decreasing proteinuria and renal lesions. A number of inflammatory molecules have been shown useful to identify diabetic patients at high risk of developing renal complications. In this review, we focus on the key role of inflammation in the genesis and progression of DN, with a special interest in effector molecules and activated intracellular pathways leading to renal damage, as well as a comprehensive update of new therapeutic strategies targeting inflammation to prevent and/or retard renal injury.
Collapse
|
9
|
Alleboina S, Wong T, Singh MV, Dokun AO. Inhibition of protein kinase C beta phosphorylation activates nuclear factor-kappa B and improves postischemic recovery in type 1 diabetes. Exp Biol Med (Maywood) 2020; 245:785-796. [PMID: 32326759 PMCID: PMC7273893 DOI: 10.1177/1535370220920832] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/30/2020] [Indexed: 01/01/2023] Open
Abstract
IMPACT STATEMENT Diabetes worsens the outcomes of peripheral arterial disease (PAD) likely in part through inducing chronic inflammation. However, in PAD, recovery requires the nuclear factor-kappa B (NF-κB) activation, a known contributor to inflammation. Our study shows that individually, both ischemia and high glucose activate the canonical and non-canonical arms of the NF-κB pathways. We show for the first time that prolonged high glucose specifically impairs ischemia-induced activation of the canonical NF-κB pathway through activation of protein kinase C beta (PKCβ). Accordingly, inhibition of PKCβ restores the ischemia-induced NF-κB activity both in vitroin endothelial cells and in vivoin hind limbs of type 1 diabetic mice and improves perfusion recovery after experimental PAD. Thus, this study provides a mechanistic insight into how diabetes contributes to poor outcomes in PAD and a potential translational approach to improve PAD outcomes.
Collapse
Affiliation(s)
- Satyanarayana Alleboina
- Division of Endocrinology, Diabetes and Metabolism,
University of Tennessee Health Sciences Center, Memphis, TN 38103,
USA
| | - Thomas Wong
- Division of Endocrinology and Metabolism, Carver
College of Medicine, University of Iowa, Iowa City, IA 52242,
USA
| | - Madhu V Singh
- Division of Endocrinology and Metabolism, Carver
College of Medicine, University of Iowa, Iowa City, IA 52242,
USA
| | - Ayotunde O Dokun
- Division of Endocrinology and Metabolism, Carver
College of Medicine, University of Iowa, Iowa City, IA 52242,
USA
| |
Collapse
|
10
|
Lee SH, Hwang SM, Kang DH, Yang HJ. Brain education-based meditation for patients with hypertension and/or type 2 diabetes: A pilot randomized controlled trial. Medicine (Baltimore) 2019; 98:e15574. [PMID: 31083232 PMCID: PMC6531095 DOI: 10.1097/md.0000000000015574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Hypertension and type 2 diabetes are chronic diseases, which generally require lifetime care. Meditation and yoga can be complementary to pharmacological therapies according to the scientific evidences so far. Brain education-based meditation (BEM) is a technique, which has been known to change brain structure, psychology, and physiology of healthy adult participants. This randomized, nonblinded pilot trial aimed to examine whether BEM affects the conditions of patients with hypertension and/or type 2 diabetes compared with health education classes. METHODS We randomly allocated 48 patients with hypertension and/or type 2 diabetes to BEM (n = 24) or health education (n = 24) classes in the Ulsan Junggu Public Health Center in Korea, where the classes were run during the same period and explored the impact of 8-week practice on the serum glutamic-oxaloacetic transaminase, serum glutamic pyruvic transaminase, gamma glutamyl transpeptidase, creatinine, high-density lipoprotein cholesterol, and low-density lipoprotein (LDL) cholesterol. Total RNA was extracted to examine inflammatory gene expressions from the whole blood using PAXgene blood RNA System. In addition, self-reports on mental/physical health were evaluated. The Student's t test, chi-squared test, and analysis of covariance were used for statistical analysis. RESULTS The number of people who participated until the completion of the study was 14 in the control and 21 in the BEM group. After 8 weeks, LDL cholesterol level was significantly decreased in the BEM group after the intervention (13.82 mg/dL reduction, P < .05), while it was not significantly altered in the control group. The expression of inflammatory genes was significantly reduced after 8 weeks of the BEM training (0.3-, 0.5-, and 0.2-fold change for NFKB2, RELA, and IL1B, respectively, all P < .05). In the item analysis of mental/physical health self-reports, a significant improvement was confirmed as follows: increases in focus, confidence, relaxation, and happiness; decreases in fatigue, anger, and loneliness (all P < .05). There were no important adverse events or side-effects by BEM intervention. CONCLUSION Compared to health education, BEM helps lower LDL cholesterol level and the inflammatory gene expression in the patients with hypertension and/or type 2 diabetes. Moreover, BEM induces positive effects on the self-reported mental/physical states, warranting further study.
Collapse
Affiliation(s)
- Seung-Ho Lee
- Department of Brain Education
- Department of Counseling Psychology, University of Brain Education, Dongnam-gu, Cheonan-si, Chungcheongnam-do
| | | | - Do-Hyung Kang
- Emotional Information and Communication Technology Association, Dae-jeon
| | - Hyun-Jeong Yang
- Department of Brain Education
- Department of Integrative Biosciences, University of Brain Education, Dongnam-gu, Cheonan-si, Chungcheongnam-do
- Korea Institute of Brain Science, Seoul, Republic of Korea
| |
Collapse
|
11
|
Takenaka T, Kobori H, Miyazaki T, Suzuki H, Nishiyama A, Ishii N, Yamashita M, Hayashi M. Klotho protein supplementation reduces blood pressure and renal hypertrophy in db/db mice, a model of type 2 diabetes. Acta Physiol (Oxf) 2019; 225:e13190. [PMID: 30251773 DOI: 10.1111/apha.13190] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 12/18/2022]
Abstract
AIMS Klotho interacts with various membrane proteins, such as receptors for transforming growth factor (TGF)-β and insulin-like growth factor (IGF), to alter their function. Renal expression of klotho is diminished in diabetes. The present study examined whether exogenous klotho protein supplementation ameliorates kidney injury and renin-angiotensin system (RAS) in db/db mice. METHODS We investigated the effects of klotho supplementation on diabetic kidney injury and RAS. Recombinant human klotho protein (10 μg/kg/d) was administered to db/db mice daily. RESULTS Klotho protein supplementation reduced kidney weight, systolic blood pressure (SBP), albuminuria, glomerular filtration rate, and 8-epi-prostaglandin F2α excretion without affecting body weight. Although klotho supplementation did not alter glycated albumin, it reduced renal angiotensin II levels associated with reduced renal expression of angiotensinogen. Klotho supplementation improved renal expression of superoxide dismutase (SOD), and endogenous renal expression of klotho. Klotho supplementation reduced the levels of hypoxia-inducible factor, phosphorylated Akt, and phosphorylated mTOR and decreased the renal expression of TGF-β, tumour necrosis factor (TNF), and fibronectin. CONCLUSIONS These data indicate that klotho supplementation reduces blood pressure and albuminuria along with ameliorating renal RAS activation in db/db mice. Furthermore, these results suggest that klotho inhibits IGF signalling, induces SOD expression to reduce oxidative stress, and suppresses Akt-mTOR signalling to inhibit abnormal kidney growth. Collectively, the results suggest that klotho inhibits TGF-β and TNF signalling, resulting in a decline in renal fibrosis.
Collapse
Affiliation(s)
- Tsuneo Takenaka
- International University of Health and Welfare; Minato Japan
| | - Hiroyuki Kobori
- International University of Health and Welfare; Minato Japan
| | | | | | | | | | | | | |
Collapse
|
12
|
Moreno JA, Gomez-Guerrero C, Mas S, Sanz AB, Lorenzo O, Ruiz-Ortega M, Opazo L, Mezzano S, Egido J. Targeting inflammation in diabetic nephropathy: a tale of hope. Expert Opin Investig Drugs 2018; 27:917-930. [PMID: 30334635 DOI: 10.1080/13543784.2018.1538352] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Diabetic nephropathy (DN) is the leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD). Beyond the new anti-diabetic drugs that possess markedly cardiovascular and renal protective effects, no novel direct therapies for DN have become available on the market in the last twenty years. Recently well-designed clinical trials for the treatment of DN, with attractive pathogenetic rationale, e.g. bardoxolone and atrasentan, were canceled or stopped because of safety concerns or lack of reaching the end points, respectively. AREAS COVERED In this review, we focus on the involvement of inflammation in the pathogenesis of DN. We update information from recent experimental and clinical studies that reported beneficial effects of several agents targeting chemokines, cytokines, transcription factors and kinases as well as several compounds with anti-inflammatory properties on DN. EXPERT OPINION Inflammation plays a key role in the DN progression. Preclinical studies have identified several anti-inflammatory molecules that effective decrease albuminuria and/or proteinuria. However, limited clinical trials in humans have been performed to confirm these results. Inhibitors of CCL2/CCR2, IL-1β and JAK/STAT pathways, and Nrf2 inducers are promising therapeutic options to improve the renal outcome of patients with DN, but appropriate clinical trials are necessary.
Collapse
Affiliation(s)
- Juan Antonio Moreno
- a Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz University Hospital-Health Research Institute (FIIS-FJD) , Autonoma University of Madrid (UAM) , Madrid , Spain
| | - Carmen Gomez-Guerrero
- a Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz University Hospital-Health Research Institute (FIIS-FJD) , Autonoma University of Madrid (UAM) , Madrid , Spain.,b Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM) , Madrid , Spain
| | - Sebastian Mas
- a Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz University Hospital-Health Research Institute (FIIS-FJD) , Autonoma University of Madrid (UAM) , Madrid , Spain.,b Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM) , Madrid , Spain
| | - Ana Belen Sanz
- a Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz University Hospital-Health Research Institute (FIIS-FJD) , Autonoma University of Madrid (UAM) , Madrid , Spain.,c Red de Investigación Renal (REDinREN) , Spain
| | - Oscar Lorenzo
- a Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz University Hospital-Health Research Institute (FIIS-FJD) , Autonoma University of Madrid (UAM) , Madrid , Spain.,b Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM) , Madrid , Spain
| | - Marta Ruiz-Ortega
- c Red de Investigación Renal (REDinREN) , Spain.,d Biology in Renal Diseases Laboratory , FIIS-FJD, UAM , Madrid , Spain
| | - Lucas Opazo
- e Division of Nephrology, School of Medicine , Universidad Austral , Valdivia , Chile
| | - Sergio Mezzano
- e Division of Nephrology, School of Medicine , Universidad Austral , Valdivia , Chile
| | - Jesus Egido
- a Renal, Vascular and Diabetes Research Laboratory, Fundacion Jimenez Diaz University Hospital-Health Research Institute (FIIS-FJD) , Autonoma University of Madrid (UAM) , Madrid , Spain.,b Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM) , Madrid , Spain
| |
Collapse
|
13
|
Gene variants in the NF-KB pathway (NFKB1, NFKBIA, NFKBIZ) and their association with type 2 diabetes and impaired renal function. Hum Immunol 2018; 79:494-498. [DOI: 10.1016/j.humimm.2018.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 03/06/2018] [Accepted: 03/26/2018] [Indexed: 12/29/2022]
|
14
|
Grinberg-Bleyer Y, Caron R, Seeley JJ, De Silva NS, Schindler CW, Hayden MS, Klein U, Ghosh S. The Alternative NF-κB Pathway in Regulatory T Cell Homeostasis and Suppressive Function. THE JOURNAL OF IMMUNOLOGY 2018; 200:2362-2371. [PMID: 29459403 DOI: 10.4049/jimmunol.1800042] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) are essential regulators of immune responses. Perturbation of Treg homeostasis or function can lead to uncontrolled inflammation and autoimmunity. Therefore, understanding the molecular mechanisms involved in Treg biology remains an active area of investigation. It has been shown previously that the NF-κB family of transcription factors, in particular, the canonical pathway subunits, c-Rel and p65, are crucial for the development, maintenance, and function of Tregs. However, the role of the alternative NF-κB pathway components, p100 and RelB, in Treg biology remains unclear. In this article, we show that conditional deletion of the p100 gene, nfkb2, in Tregs, resulted in massive inflammation because of impaired suppressive function of nfkb2-deficient Tregs. Surprisingly, mice lacking RelB in Tregs did not exhibit the same phenotype. Instead, deletion of both relb and nfkb2 rescued the inflammatory phenotype, demonstrating an essential role for p100 as an inhibitor of RelB in Tregs. Our data therefore illustrate a new role for the alternative NF-κB signaling pathway in Tregs that has implications for the understanding of molecular pathways driving tolerance and immunity.
Collapse
Affiliation(s)
- Yenkel Grinberg-Bleyer
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Rachel Caron
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - John J Seeley
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Nilushi S De Silva
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Christian W Schindler
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Matthew S Hayden
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Ulf Klein
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032.,Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032;
| |
Collapse
|
15
|
Rayego-Mateos S, Morgado-Pascual JL, Rodrigues-Diez RR, Rodrigues-Diez R, Falke LL, Mezzano S, Ortiz A, Egido J, Goldschmeding R, Ruiz-Ortega M. Connective tissue growth factor induces renal fibrosis via epidermal growth factor receptor activation. J Pathol 2018; 244:227-241. [PMID: 29160908 DOI: 10.1002/path.5007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/20/2017] [Accepted: 11/14/2017] [Indexed: 01/04/2023]
Abstract
Connective tissue growth factor (CCN2/CTGF) is a matricellular protein that is overexpressed in progressive human renal diseases, mainly in fibrotic areas. In vitro studies have demonstrated that CCN2 regulates the production of extracellular matrix (ECM) proteins and epithelial-mesenchymal transition (EMT), and could therefore contribute to renal fibrosis. CCN2 blockade ameliorates experimental renal damage, including diminution of ECM accumulation. We have reported that CCN2 and its C-terminal degradation product CCN2(IV) bind to epidermal growth factor receptor (EGFR) to modulate renal inflammation. However, the receptor involved in CCN2 profibrotic actions has not been described so far. Using a murine model of systemic administration of CCN2(IV), we have unveiled a fibrotic response in the kidney that was diminished by EGFR blockade. Additionally, in conditional CCN2 knockout mice, renal fibrosis elicited by folic acid-induced renal damage was prevented, and this was linked to inhibition of EGFR pathway activation. Our in vitro studies demonstrated a direct effect of CCN2 via the EGFR pathway on ECM production by fibroblasts and the induction of EMT in tubular epithelial cells. Our studies clearly show that the EGFR regulates CCN2 fibrotic signalling in the kidney, and suggest that EGFR pathway blockade could be a potential therapeutic option to block CCN2-mediated profibrotic effects in renal diseases. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| | - José Luis Morgado-Pascual
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| | | | - Raquel Rodrigues-Diez
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sergio Mezzano
- Division of Nephrology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz-UAM, School of Medicine, UAM, Madrid, Spain
| | - Jesús Egido
- IIS-Fundación Jiménez Díaz-UAM, School of Medicine, UAM, Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory. School of Medicine, Universidad Autónoma Madrid, Madrid, Spain
| |
Collapse
|
16
|
Abstract
The nuclear factor-κB (NF-κB) family of transcription factors is activated by canonical and non-canonical signalling pathways, which differ in both signalling components and biological functions. Recent studies have revealed important roles for the non-canonical NF-κB pathway in regulating different aspects of immune functions. Defects in non-canonical NF-κB signalling are associated with severe immune deficiencies, whereas dysregulated activation of this pathway contributes to the pathogenesis of various autoimmune and inflammatory diseases. Here we review the signalling mechanisms and the biological function of the non-canonical NF-κB pathway. We also discuss recent progress in elucidating the molecular mechanisms regulating non-canonical NF-κB pathway activation, which may provide new opportunities for therapeutic strategies.
Collapse
Affiliation(s)
- Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, MD Anderson Cancer Center UT Heath Graduate School of Biomedical Sciences, 7455 Fannin Street, Box 902, Houston, Texas 77030, USA
| |
Collapse
|
17
|
Hendrawati A, Winardi M. Effects of Quercetin and Omega-3 Combination on Nuclear Factor Kappa B (NFκB) Expression Level in Pancreatic Tissue of Rats with Type-2 Diabetes Mellitus. ACTA ACUST UNITED AC 2017. [DOI: 10.6000/1927-5951.2017.07.01.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
18
|
Expression Profiling of Genes Related to Endothelial Cells Biology in Patients with Type 2 Diabetes and Patients with Prediabetes. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1845638. [PMID: 27781209 PMCID: PMC5066000 DOI: 10.1155/2016/1845638] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 12/31/2022]
Abstract
Endothelial dysfunction appears to be an early sign indicating vascular damage and predicts the progression of atherosclerosis and cardiovascular disorders. Extensive clinical and experimental evidence suggests that endothelial dysfunction occurs in Type 2 Diabetes Mellitus (T2DM) and prediabetes patients. This study was carried out with an aim to appraise the expression levels in the peripheral blood of 84 genes related to endothelial cells biology in patients with diagnosed T2DM or prediabetes, trying to identify new genes whose expression might be changed under these pathological conditions. The study covered a total of 45 participants. The participants were divided into three groups: group 1, patients with T2DM; group 2, patients with prediabetes; group 3, control group. The gene expression analysis was performed using the Endothelial Cell Biology RT2 Profiler PCR Array. In the case of T2DM, 59 genes were found to be upregulated, and four genes were observed to be downregulated. In prediabetes patients, increased expression was observed for 49 genes, with two downregulated genes observed. Our results indicate that diabetic and prediabetic conditions change the expression levels of genes related to endothelial cells biology and, consequently, may increase the risk for occurrence of endothelial dysfunction.
Collapse
|
19
|
Ortiz A, Husi H, Gonzalez-Lafuente L, Valiño-Rivas L, Fresno M, Sanz AB, Mullen W, Albalat A, Mezzano S, Vlahou T, Mischak H, Sanchez-Niño MD. Mitogen-Activated Protein Kinase 14 Promotes AKI. J Am Soc Nephrol 2016; 28:823-836. [PMID: 27620989 DOI: 10.1681/asn.2015080898] [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: 08/15/2015] [Accepted: 07/28/2016] [Indexed: 01/20/2023] Open
Abstract
An improved understanding of pathogenic pathways in AKI may identify novel therapeutic approaches. Previously, we conducted unbiased liquid chromatography-tandem mass spectrometry-based protein expression profiling of the renal proteome in mice with acute folate nephropathy. Here, analysis of the dataset identified enrichment of pathways involving NFκB in the kidney cortex, and a targeted data mining approach identified components of the noncanonical NFκB pathway, including the upstream kinase mitogen-activated protein kinase kinase kinase 14 (MAP3K14), the NFκB DNA binding heterodimer RelB/NFκB2, and proteins involved in NFκB2 p100 ubiquitination and proteasomal processing to p52, as upregulated. Immunohistochemistry localized MAP3K14 expression to tubular cells in acute folate nephropathy and human AKI. In vivo, kidney expression levels of NFκB2 p100 and p52 increased rapidly after folic acid injection, as did DNA binding of RelB and NFκB2, detected in nuclei isolated from the kidneys. Compared with wild-type mice, MAP3K14 activity-deficient aly/aly (MAP3K14aly/aly) mice had less kidney dysfunction, inflammation, and apoptosis in acute folate nephropathy and less kidney dysfunction and a lower mortality rate in cisplatin-induced AKI. The exchange of bone marrow between wild-type and MAP3K14aly/aly mice did not affect the survival rate of either group after folic acid injection. In cultured tubular cells, MAP3K14 small interfering RNA targeting decreased inflammation and cell death. Additionally, cell culture and in vivo studies identified the chemokines MCP-1, RANTES, and CXCL10 as MAP3K14 targets in tubular cells. In conclusion, MAP3K14 promotes kidney injury through promotion of inflammation and cell death and is a promising novel therapeutic target.
Collapse
Affiliation(s)
- Alberto Ortiz
- Instituto Investigacion Sanitaria-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain; .,Red de Investigacion Rena, Madrid, Spain
| | - Holger Husi
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Laura Gonzalez-Lafuente
- Instituto Investigacion Sanitaria-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain.,Red de Investigacion Rena, Madrid, Spain
| | - Lara Valiño-Rivas
- Instituto Investigacion Sanitaria-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain.,Red de Investigacion Rena, Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas de la Universidad Autonoma de Madrid, Madrid, Spain
| | - Ana Belen Sanz
- Instituto Investigacion Sanitaria-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain.,Mosaiques diagnostics GmbH, Hannover, Germany
| | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Amaya Albalat
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sergio Mezzano
- Unidad de Nefrología, Instituto de Medicina, Universidad Austral de Chile, Valdivia, Chile; and
| | - Tonia Vlahou
- Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Harald Mischak
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom.,Mosaiques diagnostics GmbH, Hannover, Germany
| | - Maria Dolores Sanchez-Niño
- Instituto Investigacion Sanitaria-Fundacion Jimenez Diaz-Universidad Autonoma de Madrid and Fundacion Renal Iñigo Alvarez de Toledo-Instituto Reina Sofia de Investigacion Nefrologica, Madrid, Spain; .,Red de Investigacion Rena, Madrid, Spain
| |
Collapse
|
20
|
Le TKC, Hosaka T, Nguyen TT, Kassu A, Dang TO, Tran HB, Pham TP, Tran QB, Le THH, Pham XD. Bifidobacterium species lower serum glucose, increase expressions of insulin signaling proteins, and improve adipokine profile in diabetic mice. Biomed Res 2015; 36:63-70. [PMID: 25749152 DOI: 10.2220/biomedres.36.63] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This study, using C57BL/6J mice with streptozotocin (STZ)-induced diabetes, aimed to determine whether Bifidobacterium species (spp.) both induces the expressions of proteins in the insulin signaling pathway and enhances the expressions of certain adipocytokines. The protein expressions of IκB kinase alpha (IKKα), IκB kinase beta (IKKβ), nuclear factor-kappaB inhibitor alpha (IκBα), and the mitogen-activated protein kinase (MAPK) pathway were also investigated. Oral administration of Bifidobacterium spp. reduced blood glucose levels significantly and increased the protein expressions of insulin receptor beta, insulin receptor substrate 1, protein kinase B (Akt/PKB), IKKα, and IκBα. Extracellular-signal-regulated kinase 2 (ERK2) showed increased expression. Bifidobacterium spp. also induced the adiponectin expression and decreased both macrophage chemoattractant protein-1 (MCP-1) and interleukin-6 (IL-6) expression. In addition, IKKβ, c-Jun NH2-terminal kinase (JNK) and p38 MAP kinase expressions showed no significant changes in both groups. In conclusion, Bifidobacterium spp. may be the promising bacteria for treating diabetes.
Collapse
Affiliation(s)
- Thi Kim Chung Le
- Department of Food Microbiology, National Institute for Food Control
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Shi L, Feng L, Jiang WD, Liu Y, Jiang J, Wu P, Zhao J, Kuang SY, Tang L, Tang WN, Zhang YA, Zhou XQ. Folic acid deficiency impairs the gill health status associated with the NF-κB, MLCK and Nrf2 signaling pathways in the gills of young grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2015; 47:289-301. [PMID: 26381932 DOI: 10.1016/j.fsi.2015.09.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/11/2015] [Accepted: 09/11/2015] [Indexed: 06/05/2023]
Abstract
The aim of this study was to investigate the effect of dietary folic acid on fish growth, the immune and barrier functions of fish gills, and the potential mechanisms of these effects. Young grass carp (Ctenopharyngodon idella) were fed diets containing graded levels of folic acid at 0.10 (basal diet), 0.47, 1.03, 1.48, 1.88 and 3.12 mg kg(-1) diet for 8 weeks. The results showed that acid phosphatase and lysozyme activities and the complement component 3 content in fish gills decreased with folic acid deficiency (P < 0.05). Folic acid deficiency up-regulated liver-expressed antimicrobial peptide 1, interleukin 1β, interleukin 8, tumor necrosis factor α, nuclear factor κB p65, IκB kinase α (IKK-α), IKK-β and IKK-γ gene expression. Folic acid deficiency down-regulated interleukin 10, transforming growth factor β, IκB and target of rapamycin gene expression in fish gills (P < 0.05). These results showed that limited folic acid decreased fish gill immune status. Furthermore, folic acid deficiency down-regulated claudin-b, claudin-c, claudin-3, occludin and zonula occludens 1 gene expression, whereas folic acid deficiency up-regulated claudin-12, claudin-15, myosin light chain kinase and p38 mitogen activated protein kinase gene expression in fish gills (P < 0.05). These results suggested that folic acid deficiency disrupted tight junction-mediated fish gill barrier function. Additionally, folic acid deficiency increased the content of reactive oxygen species, protein carbonyl and malondialdehyde (MDA); Mn superoxide dismutase activity and gene expression; and Kelch-like-ECH-associated protein 1a (Keap1a) and Keap1b gene expression (P < 0.05). Conversely, folic acid deficiency decreased Cu/Zn superoxide dismutase, catalase, glutathione peroxidase, glutathione s-transferases and glutathione reductase activities and gene expression as well as NF-E2-related factor 2 gene expression in fish gills (P < 0.05). All of these results indicated that folic acid deficiency impaired fish gill health status via regulating gene expression of cytokines, tight junction proteins, antioxidant enzymes, NF-κB p65, MLCK and Nrf2. Based on percent weight gain, LZ activity and MDA content in the gills, the dietary folic acid requirements for young grass carp were 1.60, 2.07 and 2.08 mg kg(-1), respectively.
Collapse
Affiliation(s)
- Lei Shi
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Juan Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Wu-Neng Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
22
|
Serum amyloid A and inflammation in diabetic kidney disease and podocytes. J Transl Med 2015; 95:250-62. [PMID: 25531567 PMCID: PMC4346621 DOI: 10.1038/labinvest.2014.163] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/04/2014] [Accepted: 11/24/2014] [Indexed: 12/27/2022] Open
Abstract
Inflammatory pathways are central mechanisms in diabetic kidney disease (DKD). Serum amyloid A (SAA) is increased by chronic inflammation, but SAA has not been previously evaluated as a potential DKD mediator. The aims of this study were to determine whether SAA is increased in human DKD and corresponding mouse models and to assess effects of SAA on podocyte inflammatory responses. SAA was increased in the plasma of people with DKD characterized by overt proteinuria and inversely correlated with estimated glomerular filtration rate (creatinine-based CKD-EPI). SAA was also elevated in plasma of diabetic mouse models including type 1 diabetes (streptozotocin/C57BL/6) and type 2 diabetes (BTBR-ob/ob). SAA mRNA (Nephromine) was increased in human DKD compared with non-diabetic and/or glomerular disease controls (glomerular fold change 1.5, P=0.017; tubulointerstitium fold change 1.4, P=0.021). The kidneys of both diabetic mouse models also demonstrated increased SAA mRNA (quantitative real-time PCR) expression compared with non-diabetic controls (type 1 diabetes fold change 2.9; type 2 diabetes fold change 42.5, P=0.009; interaction by model P=0.57). Humans with DKD and the diabetic mouse models exhibited extensive SAA protein deposition in the glomeruli and tubulointerstitium in similar patterns by immunohistochemistry. SAA localized within podocytes of diabetic mice. Podocytes exposed to advanced glycation end products, metabolic mediators of inflammation in diabetes, increased expression of SAA mRNA (fold change 15.3, P=0.004) and protein (fold change 38.4, P=0.014). Podocytes exposed to exogenous SAA increased NF-κB activity, and pathway array analysis revealed upregulation of mRNA for NF-κB-dependent targets comprising numerous inflammatory mediators, including SAA itself (fold change 17.0, P=0.006). Inhibition of NF-κB reduced these pro-inflammatory responses. In conclusion, SAA is increased in the blood and produced in the kidneys of people with DKD and corresponding diabetic mouse models. Podocytes are likely to be key responder cells to SAA-induced inflammation in the diabetic kidney. SAA is a compelling candidate for DKD therapeutic and biomarker discovery.
Collapse
|
23
|
Wang Y, Wang B, Du F, Su X, Sun G, Zhou G, Bian X, Liu N. Epigallocatechin-3-Gallate Attenuates Oxidative Stress and Inflammation in Obstructive Nephropathy via NF-κB and Nrf2/HO-1 Signalling Pathway Regulation. Basic Clin Pharmacol Toxicol 2015; 117:164-72. [PMID: 25625183 DOI: 10.1111/bcpt.12383] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/09/2015] [Indexed: 01/22/2023]
Abstract
Oxidative stress and inflammation contribute importantly to the pathogenesis of chronic kidney disease (CKD). Epigallocatechin-3-gallate (EGCG), which is the most abundant and most active catechin polyphenol extracted from green tea, has been proved to have many bioactivities. In this study, the renoprotective effect of EGCG was evaluated in a widely used kidney disease model, the unilateral ureteral obstruction (UUO) mice model. After 14 days of EGCG administration, mean arterial blood pressure, body-weight and obstructed kidney weight were measured. Levels of blood urea nitrogen (BUN) and creatinine (CR) and activities of glutamic-pyruvic transaminase (GPT) and lactate dehydrogenase (LDH) in serum were estimated as indicators of renal function. Periodic acid-Schiff (PAS) staining was performed to observe the pathological changes of the obstructed kidney. Antioxidant enzymes and pro-inflammatory cytokine production were estimated to reflect the oxidative stress and inflammatory state in the obstructed kidney. Finally, the main proteins in the NF-κB and Nrf2 signalling pathway and DNA binding activity of NF-κB and Nrf2 were measured to investigate the effect of EGCG on these two pathways. The results demonstrated that EGCG could restore UUO-induced kidney weight loss and renal dysfunction. In addition, UUO-induced oxidative stress and inflammatory responses in the obstructed kidney were also prevented by EGCG. Furthermore, EGCG could induce both NF-κB and Nrf2 nuclear translocation in the UUO kidney and promote heme oxygenase-1 (HO-1) production. These results indicated that the renoprotective effect of EGCG might be through its NF-κB and Nrf2 signalling pathway regulations.
Collapse
Affiliation(s)
- Yanqiu Wang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bowen Wang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Feng Du
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuesong Su
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guangping Sun
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guangyu Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohui Bian
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Na Liu
- Department of Nephrology, Ordos Central Hospital, Ordos, Inner Mongolia, China
| |
Collapse
|
24
|
Neto WK, Gama EF, Rocha LY, Ramos CC, Taets W, Scapini KB, Ferreira JB, Rodrigues B, Caperuto É. Effects of testosterone on lean mass gain in elderly men: systematic review with meta-analysis of controlled and randomized studies. AGE (DORDRECHT, NETHERLANDS) 2015; 37:9742. [PMID: 25637335 PMCID: PMC4312307 DOI: 10.1007/s11357-014-9742-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 12/18/2014] [Indexed: 05/26/2023]
Abstract
The objective of this study was to evaluate the effects of steroid anabolic androgenic hormones use on lean mass gain in elderly men through a systematic review with a meta-analysis of randomized controlled studies. We systematically searched PubMed database until 4th October 2013. We included randomized placebo-controlled trials (RCT) that studied testosterone replacement therapy in men over 60 years of age, with total testosterone levels ≤550 ng/dl, observing gains in weight, lean mass tissue and fat mass as outcome. We excluded duplicated studies, studies which mixed men and women, and studies using weak androgens such as dehydroepiandrosterone or androstenedione. The initial search yielded 2681 articles, of which 26 were selected for full text analysis. In the end, 11 studies were included. However, 3 studies were not included in the meta-analysis. Meta-analysis showed that mean weight increased (lean mass), ranging from 1.65 (95 % CI, 1.61-1.69) to 6.20 (95 % CI, 5.22-7.18) kg, although it was heterogeneous (I (2) = 98 %). Effect estimate was 3.59 [2.38-4.81]. Androgen therapy decreased fat mass; effect estimate was -1.78 [-2.57, -0.99] that analysis had also a high level of heterogeneity (I (2) = 81 %). The results suggest that testosterone replacement therapy is able to increase muscle mass in elderly men and that is affected by the time that the treatment is carried out and the method of administration of the drug.
Collapse
Affiliation(s)
- Walter Krause Neto
- />Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, SP Brazil
| | - Eliane Florencio Gama
- />Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, SP Brazil
| | - Leandro Yanase Rocha
- />Laboratory of Human Movement, Department of Physical Education, São Judas Tadeu University, São Paulo, SP Brazil
| | - Carla Cristina Ramos
- />Laboratory of Human Movement, Department of Physical Education, São Judas Tadeu University, São Paulo, SP Brazil
| | - Wagner Taets
- />Laboratory of Human Movement, Department of Physical Education, São Judas Tadeu University, São Paulo, SP Brazil
| | | | | | - Bruno Rodrigues
- />Laboratory of Human Movement, Department of Physical Education, São Judas Tadeu University, São Paulo, SP Brazil
| | - Érico Caperuto
- />Laboratory of Human Movement, Department of Physical Education, São Judas Tadeu University, São Paulo, SP Brazil
- />Laboratory of Exercise and Movement Sciences, Mackenzie Presbiterian University, 546, Taquari St, Moóca, 03166-000 São Paulo, SP Brazil
| |
Collapse
|
25
|
Thomas CM, Yong QC, Rosa RM, Seqqat R, Gopal S, Casarini DE, Jones WK, Gupta S, Baker KM, Kumar R. Cardiac-specific suppression of NF-κB signaling prevents diabetic cardiomyopathy via inhibition of the renin-angiotensin system. Am J Physiol Heart Circ Physiol 2014; 307:H1036-45. [PMID: 25085967 DOI: 10.1152/ajpheart.00340.2014] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activation of NF-κB signaling in the heart may be protective or deleterious depending on the pathological context. In diabetes, the role of NF-κB in cardiac dysfunction has been investigated using pharmacological approaches that have a limitation of being nonspecific. Furthermore, the specific cellular pathways by which NF-κB modulates heart function in diabetes have not been identified. To address these questions, we used a transgenic mouse line expressing mutated IκB-α in the heart (3M mice), which prevented activation of canonical NF-κB signaling. Diabetes was developed by streptozotocin injections in wild-type (WT) and 3M mice. Diabetic WT mice developed systolic and diastolic cardiac dysfunction by the 12th week, as measured by echocardiography. In contrast, cardiac function was preserved in 3M mice up to 24 wk of diabetes. Diabetes induced an elevation in cardiac oxidative stress in diabetic WT mice but not 3M mice compared with nondiabetic control mice. In diabetic WT mice, an increase in the phospholamban/sarco(endo)plasmic reticulum Ca(2+)-ATPase 2 ratio and decrease in ryanodine receptor expression were observed, whereas diabetic 3M mice showed an opposite effect on these parameters of Ca(2+) handling. Significantly, renin-angiotensin system activity was suppressed in diabetic 3M mice compared with an increase in WT animals. In conclusion, these results demonstrate that inhibition of NF-κB signaling in the heart prevents diabetes-induced cardiac dysfunction through preserved Ca(2+) handling and inhibition of the cardiac renin-angiotensin system.
Collapse
Affiliation(s)
- Candice M Thomas
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Qian Chen Yong
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Rodolfo M Rosa
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil; and
| | - Rachid Seqqat
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Shanthi Gopal
- Central Texas Veterans Health Care System, Temple, Texas
| | - Dulce E Casarini
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil; and
| | - W Keith Jones
- Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, Illinois
| | - Sudhiranjan Gupta
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Kenneth M Baker
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Rajesh Kumar
- Division of Molecular Cardiology, Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Baylor Scott & White Health, Temple, Texas; Central Texas Veterans Health Care System, Temple, Texas;
| |
Collapse
|
26
|
Liu R, Zhong Y, Li X, Chen H, Jim B, Zhou MM, Chuang PY, He JC. Role of transcription factor acetylation in diabetic kidney disease. Diabetes 2014; 63:2440-53. [PMID: 24608443 PMCID: PMC4066331 DOI: 10.2337/db13-1810] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nuclear factor (NF)-κB and signal transducer and activator of transcription 3 (STAT3) play a critical role in diabetic nephropathy (DN). Sirtuin-1 (SIRT1) regulates transcriptional activation of target genes through protein deacetylation. Here, we determined the roles of Sirt1 and the effect of NF-κB (p65) and STAT3 acetylation in DN. We found that acetylation of p65 and STAT3 was increased in both mouse and human diabetic kidneys. In human podocytes, advanced glycation end products (AGEs) induced p65 and STAT3 acetylation and overexpression of acetylation-incompetent mutants of p65 and STAT3 abrogated AGE-induced expression of NF-κB and STAT3 target genes. Inhibition of AGE formation in db/db mice by pyridoxamine treatment attenuated proteinuria and podocyte injury, restored SIRT1 expression, and reduced p65 and STAT3 acetylation. Diabetic db/db mice with conditional deletion of SIRT1 in podocytes developed more proteinuria, kidney injury, and acetylation of p65 and STAT3 compared with db/db mice without SIRT1 deletion. Treatment of db/db mice with a bromodomain and extraterminal (BET)-specific bromodomain inhibitor (MS417) which blocks acetylation-mediated association of p65 and STAT3 with BET proteins, attenuated proteinuria, and kidney injury. Our findings strongly support a critical role for p65 and STAT3 acetylation in DN. Targeting protein acetylation could be a potential new therapy for DN.
Collapse
Affiliation(s)
- Ruijie Liu
- Department of Medicine/Nephrology, Mount Sinai School of Medicine, New York, NY
| | - Yifei Zhong
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuezhu Li
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haibing Chen
- Department of Endocrinology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Belinda Jim
- Division of Nephrology, Jacobi Medical Center, Bronx, NY
| | - Ming-Ming Zhou
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine, New York, NY
| | - Peter Y Chuang
- Department of Medicine/Nephrology, Mount Sinai School of Medicine, New York, NY
| | - John Cijiang He
- Department of Medicine/Nephrology, Mount Sinai School of Medicine, New York, NYRenal Section, James J. Peters VA Medical Center, Bronx, NY
| |
Collapse
|
27
|
Lim AI, Chan LYY, Tang SCW, Yiu WH, Li R, Lai KN, Leung JCK. BMP-7 represses albumin-induced chemokine synthesis in kidney tubular epithelial cells through destabilization of NF-κB-inducing kinase. Immunol Cell Biol 2014; 92:427-35. [PMID: 24418819 DOI: 10.1038/icb.2013.106] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/28/2013] [Accepted: 12/12/2013] [Indexed: 01/28/2023]
Abstract
Protein overload activates proximal tubule epithelial cells (PTECs) to release chemokines. Bone morphogenetic protein-7 (BMP-7) reduces infiltrating cells and tissue damage in acute and chronic renal injuries. The present study examines the inhibitory effect and related molecular mechanism of BMP-7 on chemokine and adhesion molecule synthesis by PTECs activated with human serum albumin (HSA). The expression profiles of chemokines and adhesion molecules in cultured human PTECs were screened by PCR array. Expression of CXCL1, CXCL2 and vascular cell adhesion protein 1 (VCAM-1) by PTECs was significantly upregulated by HSA and reduced by BMP-7. HSA activated both the canonical and noncanonical nuclear factor (NF)-κB pathways in PTECs, as indicated by the increased nuclear translocation of NF-κB p50 and p52 subunits. The nuclear translocation of NF-κB p52 was completely abrogated by BMP-7, whereas NF-κB p50 activation was only partially repressed. BMP-7 increased the expression of cellular inhibitor of apoptosis 1 (cIAP1), tumor necrosis factor receptor-associated factor (TRAF)2 and TRAF3, but not of NF-κB-inducing kinase (NIK) that was significantly upregulated by HSA. Silencing NIK recapitulated the partial inhibitory effect on HSA-induced chemokine synthesis by BMP-7. Complete abolishment of the chemokine synthesis was only achieved by including additional blockade of the NF-κB p65 translocation on top of NIK silencing. Our data suggest that BMP-7 represses the NIK-dependent chemokine synthesis in PTECs activated with HSA through blocking the noncanonical NF-κB pathway and partially interfering with the canonical NF-κB pathway.
Collapse
Affiliation(s)
- Ai Ing Lim
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong
| | - Loretta Y Y Chan
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong
| | - Sydney C W Tang
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong
| | - Wai Han Yiu
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong
| | - Ruixi Li
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong
| | - Kar Neng Lai
- Nephrology Center, Hong Kong Sanatorium and Hospital, Happy Valley, Hong Kong
| | - Joseph C K Leung
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
28
|
Poveda J, Tabara LC, Fernandez-Fernandez B, Martin-Cleary C, Sanz AB, Selgas R, Ortiz A, Sanchez-Niño MD. TWEAK/Fn14 and Non-Canonical NF-kappaB Signaling in Kidney Disease. Front Immunol 2013; 4:447. [PMID: 24339827 PMCID: PMC3857575 DOI: 10.3389/fimmu.2013.00447] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/26/2013] [Indexed: 12/27/2022] Open
Abstract
The incidence of acute kidney injury (AKI) and chronic kidney disease (CKD) is increasing. However, there is no effective therapy for AKI and current approaches only slow down, but do not prevent progression of CKD. TWEAK is a TNF superfamily cytokine. A solid base of preclinical data suggests a role of therapies targeting the TWEAK or its receptor Fn14 in AKI and CKD. In particular TWEAK/Fn14 targeting may preserve renal function and decrease cell death, inflammation, proteinuria, and fibrosis in mouse animal models. Furthermore there is clinical evidence for a role of TWEAK in human kidney injury including increased tissue and/or urinary levels of TWEAK and parenchymal renal cell expression of the receptor Fn14. In this regard, clinical trials of TWEAK targeting are ongoing in lupus nephritis. Nuclear factor-kappa B (NF-κB) activation plays a key role in TWEAK-elicited inflammatory responses. Activation of the non-canonical NF-κB pathway is a critical difference between TWEAK and TNF. TWEAK activation of the non-canonical NF-κB pathways promotes inflammatory responses in tubular cells. However, there is an incomplete understanding of the role of non-canonical NF-κB activation in kidney disease and on its contribution to TWEAK actions in vivo.
Collapse
Affiliation(s)
- Jonay Poveda
- Department of Nephrology, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid and IRSIN , Madrid , Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Shahsavar F, Jafarzadeh M, Azargoon A, Hedayati M, Asadifar B. Small ubiquitin-like modifier 4 M55V polymorphism is not associated with diabetic nephropathy in Iranian type 2 diabetes patients. INDIAN JOURNAL OF HUMAN GENETICS 2013; 19:179-82. [PMID: 24019619 PMCID: PMC3758724 DOI: 10.4103/0971-6866.116121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
INTRODUCTION: We studied the impact of small ubiquitin-like modifier 4 (SUMO4) M55V polymorphism on susceptibility to diabetic nephropathy in Iranian type 2 diabetes patients. MATERIALS AND METHODS: The patient group consisted of 50 Iranian type 2 diabetes patients with nephropathy, and the control group consisted of 50 Iranian type 2 diabetes patients without nephropathy. Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism method for the M55V. RESULTS: The frequency of SUMO4 AA, AG, and GG genotypes were 23%, 18%, and 9% in the patient group and 10%, 22%, and 18% in the control group. There was no significant difference in frequency of SUMO4 genotypes in patients compared to controls. CONCLUSION: These findings indicate that SUMO4 M55V polymorphism is not associated with diabetic nephropathy in Iranian type 2 diabetes patients.
Collapse
Affiliation(s)
- Farhad Shahsavar
- Department of Immunology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | | | | | | |
Collapse
|
30
|
Abstract
A strong case for the deregulation of epigenetic chromatin modifications in the development and progression of various chronic complications of diabetes has emerged from recent experimental observations. Clinical trials of type 1 and type 2 diabetes patients highlight the importance of early and intensive treatment and the prolonged damage of hyperglycemia on organs such as the kidney. The functional relationship between the regulation of chromatin architecture and persistent gene expression changes conferred by prior hyperglycemia represents an important avenue of investigation for explaining diabetic nephropathy. While several studies implicate epigenetic changes at the chromatin template in the deregulated gene expression associated with diabetic nephropathy, the molecular determinants of metabolic memory in renal cells remain poorly understood. There is now strong evidence from experimental animals and cell culture of persistent glucose-driven changes in vascular endothelial gene expression that may also have relevance for the microvasculature of the kidney. Exploration of epigenetic mechanisms underlying the hyperglycemic cue mediating persistent transcriptional changes in renal cells holds novel therapeutic potential for diabetic nephropathy.
Collapse
Affiliation(s)
- Samuel T Keating
- Epigenetics in Human Health and Disease Laboratory, Baker IDI Heart and Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, 3004, Australia
| | | |
Collapse
|
31
|
Choudhary S, Kalita M, Fang L, Patel KV, Tian B, Zhao Y, Edeh CB, Brasier AR. Inducible tumor necrosis factor (TNF) receptor-associated factor-1 expression couples the canonical to the non-canonical NF-κB pathway in TNF stimulation. J Biol Chem 2013; 288:14612-14623. [PMID: 23543740 PMCID: PMC3656313 DOI: 10.1074/jbc.m113.464081] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 03/28/2013] [Indexed: 11/06/2022] Open
Abstract
The NF-κB transcription factor mediates the inflammatory response through distinct (canonical and non-canonical) signaling pathways. The mechanisms controlling utilization of either of these pathways are largely unknown. Here we observe that TNF stimulation induces delayed NF-κB2/p100 processing and investigate the coupling mechanism. TNF stimulation induces TNF-associated factor-1 (TRAF-1) that directly binds NF-κB-inducing kinase (NIK) and stabilizes it from degradation by disrupting its interaction with TRAF2·cIAP2 ubiquitin ligase complex. We show that TRAF1 depletion prevents TNF-induced NIK stabilization and reduces p52 production. To further examine the interactions of TRAF1 and NIK with NF-κB2/p100 processing, we mathematically modeled TRAF1·NIK as a coupling signaling complex and validated computational inference by siRNA knockdown to show non-canonical pathway activation is dependent not only on TRAF1 induction but also NIK stabilization by forming TRAF1·NIK complex. Thus, these integrated computational-experimental studies of TNF-induced TRAF1 expression identified TRAF1·NIK as a central complex linking canonical and non-canonical pathways by disrupting the TRAF2-cIAP2 ubiquitin ligase complex. This feed-forward kinase pathway is essential for the activation of non-canonical pathway.
Collapse
Affiliation(s)
- Sanjeev Choudhary
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas 77555; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas 77555.
| | - Mridul Kalita
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Ling Fang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Kershaw V Patel
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas 77555; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas 77555
| | - Chukwudi B Edeh
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas 77555; Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas 77555
| |
Collapse
|
32
|
Day RT, Cavaglieri RC, Feliers D. Apelin retards the progression of diabetic nephropathy. Am J Physiol Renal Physiol 2013; 304:F788-800. [PMID: 23303408 PMCID: PMC3602700 DOI: 10.1152/ajprenal.00306.2012] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 01/04/2013] [Indexed: 12/14/2022] Open
Abstract
Apelin and its receptor APJ have pleiotropic effects in mice and humans and play a protective role in cardiovascular diseases at least partially by inhibiting oxidative stress. Our objective was to study the effect of apelin on the progression of kidney disease in mice with established type 1 diabetes. Ove26 mice with type 1 diabetes received daily subcutaneous injections of apelin for 2 or 14 wk. APJ localizes in the glomeruli and blood vessels of kidneys. Renal APJ expression was reduced in diabetic mice but increased after treatment with apelin. Apelin treatment did not affect glycemia, body weight, or blood pressure in diabetic mice. Whole kidney and glomerular hypertrophy, as well as renal inflammation, including monocyte chemoattractant protein 1 and vascular cell adhesion molecule 1 expression, NF-κB activation, and monocyte infiltration, was inhibited after short and long treatment with apelin. Apelin administration significantly reduced albuminuria at 6 mo. Short treatment with apelin was sufficient to reverse the downregulation of the antioxidant enzyme catalase. Expression of angiotensin II and angiotensin type 1 receptor (AT1) in kidneys from diabetic mice treated was not affected by apelin. These findings show for the first time that apelin exerts a protective effect on the diabetic kidney. Short administration is sufficient to reduce kidney and glomerular hypertrophy as well as renal inflammation, but prolonged treatment is required to improve albuminuria. This effect was independent of the activation of the renin angiotensin system but correlated with upregulation of the antioxidant catalase. Apelin may represent a novel tool to treat diabetic nephropathy.
Collapse
Affiliation(s)
- Robert T Day
- Department of Medicine/Renal Diseases, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | | | | |
Collapse
|
33
|
Huang WC, Hung MC. Beyond NF-κB activation: nuclear functions of IκB kinase α. J Biomed Sci 2013; 20:3. [PMID: 23343355 PMCID: PMC3563485 DOI: 10.1186/1423-0127-20-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/11/2013] [Indexed: 12/15/2022] Open
Abstract
IκB kinase (IKK) complex, the master kinase for NF-κB activation, contains two kinase subunits, IKKα and IKKβ. In addition to mediating NF-κB signaling by phosphorylating IκB proteins during inflammatory and immune responses, the activation of the IKK complex also responds to various stimuli to regulate diverse functions independently of NF-κB. Although these two kinases share structural and biochemical similarities, different sub-cellular localization and phosphorylation targets between IKKα and IKKβ account for their distinct physiological and pathological roles. While IKKβ is predominantly cytoplasmic, IKKα has been found to shuttle between the cytoplasm and the nucleus. The nuclear-specific roles of IKKα have brought increasing complexity to its biological function. This review highlights major advances in the studies of the nuclear functions of IKKα and the mechanisms of IKKα nuclear translocation. Understanding the nuclear activity is essential for targeting IKKα for therapeutics.
Collapse
Affiliation(s)
- Wei-Chien Huang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 40447, Taiwan.
| | | |
Collapse
|
34
|
Kulp GA, Tilton RG, Herndon DN, Jeschke MG. Hyperglycemia exacerbates burn-induced liver inflammation via noncanonical nuclear factor-κB pathway activation. Mol Med 2012; 18:948-56. [PMID: 22572938 PMCID: PMC3459487 DOI: 10.2119/molmed.2011.00357] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 05/03/2012] [Indexed: 01/04/2023] Open
Abstract
Hyperglycemia and inflammation are hallmarks of burn injury. In this study, we used a rat model of hyperglycemia and burn injury to investigate the effects of hyperglycemia on inflammatory responses in the liver. Hyperglycemia was induced in male Sprague-Dawley rats with streptozotocin (STZ) (35-40 mg/kg), followed by a 60% third-degree scald burn injury. Cytokine levels (by multiplex, in cytosolic liver extracts), hormones (by enzyme-linked immunosorbent assay [ELISA], in serum), nuclear factor (NF)-κB protein deoxyribonucleic acid (DNA) binding (by ELISA, in nuclear liver extracts) and liver functional panel (using VetScan, in serum) were measured at different time points up to 7 d after burn injury. Blood glucose significantly increased after burn injury in both groups with different temporal patterns. Hyperglycemic rats were capable of endogenous insulin secretion, which was enhanced significantly versus controls 12 h after burn injury. DNA binding data of liver nuclear extracts showed a robust and significant activation of the noncanonical NF-κB pathway in the hyperglycemic versus control burn animals, including increased NF-κB-inducing kinase expression (p < 0.05). Liver acute-phase proteins and cytokine expression were increased, whereas secretion of constitutive proteins was decreased after burn injury in hyperglycemic versus control animals (p < 0.05). These results indicate that burn injury to the skin rapidly activated canonical and noncanonical NF-κB pathways in the liver. Robust activation of the NF-κB noncanonical pathway was associated with increased expression of inflammatory markers and acute-phase proteins, and impaired glucose metabolism. Hyperglycemia is detrimental to burn outcome by augmenting inflammation mediated by hepatic noncanonical NF-κB pathway activation.
Collapse
Affiliation(s)
- Gabriela A Kulp
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Biochemistry and Molecular Biology Graduate Program, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ronald G Tilton
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - David N Herndon
- Shriners Hospital for Children and Department of Surgery, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Science Centre, Sunnybrook Research Institute, Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Martínez-García C, Izquierdo A, Velagapudi V, Vivas Y, Velasco I, Campbell M, Burling K, Cava F, Ros M, Orešič M, Vidal-Puig A, Medina-Gomez G. Accelerated renal disease is associated with the development of metabolic syndrome in a glucolipotoxic mouse model. Dis Model Mech 2012; 5:636-48. [PMID: 22773754 PMCID: PMC3424461 DOI: 10.1242/dmm.009266] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 06/10/2012] [Indexed: 12/23/2022] Open
Abstract
Individuals with metabolic syndrome are at high risk of developing chronic kidney disease (CKD) through unclear pathogenic mechanisms. Obesity and diabetes are known to induce glucolipotoxic effects in metabolically relevant organs. However, the pathogenic role of glucolipotoxicity in the aetiology of diabetic nephropathy is debated. We generated a murine model, the POKO mouse, obtained by crossing the peroxisome proliferator-activated receptor gamma 2 (PPARγ2) knockout (KO) mouse into a genetically obese ob/ob background. We have previously shown that the POKO mice showed: hyperphagia, insulin resistance, hyperglycaemia and dyslipidaemia as early as 4 weeks of age, and developed a complete loss of normal β-cell function by 16 weeks of age. Metabolic phenotyping of the POKO model has led to investigation of the structural and functional changes in the kidney and changes in blood pressure in these mice. Here we demonstrate that the POKO mouse is a model of renal disease that is accelerated by high levels of glucose and lipid accumulation. Similar to ob/ob mice, at 4 weeks of age these animals exhibited an increased urinary albumin:creatinine ratio and significantly increased blood pressure, but in contrast showed a significant increase in the renal hypertrophy index and an associated increase in p27(Kip1) expression compared with their obese littermates. Moreover, at 4 weeks of age POKO mice showed insulin resistance, an alteration of lipid metabolism and glomeruli damage associated with increased transforming growth factor beta (TGFβ) and parathyroid hormone-related protein (PTHrP) expression. At this age, levels of proinflammatory molecules, such as monocyte chemoattractant protein-1 (MCP-1), and fibrotic factors were also increased at the glomerular level compared with levels in ob/ob mice. At 12 weeks of age, renal damage was fully established. These data suggest an accelerated lesion through glucolipotoxic effects in the renal pathogenesis in POKO mice.
Collapse
Affiliation(s)
- Cristina Martínez-García
- Universidad Rey Juan Carlos, Dpto. de Bioquímica, Fisiología y Genética Molecular, Avda. de Atenas s/n. 28922, Alcorcón, Madrid, Spain
| | - Adriana Izquierdo
- Universidad Rey Juan Carlos, Dpto. de Bioquímica, Fisiología y Genética Molecular, Avda. de Atenas s/n. 28922, Alcorcón, Madrid, Spain
| | - Vidya Velagapudi
- VTT Technical Research Centre of Finland, Tietotie 2, Espoo, PO Box 1500, FIN-02044 VTT, Finland
| | - Yurena Vivas
- Universidad Rey Juan Carlos, Dpto. de Bioquímica, Fisiología y Genética Molecular, Avda. de Atenas s/n. 28922, Alcorcón, Madrid, Spain
| | - Ismael Velasco
- Universidad Rey Juan Carlos, Dpto. de Bioquímica, Fisiología y Genética Molecular, Avda. de Atenas s/n. 28922, Alcorcón, Madrid, Spain
| | - Mark Campbell
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Level 4, Box 289, Addenbrookes Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Keith Burling
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Level 4, Box 289, Addenbrookes Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Fernando Cava
- Área de Laboratorio – Hospital Universitario Fundación Alcorcón, C/Budapest 1. 28922, Alcorcón, Madrid, Spain
| | - Manuel Ros
- Universidad Rey Juan Carlos, Dpto. de Bioquímica, Fisiología y Genética Molecular, Avda. de Atenas s/n. 28922, Alcorcón, Madrid, Spain
| | - Matej Orešič
- VTT Technical Research Centre of Finland, Tietotie 2, Espoo, PO Box 1500, FIN-02044 VTT, Finland
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Level 4, Box 289, Addenbrookes Hospital, Hills Road, Cambridge, CB2 OQQ, UK
| | - Gema Medina-Gomez
- Universidad Rey Juan Carlos, Dpto. de Bioquímica, Fisiología y Genética Molecular, Avda. de Atenas s/n. 28922, Alcorcón, Madrid, Spain
| |
Collapse
|
36
|
Combined aliskiren and amlodipine reduce albuminuria via reduction in renal inflammation in diabetic rats. J Cardiovasc Pharmacol 2012; 59:281-7. [PMID: 22075749 DOI: 10.1097/fjc.0b013e31823fc3f5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We hypothesized that compared with hydrochlorothiazide (HCTZ), the renin inhibitor aliskiren (ALISK) or amlodipine (AMLO) and their combination reduce albuminuria via reduction in renal inflammation, independent of blood pressure (BP) changes. We studied normal and streptozotocin-induced diabetic (DM) Sprague-Dawley rats treated for 6 weeks with vehicle, ALISK, HCTZ, or AMLO individually and combined and evaluated the effects of treatments on BP, urine albumin to creatinine ratio, renal interstitial fluid levels of angiotensin II, tumor necrosis factor alpha (TNF-α), and interleukin 6 (IL-6) and renal expression of TNF-α, IL-6, transforming growth factor beta 1, and nuclear factor kappa B. There were no differences in BP between treatments. Only ALISK and its combinations reduced renal interstitial fluid angiotensin II. Urine albumin to creatinine ratio increased in DM rats and decreased with ALISK alone or combined with HCTZ or AMLO. HCTZ or AMLO individually and combined did not influence urine albumin to creatinine ratio. Renal interstitial fluid TNF-α and IL-6, and the renal expression of TNF-α, IL-6, transforming growth factor beta 1, and nuclear factor kappa B were increased in DM rats. These renal inflammatory markers were reduced only with ALISK or AMLO individually or combined with other treatments. We conclude that ALISK alone and combined with HCTZ or AMLO reduced albuminuria in diabetes via reduction in renal inflammation, independent of BP changes.
Collapse
|
37
|
Oh SW, Ahn JM, Lee YM, Kim S, Chin HJ, Chae DW, Na KY. Activation of hypoxia-inducible factor by cobalt is associated with the attenuation of tissue injury and apoptosis in cyclosporine-induced nephropathy. TOHOKU J EXP MED 2012; 226:197-206. [PMID: 22343435 DOI: 10.1620/tjem.226.197] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hypoxia-inducible factor (HIF) is a transcription factor that regulates cellular hypoxic responses. Despite the therapeutic benefits of cyclosporine A (CsA) in organ transplantation, its clinical use is limited due to chronic nephropathy. We investigated whether HIF activation by cobalt could improve CsA-induced nephropathy, and investigated the related mechanism. In animal experiments, rats were kept on a 0.05% low-salt diet and administered CsA subcutaneously for 28 days (15 mg/kg/day). They also received cobalt (10 mg/kg/day) during the entire experimental period. The administration of cobalt significantly increased HIF-1α expression in the kidney. The increased expression of HIF-1α ameliorated CsA-induced afferent arteriolopathy and tubulointerstitial injury in the kidney. Cobalt significantly reduced the infiltration of macrophages/monocytes into the renal tubulointerstitium. In addition, HIF activation by cobalt reduced the number of CsA-induced apoptotic cells in the kidney. Subsequently, HK-2 human renal tubular epithelial cells were used for in vitro experiments. They were pre-treated with 150 µM of cobalt to activate HIF, and then exposed to 10 µM CsA. HIF activation by cobalt decreased the CsA-induced apoptosis in HK-2 cells, as judged by the decreases in the number of apoptotic cells, pro-apoptotic caspase-3 activity, and the expression level of cleaved caspase-3, together with the increase in the expression of anti-apoptotic bcl-2. Cobalt pretreatment also reduced the CsA-induced phosphorylation of NF-κB and the CsA-induced expression of vimentin and α-smooth muscle actin, suggesting the attenuation of inflammation and fibrosis. In conclusion, the activation of HIF by cobalt may ameliorate the CsA-induced nephropathy by inhibiting apoptosis, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Se Won Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | | | | | | | | | | | | |
Collapse
|
38
|
Choudhary S, Sinha S, Zhao Y, Banerjee S, Sathyanarayana P, Shahani S, Sherman V, Tilton RG, Bajaj M. NF-kappaB-inducing kinase (NIK) mediates skeletal muscle insulin resistance: blockade by adiponectin. Endocrinology 2011; 152:3622-7. [PMID: 21846802 PMCID: PMC3176647 DOI: 10.1210/en.2011-1343] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 07/25/2011] [Indexed: 11/19/2022]
Abstract
Enhanced levels of nuclear factor (NF)-κB-inducing kinase (NIK), an upstream kinase in the NF-κB pathway, have been implicated in the pathogenesis of chronic inflammation in diabetes. We investigated whether increased levels of NIK could induce skeletal muscle insulin resistance. Six obese subjects with metabolic syndrome underwent skeletal muscle biopsies before and six months after gastric bypass surgery to quantitate NIK protein levels. L6 skeletal myotubes, transfected with NIK wild-type or NIK kinase-dead dominant negative plasmids, were treated with insulin alone or with adiponectin and insulin. Effects of NIK overexpression on insulin-stimulated glucose uptake were estimated using tritiated 2-deoxyglucose uptake. NF-κB activation (EMSA), phosphatidylinositol 3 (PI3) kinase activity, and phosphorylation of inhibitor κB kinase β and serine-threonine kinase (Akt) were measured. After weight loss, skeletal muscle NIK protein was significantly reduced in association with increased plasma adiponectin and enhanced AMP kinase phosphorylation and insulin sensitivity in obese subjects. Enhanced NIK expression in cultured L6 myotubes induced a dose-dependent decrease in insulin-stimulated glucose uptake. The decrease in insulin-stimulated glucose uptake was associated with a significant decrease in PI3 kinase activity and protein kinase B/Akt phosphorylation. Overexpression of NIK kinase-dead dominant negative did not affect insulin-stimulated glucose uptake. Adiponectin treatment inhibited NIK-induced NF-κB activation and restored insulin sensitivity by restoring PI3 kinase activation and subsequent Akt phosphorylation. These results indicate that NIK induces insulin resistance and further indicate that adiponectin exerts its insulin-sensitizing effect by suppressing NIK-induced skeletal muscle inflammation. These observations suggest that NIK could be an important therapeutic target for the treatment of insulin resistance associated with inflammation in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Sanjeev Choudhary
- Department of Internal Medicine, Division of Endocrinology, Sealy Center of Molecular Medicine, 8.138 Medical Research Building, The University of Texas Medical Branch, 301 University Boulevard, Galveston, Texas 77555-1060, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
OBJECTIVE Diabetic kidney disease (DKD) is the single leading cause of kidney failure in the U.S., for which a cure has not yet been found. The aim of our study was to provide an unbiased catalog of gene-expression changes in human diabetic kidney biopsy samples. RESEARCH DESIGN AND METHODS Affymetrix expression arrays were used to identify differentially regulated transcripts in 44 microdissected human kidney samples. DKD samples were significant for their racial diversity and decreased glomerular filtration rate (~25-35 mL/min). Stringent statistical analysis, using the Benjamini-Hochberg corrected two-tailed t test, was used to identify differentially expressed transcripts in control and diseased glomeruli and tubuli. Two different web-based algorithms were used to define differentially regulated pathways. RESULTS We identified 1,700 differentially expressed probesets in DKD glomeruli and 1,831 in diabetic tubuli, and 330 probesets were commonly differentially expressed in both compartments. Pathway analysis highlighted the regulation of Ras homolog gene family member A, Cdc42, integrin, integrin-linked kinase, and vascular endothelial growth factor signaling in DKD glomeruli. The tubulointerstitial compartment showed strong enrichment for inflammation-related pathways. The canonical complement signaling pathway was determined to be statistically differentially regulated in both DKD glomeruli and tubuli and was associated with increased glomerulosclerosis even in a different set of DKD samples. CONCLUSIONS Our studies have cataloged gene-expression regulation and identified multiple novel genes and pathways that may play a role in the pathogenesis of DKD or could serve as biomarkers.
Collapse
Affiliation(s)
- Karolina I. Woroniecka
- Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, Bronx, New York
| | - Ae Seo Deok Park
- Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, Bronx, New York
| | - Davoud Mohtat
- Department of Pediatrics, Division of Nephrology, Albert Einstein College of Medicine, Bronx, New York
| | | | - James M. Pullman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Katalin Susztak
- Department of Medicine, Division of Nephrology, Albert Einstein College of Medicine, Bronx, New York
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York
- Corresponding author: Katalin Susztak,
| |
Collapse
|
40
|
Zhao Y, Banerjee S, LeJeune WS, Choudhary S, Tilton RG. NF-κB-inducing kinase increases renal tubule epithelial inflammation associated with diabetes. EXPERIMENTAL DIABETES RESEARCH 2011; 2011:192564. [PMID: 21869881 PMCID: PMC3159020 DOI: 10.1155/2011/192564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 06/22/2011] [Indexed: 02/07/2023]
Abstract
The impact of increased NF-κB-inducing kinase (NIK), a key component of the NF-κB activation pathways, on diabetes-induced renal inflammation remains unknown. We overexpressed NIK wild type (NIKwt) or kinase-dead dominant negative mutants (NIKdn) in HK-2 cells and demonstrated that RelB and p52, but not RelA, abundance and DNA binding increased in nuclei of NIKwt but not NIKdn overexpressed cells, and this corresponded with increases in multiple proinflammatory cytokines. Since TRAF3 negatively regulates NIK expression, we silenced TRAF3 by >50%; this increased nuclear levels of p52 and RelB, and transcript levels of proinflammatory cytokines and transcription factors. In HK-2 cells and mouse primary proximal tubule epithelial cells treated with methylglyoxal-modified albumin, multiple proinflammatory cytokines and NIK were increased in association with increased nuclear RelB and p52. These observations indicate that NIK regulates proinflammatory responses of renal proximal tubular epithelial cells via mechanisms involving TRAF3 and suggest a role for NF-κB noncanonical pathway activation in modulating diabetes-induced inflammation in renal tubular epithelium.
Collapse
Affiliation(s)
- Yanhua Zhao
- Division of Endocrinology and Stark Diabetes Center, Department of Internal Medicine, The University of Texas Medical Branch, 301 University Baulevard, Galveston, TX 77555-1060, USA
| | - Srijita Banerjee
- Division of Endocrinology and Stark Diabetes Center, Department of Internal Medicine, The University of Texas Medical Branch, 301 University Baulevard, Galveston, TX 77555-1060, USA
| | - Wanda S. LeJeune
- Division of Endocrinology and Stark Diabetes Center, Department of Internal Medicine, The University of Texas Medical Branch, 301 University Baulevard, Galveston, TX 77555-1060, USA
| | - Sanjeev Choudhary
- Division of Endocrinology and Stark Diabetes Center, Department of Internal Medicine, The University of Texas Medical Branch, 301 University Baulevard, Galveston, TX 77555-1060, USA
- Sealy Center for Molecular Medicine, The University of Texas Medical Branch, Galveston, TX 77555-1060, USA
| | - Ronald G. Tilton
- Division of Endocrinology and Stark Diabetes Center, Department of Internal Medicine, The University of Texas Medical Branch, 301 University Baulevard, Galveston, TX 77555-1060, USA
- Sealy Center for Molecular Medicine, The University of Texas Medical Branch, Galveston, TX 77555-1060, USA
- Department of Ophthalmology & Visual Sciences, The University of Texas Medical Branch, Galveston, TX 77555-1060, USA
| |
Collapse
|
41
|
Zhao Y, Banerjee S, Dey N, LeJeune WS, Sarkar PS, Brobey R, Rosenblatt KP, Tilton RG, Choudhary S. Klotho depletion contributes to increased inflammation in kidney of the db/db mouse model of diabetes via RelA (serine)536 phosphorylation. Diabetes 2011; 60:1907-16. [PMID: 21593200 PMCID: PMC3121423 DOI: 10.2337/db10-1262] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Klotho is an antiaging hormone present in the kidney that extends the lifespan, regulates kidney function, and modulates cellular responses to oxidative stress. We investigated whether Klotho levels and signaling modulate inflammation in diabetic kidneys. RESEARCH DESIGN AND METHODS Renal Klotho expression was determined by quantitative real-time PCR and immunoblot analysis. Primary mouse tubular epithelial cells were treated with methylglyoxalated albumin, and Klotho expression and inflammatory cytokines were measured. Nuclear factor (NF)-κB activation was assessed by treating human embryonic kidney (HEK) 293 and HK-2 cells with tumor necrosis factor (TNF)-α in the presence or absence of Klotho, followed by immunoblot analysis to evaluate inhibitor of κB (IκB)α degradation, IκB kinase (IKK) and p38 activation, RelA nuclear translocation, and phosphorylation. A chromatin immunoprecipitation assay was performed to analyze the effects of Klotho signaling on interleukin-8 and monocyte chemoattractant protein-1 promoter recruitment of RelA and RelA serine (Ser)(536). RESULTS Renal Klotho mRNA and protein were significantly decreased in db/db mice, and a similar decline was observed in the primary cultures of mouse tubule epithelial cells treated with methylglyoxal-modified albumin. The exogenous addition of soluble Klotho or overexpression of membranous Klotho in tissue culture suppressed NF-κB activation and subsequent production of inflammatory cytokines in response to TNF-α stimulation. Klotho specifically inhibited RelA Ser(536) phosphorylation as well as promoter DNA binding of this phosphorylated form of RelA without affecting IKK-mediated IκBα degradation, total RelA nuclear translocation, and total RelA DNA binding. CONCLUSIONS These findings suggest that Klotho serves as an anti-inflammatory modulator, negatively regulating the production of NF-κB-linked inflammatory proteins via a mechanism that involves phosphorylation of Ser(536) in the transactivation domain of RelA.
Collapse
Affiliation(s)
- Yanhua Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Srijita Banerjee
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Nilay Dey
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Wanda S. LeJeune
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Partha S. Sarkar
- Department of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| | - Reynolds Brobey
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas
| | - Kevin P. Rosenblatt
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas
| | - Ronald G. Tilton
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Sanjeev Choudhary
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas
- Corresponding author: Sanjeev Choudhary,
| |
Collapse
|
42
|
Chen HY, Huang XR, Wang W, Li JH, Heuchel RL, Chung AC, Lan HY. The protective role of Smad7 in diabetic kidney disease: mechanism and therapeutic potential. Diabetes 2011; 60:590-601. [PMID: 20980457 PMCID: PMC3028360 DOI: 10.2337/db10-0403] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Although Smad3 has been considered as a downstream mediator of transforming growth factor-β (TGF-β) signaling in diabetes complications, the role of Smad7 in diabetes remains largely unclear. The current study tests the hypothesis that Smad7 may play a protective role and has therapeutic potential for diabetic kidney disease. RESEARCH DESIGN AND METHODS Protective role of Smad7 in diabetic kidney disease was examined in streptozotocin-induced diabetic mice that have Smad7 gene knockout (KO) and in diabetic rats given Smad7 gene transfer using an ultrasound-microbubble-mediated technique. RESULTS We found that mice deficient for Smad7 developed more severe diabetic kidney injury than wild-type mice as evidenced by a significant increase in microalbuminuria, renal fibrosis (collagen I, IV, and fibronectin), and renal inflammation (interleukin-1β [IL-1β], tumor necrosis factor-α [TNF-α], monocyte chemoattractant protein-1 [MCP-1], intracellular adhesion molecule-1 [ICAM-1], and macrophages). Further studies revealed that enhanced renal fibrosis and inflammation in Smad7 KO mice with diabetes were associated with increased activation of both TGF-β/Smad2/3 and nuclear factor-κB (NF-κB) signaling pathways. To develop a therapeutic potential for diabetic kidney disease, Smad7 gene was transferred into the kidney in diabetic rats by an ultrasound-microbubble-mediated technique. Although overexpression of renal Smad7 had no effect on levels of blood glucose, it significantly attenuated the development of microalbuminuria, TGF-β/Smad3-mediated renal fibrosis such as collagen I and IV and fibronectin accumulation and NF-κB/p65-driven renal inflammation including IL-1β, TNF-α, MCP-1, and ICAM-1 expression and macrophage infiltration in diabetic rats. CONCLUSIONS Smad7 plays a protective role in diabetic renal injury. Overexpression of Smad7 may represent a novel therapy for the diabetic kidney complication.
Collapse
Affiliation(s)
- Hai Yong Chen
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao R. Huang
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wansheng Wang
- Department of Pediatrics, Texas Tech University, Health Science Center at El Paso, El Paso, Texas
| | - Jin Hua Li
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | | | - Arthur C.K. Chung
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Yao Lan
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Corresponding author: Hui Y. Lan,
| |
Collapse
|
43
|
Fallah S, Jafarzadeh M, Hedayati M. No association of the SUMO4 polymorphism M55V variant in type 2 diabetes in Iranian subjects. Diabetes Res Clin Pract 2010; 90:191-5. [PMID: 20728233 DOI: 10.1016/j.diabres.2010.05.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Revised: 05/02/2010] [Accepted: 05/06/2010] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Diabetes mellitus incidence has an increasing rate and it's genetic aspect is an important approach as a risk factor and predictive value in this disorder. In some population, SUMO4, a regulator of NF-κB, gene polymorphism is associated with diabetes. A single-nucleotide polymorphism was detected in SUMO4; substituting a highly conserved methionine with a valine residue (M55V). We studied the association between M55V polymorphism in the SUMO4 gene insusceptibility of type 2 diabetes in patients with type 2 diabetes. MATERIALS AND METHODS Participants were 50 patients with type 2 diabetes and 50 control Iranian subjects. Genotyping was done using polymorphism chain reaction (PCR) technique and subsequent cleavage by restriction endonuclease (RFLP) for the M55V SUMO4 gene variant. RESULTS The frequency of SUMO4 AA, AG and GG were 13%, 25% and 12% in control group and 20%, 22%, 18% in the type 2 diabetes patients respectively. The SUMO4 M55V variant was not associated with the susceptibility of type 2 diabetes. CONCLUSION The study indicates that the SUMO4 gene M55V variant was not associated with the susceptibility of the type 2 diabetes polymorphism.
Collapse
Affiliation(s)
- Soudabeh Fallah
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | | | | |
Collapse
|
44
|
Chen YW, Chenier I, Chang SY, Tran S, Ingelfinger JR, Zhang SL. High glucose promotes nascent nephron apoptosis via NF-kappaB and p53 pathways. Am J Physiol Renal Physiol 2010; 300:F147-56. [PMID: 20962117 DOI: 10.1152/ajprenal.00361.2010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A hyperglycemic environment in utero reduces kidney size and nephron number due to nascent nephron apoptosis. However, the underlying mechanisms are incompletely understood. The present study investigated whether the nascent nephron apoptosis promoted by high glucose is mediated via the transcription factor NF-κB and p53 signaling pathways. Neonatal mouse kidneys from the offspring of nondiabetic, diabetic, and insulin-treated diabetic dams were used for in vivo studies, and MK4 cells, an embryonic metanephric mesenchymal (MM) cell line, were used for in vitro studies. Neonatal kidneys of the offspring of diabetic mothers exhibited an increased number of apoptotic cells and reactive oxygen species (ROS) generation, enhanced NF-κB activation, and nuclear translocation of its subunits (p50 and p65 subunits) as well as phosphorylation (Ser 15) of p53 compared with kidneys of offspring of nondiabetic mothers. Insulin treatment of diabetic dams normalized these parameters in the offspring. In vitro, high-glucose (25 mM) induced ROS generation and significantly increased MK4 cell apoptosis and caspase-3 activity via activation of NF-κB pathway, with p53 phosphorylation and nuclear translocation compared with normal glucose (5 mM). These changes in a high-glucose milieu were prevented by transient transfection of small interfering RNAs for dominant negative IκBα or IKK or p53. Our data demonstrate that high glucose-induced nascent nephron apoptosis is mediated, at least in part, via ROS generation and the activation of NF-κB and p53 pathways.
Collapse
Affiliation(s)
- Yun-Wen Chen
- Université de Montréal, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, CRCHUM, Hôpital Hôtel-Dieu, Pavillon Masson, 3850 Saint-Urbain St., Montreal, Quebec, Canada H2W 1T7
| | | | | | | | | | | |
Collapse
|
45
|
Starkey JM, Zhao Y, Sadygov RG, Haidacher SJ, LeJeune WS, Dey N, Luxon BA, Kane MA, Napoli JL, Denner L, Tilton RG. Altered retinoic acid metabolism in diabetic mouse kidney identified by O isotopic labeling and 2D mass spectrometry. PLoS One 2010; 5:e11095. [PMID: 20559430 PMCID: PMC2885420 DOI: 10.1371/journal.pone.0011095] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 05/14/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Numerous metabolic pathways have been implicated in diabetes-induced renal injury, yet few studies have utilized unbiased systems biology approaches for mapping the interconnectivity of diabetes-dysregulated proteins that are involved. We utilized a global, quantitative, differential proteomic approach to identify a novel retinoic acid hub in renal cortical protein networks dysregulated by type 2 diabetes. METHODOLOGY/PRINCIPAL FINDINGS Total proteins were extracted from renal cortex of control and db/db mice at 20 weeks of age (after 12 weeks of hyperglycemia in the diabetic mice). Following trypsinization, (18)O- and (16)O-labeled control and diabetic peptides, respectively, were pooled and separated by two dimensional liquid chromatography (strong cation exchange creating 60 fractions further separated by nano-HPLC), followed by peptide identification and quantification using mass spectrometry. Proteomic analysis identified 53 proteins with fold change >or=1.5 and p<or=0.05 after Benjamini-Hochberg adjustment (out of 1,806 proteins identified), including alcohol dehydrogenase (ADH) and retinaldehyde dehydrogenase (RALDH1/ALDH1A1). Ingenuity Pathway Analysis identified altered retinoic acid as a key signaling hub that was altered in the diabetic renal cortical proteome. Western blotting and real-time PCR confirmed diabetes-induced upregulation of RALDH1, which was localized by immunofluorescence predominantly to the proximal tubule in the diabetic renal cortex, while PCR confirmed the downregulation of ADH identified with mass spectrometry. Despite increased renal cortical tissue levels of retinol and RALDH1 in db/db versus control mice, all-trans-retinoic acid was significantly decreased in association with a significant decrease in PPARbeta/delta mRNA. CONCLUSIONS/SIGNIFICANCE Our results indicate that retinoic acid metabolism is significantly dysregulated in diabetic kidneys, and suggest that a shift in all-trans-retinoic acid metabolism is a novel feature in type 2 diabetic renal disease. Our observations provide novel insights into potential links between altered lipid metabolism and other gene networks controlled by retinoic acid in the diabetic kidney, and demonstrate the utility of using systems biology to gain new insights into diabetic nephropathy.
Collapse
Affiliation(s)
- Jonathan M. Starkey
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Stark Diabetes Center, University of Texas Medical Branch, Galveston, Texas, United States of America
- McCoy Diabetes Mass Spectrometry Research Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Rovshan G. Sadygov
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Sigmund J. Haidacher
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- McCoy Diabetes Mass Spectrometry Research Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Wanda S. LeJeune
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- McCoy Diabetes Mass Spectrometry Research Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nilay Dey
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bruce A. Luxon
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Translational Science Biomedical Informatics Program, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Maureen A. Kane
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California, United States of America
| | - Joseph L. Napoli
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California, United States of America
| | - Larry Denner
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Stark Diabetes Center, University of Texas Medical Branch, Galveston, Texas, United States of America
- McCoy Diabetes Mass Spectrometry Research Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ronald G. Tilton
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- Stark Diabetes Center, University of Texas Medical Branch, Galveston, Texas, United States of America
- McCoy Diabetes Mass Spectrometry Research Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
46
|
Brasier AR. The nuclear factor-kappaB-interleukin-6 signalling pathway mediating vascular inflammation. Cardiovasc Res 2010; 86:211-8. [PMID: 20202975 DOI: 10.1093/cvr/cvq076] [Citation(s) in RCA: 406] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vascular inflammation is a common pathophysiological response to diverse cardiovascular disease processes, including atherosclerosis, myocardial infarction, congestive heart failure, and aortic aneurysms/dissection. Inflammation is an ordered process initiated by vascular injury that produces enhanced leucocyte adherence, chemotaxis, and finally activation in situ. This process is coordinated by local secretion of adhesion molecules, chemotactic factors, and cytokines whose expression is the result of vascular injury-induced signal transduction networks. A wide variety of mediators of the vascular injury response have been identified; these factors include vasoactive peptides (angiotensin II, Ang II), CD40 ligands, oxidized cholesterol, and advanced glycation end-products. Downstream, the nuclear factor-kappaB (NF-kappaB) transcription factor performs an important signal integration step, responding to mediators of vascular injury in a stimulus-dependent and cell type-specific manner. The ultimate consequence of NF-kappaB signalling is the activation of inflammatory genes including adhesion molecules and chemotaxins. However, clinically, the hallmark of vascular NF-kappaB activation is the production of interleukin-6 (IL-6), whose local role in vascular inflammation is relatively unknown. The recent elucidation for the role of the IL-6 signalling pathway in Ang II-induced vascular inflammation as one that controls monocyte activation as well as its diverse signalling mechanism will be reviewed. These new discoveries further our understanding for the important role of the NF-kappaB-IL-6 signalling pathway in the process of vascular inflammation.
Collapse
Affiliation(s)
- Allan R Brasier
- Division of Endocrinology, Department of Internal Medicine, Sealy Center for Molecular Medicine and Institute for Translational Sciences, University of Texas Medical Branch, MRB 8.122, 301 University Blvd, Galveston, TX 77555-1060, USA.
| |
Collapse
|
47
|
Sanz AB, Sanchez-Niño MD, Izquierdo MC, Jakubowski A, Justo P, Blanco-Colio LM, Ruiz-Ortega M, Selgas R, Egido J, Ortiz A. TWEAK activates the non-canonical NFkappaB pathway in murine renal tubular cells: modulation of CCL21. PLoS One 2010; 5:e8955. [PMID: 20126461 PMCID: PMC2813291 DOI: 10.1371/journal.pone.0008955] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 01/04/2010] [Indexed: 12/21/2022] Open
Abstract
TWEAK is a member of the TNF superfamily of cytokines that contribute to kidney tubulointerstitial injury. It has previously been reported that TWEAK induces transient nuclear translocation of RelA and expression of RelA-dependent cytokines in renal tubular cells. Additionally, TWEAK induced long-lasting NFκB activation suggestive of engagement of the non-canonical NFκB pathway. We now explore TWEAK-induced activation of NFκB2 and RelB, as well as expression of CCL21, a T-cell chemotactic factor, in cultured murine tubular epithelial cells and in healthy kidneys in vivo. In cultured tubular cells, TWEAK and TNFα activated different DNA-binding NFκB complexes. TWEAK-induced sustained NFκB activation was associated with NFκB2 p100 processing to p52 via proteasome and nuclear translocation and DNA-binding of p52 and RelB. TWEAK, but not TNFα used as control), induced a delayed increase in CCL21a mRNA (3.5±1.22-fold over control) and CCL21 protein (2.5±0.8-fold over control), which was prevented by inhibition of the proteasome, or siRNA targeting of NIK or RelB, but not by RelA inhibition with parthenolide. A second NFκB2-dependent chemokine, CCL19, was upregulates by TWEAK, but not by TNFα. However, both cytokines promoted chemokine RANTES expression (3-fold mRNA at 24 h). In vivo, TWEAK induced nuclear NFκB2 and RelB translocation and CCL21a mRNA (1.5±0.3-fold over control) and CCL21 protein (1.6±0.5-fold over control) expression in normal kidney. Increased tubular nuclear RelB and tubular CCL21 expression in acute kidney injury were decreased by neutralization (2±0.9 vs 1.3±0.6-fold over healthy control) or deficiency of TWEAK (2±0.9 vs 0.8±0.6-fold over healthy control). Moreover, anti-TWEAK treatment prevented the recruitment of T cells to the kidney in this model (4.1±1.4 vs 1.8±1-fold over healthy control). Our results thus identify TWEAK as a regulator of non-canonical NFκB activation and CCL21 expression in tubular cells thus promoting lymphocyte recruitment to the kidney during acute injury.
Collapse
Affiliation(s)
- Ana B. Sanz
- Servicio de Nefrologia, Fundación para la Investigación Biomédica del Hospital Universitario La Paz, Madrid, Spain
| | - Maria D. Sanchez-Niño
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | - Maria C. Izquierdo
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | - Aniela Jakubowski
- Department of Immunobiology, Biogen Idec, Inc., Cambridge, Massachusetts, United States of America
| | - Pilar Justo
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | - Luis M. Blanco-Colio
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | - Marta Ruiz-Ortega
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | - Rafael Selgas
- Servicio de Nefrologia, Fundación para la Investigación Biomédica del Hospital Universitario La Paz, Madrid, Spain
| | - Jesús Egido
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
| | - Alberto Ortiz
- Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo, Madrid, Spain
- * E-mail:
| |
Collapse
|
48
|
Li X, Hu J, Zhang R, Sun X, Zhang Q, Guan X, Chen J, Zhu Q, Li S. Urocortin ameliorates diabetic nephropathy in obese db/db mice. Br J Pharmacol 2008; 154:1025-34. [PMID: 18587447 DOI: 10.1038/bjp.2008.155] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Hyperglycaemia induces overproduction of mitochondrial reactive oxygen species (ROS) in endothelial cells, which is believed to be a major molecular mechanism underlying complications of diabetes, including diabetic nephropathy. Impairment of endothelium-dependent vasodilatation is found in type 2 diabetes. Urocortin is a 40 amino-acid peptide related to the corticotrophin-releasing factor (CRF) family, which suppresses production of ROS in endothelial cells and sustains endothelium-dependent relaxations of rat coronary artery. However, it is not clear if urocortin has any effect on diabetic nephropathy. EXPERIMENTAL APPROACH Possible mechanisms underlying the effects of urocortin on diabetic nephropathy were investigated in db/db mice and cultured rat mesangial cells. KEY RESULTS Urocortin decreased body weight, plasma levels of advanced glycation end-products, blood urea nitrogen and creatinine levels. However, food intake, plasma insulin and glucose levels remained unaffected. Superoxide dismutase activity was increased markedly, whereas malonaldehyde levels in kidney homogenate and sorbitol concentrations in red blood cells were decreased significantly in urocortin-treated mice. Urocortin significantly decreased glomerular extracellular matrix expansion and accumulation in kidney. Moreover, urocortin inhibited the overexpression of transforming growth factor-beta 1 and connective tissue growth factor in rat mesangial cells induced by 25 mM glucose. All the effects of urocortin, except sorbitol accumulation, were abolished by the non-selective CRF receptor blocker, astressin. CONCLUSION AND IMPLICATIONS Urocortin could significantly ameliorate diabetic nephropathy and this effect was mediated via the CRF receptor.
Collapse
Affiliation(s)
- X Li
- Department of Pharmacology, Nanjing Medical University, Nanjing, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tran S, Chen YW, Chenier I, Chan JSD, Quaggin S, Hébert MJ, Ingelfinger JR, Zhang SL. Maternal diabetes modulates renal morphogenesis in offspring. J Am Soc Nephrol 2008; 19:943-52. [PMID: 18305124 DOI: 10.1681/asn.2007080864] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Maternal diabetes leads to an adverse in utero environment, but whether maternal diabetes impairs nephrogenesis is unknown. Diabetes was induced with streptozotocin in pregnant Hoxb7-green fluorescence protein mice at embryonic day 13, and the offspring were examined at several time points after birth. Compared with offspring of nondiabetic controls, offspring of diabetic mice had lower body weight, body size, kidney weight, and nephron number. The observed renal dysmorphogenesis may be the result of increased apoptosis, because immunohistochemical analysis revealed significantly more apoptotic podocytes as well as increased active caspase-3 immunostaining in the renal tubules compared with control mice. Regarding potential mediators of these differences, offspring of diabetic mice had increased expression of intrarenal angiotensinogen and renin mRNA, upregulation of NF-kappaB isoforms p50 and p65, and activation of the NF-kappaB pathway. In conclusion, maternal diabetes impairs nephrogenesis, possibly via enhanced intrarenal activation of the renin-angiotensin system and NF-kappaB signaling.
Collapse
Affiliation(s)
- Stella Tran
- University of Montreal, Centre Hospitalier de l'Université de Montréal-Hôtel-Dieu, Research Centre, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Bibliography. Current world literature. Diabetes and the endocrine pancreas II. Curr Opin Endocrinol Diabetes Obes 2007; 14:329-57. [PMID: 17940461 DOI: 10.1097/med.0b013e3282c3a898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|