1
|
Annunziata G, Caprio M, Verde L, Carella AM, Camajani E, Benvenuto A, Paolini B, De Nicola L, Aucella F, Bellizzi V, Barberi S, Grassi D, Fogacci F, Colao A, Cicero AFG, Prodam F, Aimaretti G, Muscogiuri G, Barrea L. Nutritional assessment and medical dietary therapy for management of obesity in patients with non-dialysis chronic kidney disease: a practical guide for endocrinologist, nutritionists and nephrologists. A consensus statement from the Italian society of endocrinology (SIE), working group of the club nutrition-hormones and metabolism; the Italian society of nutraceuticals (SINut), club ketodiets and nutraceuticals "KetoNut-SINut"; and the Italian society of nephrology (SIN). J Endocrinol Invest 2024; 47:2889-2913. [PMID: 39292364 DOI: 10.1007/s40618-024-02446-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE Chronic kidney disease (CKD) is a serious health concern with an estimated prevalence of about 13.4% worldwide. It is cause and consequence of various comorbidities, including cardiovascular diseases. In parallel, common pathological conditions closely related to ageing and unhealthy dietary habits increase the risk of CKD development and progression, including type 2 diabetes and obesity. Among these, obesity is either independent risk factor for new onset kidney disease or accelerates the rate of decline of kidney function by multiple mechanisms. Therefore, the role of diets aimed at attaining weight loss in patients with obesity is clearly essential to prevent CKD as to slow disease progression. Various dietary approaches have been licensed for the medical dietary therapy in CKD, including low-protein diet and Mediterranean diet. Interestingly, emerging evidence also support the use of low-carbohydrate/ketogenic diet (LCD/KD) in these patients. More specifically, LCD/KDs may efficiently promote weight loss, improve metabolic parameters, and reduce inflammation and oxidative stress, resulting in a dietary strategy that act globally in managing collateral conditions that are directly and indirectly related to the kidney function. CONCLUSION This consensus statement from the Italian Society of Endocrinology (SIE), working group of the Club Nutrition - Hormones and Metabolism; the Italian Society of Nutraceuticals (SINut), Club Ketodiets and Nutraceuticals "KetoNut-SINut"; and the Italian Society of Nephrology (SIN) is intended to be a guide for Endocrinologist, Nutritionists and Nephrologist who deal with the management of patients with obesity with non-dialysis CKD providing a practical guidance on assessing nutritional status and prescribing the optimal diet in order to best manage obesity to prevent CKD and its progression to dialysis.
Collapse
Affiliation(s)
- G Annunziata
- Facoltà di Scienze Umane, della Formazione e dello Sport, Università Telematica Pegaso, Via Porzio, Centro Direzionale, Isola F2, 80143, Naples, Italy
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - M Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele, Rome, Italy
- Department for the Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166, Rome, Italy
| | - L Verde
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - A M Carella
- Facoltà di Scienze Umane, della Formazione e dello Sport, Università Telematica Pegaso, Via Porzio, Centro Direzionale, Isola F2, 80143, Naples, Italy
- Internal Medicine Department, "T. Masselli-Mascia" Hospital-San Severo (Foggia), Foggia, Italy
| | - E Camajani
- Department for the Promotion of Human Sciences and Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166, Rome, Italy
| | - A Benvenuto
- Internal Medicine Department, "T. Masselli-Mascia" Hospital-San Severo (Foggia), Foggia, Italy
| | - B Paolini
- Department of Innovation, experimentation and clinical research, Unit of dietetics and clinical nutrition, S. Maria Alle Scotte Hospital, University of Siena, Siena, SI, Italy
| | - L De Nicola
- Nephrology and Dialysis Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - F Aucella
- Nephrology and Dialysis Unit, "Casa Sollievo Della Sofferenza" Foundation, Scientific Institut for Reserch and Health Care, San Giovanni Rotondo, FG, Italy
| | - V Bellizzi
- Nephrology and Dialysis Division, AORN "Sant'Anna E San Sebastiano" Hospital, Caserta, Italy
| | - S Barberi
- Department of Clinical and Molecular Medicine, Renal Unit, Sant'Andrea University Hospital, "Sapienza" University of Rome, Rome, Italy
| | - D Grassi
- Internal Medicine Unit-Val Vibrata Hospital-Sant'Omero (TE)-Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - F Fogacci
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100, Bologna, Italy
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
| | - A Colao
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Centro Italiano per la Cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università Degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco "Educazione Alla Salute e Allo Sviluppo Sostenibile", University Federico II, 80131, Naples, Italy
| | - A F G Cicero
- Hypertension and Cardiovascular Risk Factors Research Centre, Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40100, Bologna, Italy
- Cardiovascular Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, 40138, Bologna, Italy
| | - F Prodam
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | - G Aimaretti
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - G Muscogiuri
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Centro Italiano per la Cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università Degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Cattedra Unesco "Educazione Alla Salute e Allo Sviluppo Sostenibile", University Federico II, 80131, Naples, Italy.
| | - L Barrea
- Centro Italiano per la Cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università Degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Benessere, Nutrizione e Sport, Università Telematica Pegaso, Centro Direzionale, Via Porzio, Isola F2, 80143, Naples, Italy
| |
Collapse
|
2
|
Han Z, Gong L, Xue Y, Wang R, Liu J, Wang X, Zhao W, Liao H, Li R. Effects of Inonotus obliquus on ameliorating podocyte injury in ORG mice through TNF pathway and prediction of active compounds. Front Pharmacol 2024; 15:1426917. [PMID: 39234117 PMCID: PMC11371614 DOI: 10.3389/fphar.2024.1426917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024] Open
Abstract
Background Podocyte injury is a common pathologic mechanism in diabetic kidney disease (DKD) and obesity-related glomerulopathy (ORG). Our previous study confirmed that Inonotus obliquus (IO) improved podocyte injury on DKD rats. The current study explored the pharmacological effects, related mechanisms and possible active components of IO on ORG mice. Methods Firstly, by combining ultra-high performance liquid chromatography tandem mass spectrometry analysis (UPLC-Q-TOF-MS) with network pharmacology to construct the human protein-protein interaction mechanism and enrich the pathway, which led to discover the crucial mechanism of IO against ORG. Then, ORG mice were established by high-fat diet and biochemical assays, histopathology, and Western blot were used to explore the effects of IO on obesity and podocyte injury. Finally, network pharmacology-based findings were confirmed by immunohistochemistry. The compositions of IO absorbed in mice plasma were analyzed by UPLC-Q-TOF-MS and molecular docking was used to predict the possible active compounds. Results The network pharmacology result suggested that IO alleviated the inflammatory response of ORG by modulating TNF signal. The 20-week in vivo experiment confirmed that IO improved glomerular hypertrophy, podocyte injury under electron microscopy, renal nephrin, synaptopodin, TNF-α and IL-6 expressions with Western blotting and immunohistochemical staining. Other indicators of ORG such as body weight, kidney weight, serum total cholesterol, liver triglyceride also improved by IO intervention. The components analysis showed that triterpenoids, including inoterpene F and trametenolic acid, might be the pharmacodynamic basis. Conclusion The research based on UPLC-Q-TOF-MS analysis, network pharmacology and in vivo experiment suggested that the amelioration of IO on podocyte injury in ORG mice via its modulation on TNF signal. Triterpenoids were predicated as acting components.
Collapse
Affiliation(s)
- Zhaodi Han
- Drug Clinical Trial Institution, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Le Gong
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Yani Xue
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Rui Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Jing Liu
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Xinyu Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Wenyan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Hui Liao
- Drug Clinical Trial Institution, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Rongshan Li
- Department of Nephrology, Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
3
|
Ali MM, Parveen S, Williams V, Dons R, Uwaifo GI. Cardiometabolic comorbidities and complications of obesity and chronic kidney disease (CKD). J Clin Transl Endocrinol 2024; 36:100341. [PMID: 38616864 PMCID: PMC11015524 DOI: 10.1016/j.jcte.2024.100341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024] Open
Abstract
Obesity and chronic kidney disease are two ongoing progressive clinical pandemics of major public health and clinical care significance. Because of their growing prevalence, chronic indolent course and consequent complications both these conditions place significant burden on the health care delivery system especially in developed countries like the United States. Beyond the chance coexistence of both of these conditions in the same patient based on high prevalence it is now apparent that obesity is associated with and likely has a direct causal role in the onset, progression and severity of chronic kidney disease. The causes and underlying pathophysiology of this are myriad, complicated and multi-faceted. In this review, continuing the theme of this special edition of the journal on " The Cross roads between Endocrinology and Nephrology" we review the epidemiology of obesity related chronic kidney disease (ORCKD), and its various underlying causes and pathophysiology. In addition, we delve into the consequent comorbidities and complications associated with ORCKD with particular emphasis on the cardio metabolic consequences and then review the current body of evidence for available strategies for chronic kidney disease modulation in ORCKD as well as the potential unique role of weight reduction and management strategies in its improvement and risk reduction.
Collapse
Affiliation(s)
- Mariam M. Ali
- Southern Illinois School of Medicine, Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, 751 North Rutledge Street, Moy Building, Suite 1700, Springfield, Il 62702, United States
| | - Sanober Parveen
- Southern Illinois School of Medicine, Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, 751 North Rutledge Street, Moy Building, Suite 1700, Springfield, Il 62702, United States
| | - Vanessa Williams
- Southern Illinois School of Medicine, Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, 751 North Rutledge Street, Moy Building, Suite 1700, Springfield, Il 62702, United States
| | - Robert Dons
- Southern Illinois School of Medicine, Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, 751 North Rutledge Street, Moy Building, Suite 1700, Springfield, Il 62702, United States
| | - Gabriel I. Uwaifo
- Section of Endocrinology, Dept of Medicine, SIU School of Medicine, 751 N Rutledge St, Moy Building, Suite 1700, Room #1813, Springfield, Il 62702, United States
| |
Collapse
|
4
|
Hao M, Lv Y, Liu S, Guo W. The New Challenge of Obesity - Obesity-Associated Nephropathy. Diabetes Metab Syndr Obes 2024; 17:1957-1971. [PMID: 38737387 PMCID: PMC11086398 DOI: 10.2147/dmso.s433649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/28/2024] [Indexed: 05/14/2024] Open
Abstract
In recent years, obesity has become one of the major diseases that affect human health and consume human health resources, especially when it causes comorbidities such as hypertension, diabetes, cardiovascular disease and kidney disease. Many studies have demonstrated that obesity is associated with the development of chronic kidney disease and can exacerbate the progression of end-stage renal disease. This review described the mechanisms associated with the development of obesity-associated nephropathy and the current relevant therapeutic modalities, with the aim of finding new therapeutic targets for obesity-associated nephropathy. The mechanisms of obesity-induced renal injury include, in addition to the traditional alterations in renal hemodynamics, the involvement of various mechanisms such as macrophage infiltration in adipose tissue, alterations in adipokines (leptin and adiponectin), and ectopic deposition of lipids. At present, there is no "point-to-point" treatment for obesity-induced kidney injury. The renin-angiotensin-aldosterone system (RAAS) inhibitors, sodium-dependent glucose transporter 2 (SGLT-2) inhibitors and bariatric surgery described in this review can reduce urinary protein to varying degrees and delay the progression of kidney disease. In addition, recent studies on the therapeutic effects of intestinal flora on obesity may reduce the incidence of obesity-related kidney disease from the perspective of primary prevention. Both of these interventions have their own advantages and disadvantages, so the continuous search for the mechanism of obesity-induced related kidney disease will be extremely helpful for the future treatment of obesity-related kidney disease.
Collapse
Affiliation(s)
- Mengjin Hao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
- Department of Endocrinology, Jining No. 1 People’s Hospital, Jining, Shandong, 272000, People’s Republic of China
| | - You Lv
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
| | - Siyuan Liu
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
| | - Weiying Guo
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China
| |
Collapse
|
5
|
Peng L, Sun X, Yi X, Wang Z, Chen K. EGR3 reduces podocyte inflammatory damage in obesity related glomerulopathy by inhibiting the PRMT1 /p -STAT3 pathway. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2024; 49:349-358. [PMID: 38970508 PMCID: PMC11208401 DOI: 10.11817/j.issn.1672-7347.2024.230394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Indexed: 07/08/2024]
Abstract
OBJECTIVES Obesity related glomerulopathy (ORG) is induced by obesity, but the pathogenesis remains unclear. This study aims to investigate the expression of early growth response protein 3 (EGR3) in the renal cortex tissues of ORG patients and high-fat diet-induced obese mice, and to further explore the molecular mechanism of EGR3 in inhibiting palmitic acid (PA) induced human podocyte inflammatory damage. METHODS Renal cortex tissues were collected from ORG patients (n=6) who have been excluded from kidney damage caused by other diseases and confirmed by histopathology, and from obese mice induced by high-fat diet (n=10). Human and mouse podocytes were intervened with 150 μmol/L PA for 48 hours. EGR3 was overexpressed or silenced in human podocytes. Enzyme linked immunosorbent assay (ELISA) was used to detcet the levels of interleukin-6 (IL-6) and interleukin-1β (IL-1β). Real-time RT-PCR was used to detect the mRNA expressions of EGR3, podocytes molecular markers nephrosis 1 (NPHS1), nephrosis 2 (NPHS2), podocalyxin (PODXL), and podoplanin (PDPN). RNA-seq was performed to detect differentially expressed genes (DEGs) after human podocytes overexpressing EGR3 and treated with 150 μmol/L PA compared with the control group. Co-immunoprecipitation (Co-IP) combined with liquid chromatography tandem mass spectrometry (LC-MS) was used to detect potential interacting proteins of EGR3 and the intersected with the RNA-seq results. Co-IP confirmed the interaction between EGR3 and protein arginine methyltransferases 1 (PRMT1), after silencing EGR3 and PRMT1 inhibitor intervention, the secretion of IL-6 and IL-1β in PA-induced podocytes was detected. Western blotting was used to detect the expression of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) after overexpression or silencing of EGR3. RESULTS EGR3 was significantly upregulated in renal cortex tissues of ORG patients and high-fat diet-induced obese mice (both P<0.01). In addition, after treating with 150 μmol/L PA for 48 hours, the expression of EGR3 in human and mouse podocytes was significantly upregulated (both P<0.05). Overexpression or silencing of EGR3 in human podocytes inhibited or promoted the secretion of IL-6 and IL-1β in the cell culture supernatant after PA intervention, respectively, and upregulated or downregulated the expression of NPHS1, PODXL, NPHS2,and PDPN (all P<0.05). RNA-seq showed a total of 988 DEGs, and Co-IP+LC-MS identified a total of 238 proteins that may interact with EGR3. Co-IP confirmed that PRMT1 was an interacting protein with EGR3. Furthermore, PRMT1 inhibitors could partially reduce PA-induced IL-6 and IL-1β secretion after EGR3 silencing in human podocytes (both P<0.05). Overexpression or silencing of EGR3 negatively regulated the expression of PRMT1 and p-STAT3. CONCLUSIONS EGR3 may reduce ORG podocyte inflammatory damage by inhibiting the PRMT1/p-STAT3 pathway.
Collapse
Affiliation(s)
- Lin Peng
- Department of Nephrology, First Hospital of Changsha, Changsha 410005.
| | - Xiaoying Sun
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xuan Yi
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhouqi Wang
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ke Chen
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
6
|
Ibrahim M, Khalife L, Abdel-Latif R, Faour WH. Ghrelin hormone a new molecular modulator between obesity and glomerular damage. Mol Biol Rep 2023; 50:10525-10533. [PMID: 37924451 DOI: 10.1007/s11033-023-08866-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/27/2023] [Indexed: 11/06/2023]
Abstract
The incidence of glomerular diseases is increasing worldwide due to increased prevalence of obesity which is a major risk factor for type-2 diabetes mellitus and cardiovascular disorders.Ghrelin, an orexigenic peptide hormone, has been implicated in obesity, and its impact on the pathology and function of the kidneys was found to be significant. Ghrelin known to regulate energy homeostasis and growth hormone release, has been shown to modulate critical signaling pathways involved in the health and survival of podocytes. These derangements directly affect glomerular function and manifest as impaired glomerular filtration barrier and leakage of albumin into urine. Although the pathological features of the above-mentioned disorders are different, they interestingly lead to similar clinical features of glomerular damage. The pathological events are majorly initiated by endocrine imbalance leading to abnormal activation of downstream signaling pathways involved in the development of glomerulosclerosis. In fact, obesity increases the risk of developing chronic kidney disease by altering the secretion of pro-inflammatory cytokines and adipokines, activating the renin-angiotensin-aldosterone system (RAAS), promoting lipotoxicity, oxidative stress and fibrosis within the kidneys. Whilst these bioregulators are well described, their direct involvement in renal homeostasis is still mostly elusive. This review summarized previous and recent evidence on the endocrine properties of ghrelin and perivascular adipose tissue involved in modulating kidney physiology.
Collapse
Affiliation(s)
- Maroun Ibrahim
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Lynn Khalife
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Rania Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Miniya, Egypt
| | - Wissam H Faour
- Gilbert & Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon.
| |
Collapse
|
7
|
Zhou Y, Chen Z, Zhou H, Niu B, Liu J, Li Y, Mi Y, Li P. ACT001 Alleviates chronic kidney injury induced by a high-fat diet in mice through the GPR43/AMPK pathway. Lipids Health Dis 2023; 22:198. [PMID: 37978497 PMCID: PMC10657122 DOI: 10.1186/s12944-023-01949-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Roughly 10 -15% of global populace suffer from Chronic Kidney Disease(CKD). A major secondary disease that can progress to end-stage renal disease (ESRD) is obesity-associated kidney disease (ORG). Although clinical management strategies are currently available, morbidity and mortality rates are increasing. Thus, new solutions are needed. Intestinal permeability, systemic inflammation, and aberrant intestinal metabolites have all been linked to ORG. PURPOSE ACT001 has anti-inflammatory, redox-regulatory and antitumour activities. The current study was designed to examine how ACT001 affects ORG and analyze the fundamental processes. METHODS A high-fat diet (HFD) was used to generate ORG in female C57BL/6 J mice. ORG mice were divided into three groups at random: HFD, HFD + ACT001, HFD + polyphosphocholine (PPC). To assess renal and colonic damage, periodic acid-Schiff (PAS) and hematoxylin-eosin (HE) staining were used. Following that, renal inflammation, oxidative stress, lipid deposition, colonic inflammation, and intestinal permeability were evaluated by protein blotting, polymerase chain reaction (PCR), immunohistochemistry, and immunofluorescence staining. Lastly, the SCFAs content was assessed by gas chromatographymass spectrometry. RESULTS Mice in the HFD group displayed more severe albuminuria, glomerular hypertrophy, renal oxidative damage, inflammation, and lipid accumulation than mice with the normal diet (ND) group, as well as lower levels of intestinal SCFA valproic acid, colonic inflammation, and tight junction protein downregulation. ACT001 treatment restores the content of valproic acid in intestinal SCFAs, promotes the binding of SCFAs to renal GPR43, activates the AMPK signalling pathway. Therefore, it promotes the Nrf2-Keap1 signalling pathway and inhibits the NF-κB signalling pathway. SCFAs, additionally, augment colonic GPR43 concentrations, diminishing NLRP3 inflammasome expression and restoring ZO-1 and occludin protein levels. CONCLUSION This study is the first to look at ACT001's potential as a treatment for obesity-related kidney disease. Regulating GPR43 and AMPK signalling pathways, By controlling the GPR43 and AMPK signalling pathways, ACT001 improves colitis and the intestinal mucosal barrier, decreases renal lipid deposition, and suppresses inflammation and oxidative stress in the kidneys. According to this study, ACT001 could be a viable ORG therapy option.
Collapse
Affiliation(s)
- Yibing Zhou
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
| | - Ze Chen
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
| | - Hui Zhou
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
| | - Bin Niu
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
| | - Jing Liu
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
| | - Yinglun Li
- Clinical School of the Second People's Hospital, Tianjin Medical University, Tianjin, China
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
| | - Yuqiang Mi
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China
- Tianjin Research Institute of Liver Disease, Tianjin, China
| | - Ping Li
- Department of Hepatology, Tianjin Second People's Hospital, Tianjin, China.
- Tianjin Research Institute of Liver Disease, Tianjin, China.
| |
Collapse
|
8
|
Hti Lar Seng NS, Lohana P, Chandra S, Jim B. The Fatty Kidney and Beyond: A Silent Epidemic. Am J Med 2023; 136:965-974. [PMID: 37451390 DOI: 10.1016/j.amjmed.2023.06.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
As the prevalence of obesity rises in the United States, so does the incidence of obesity-related kidney disease. Obesity itself is an independent risk factor for chronic kidney disease where the pathophysiology is complex, involving altered hemodynamics, renin-angiotensin-aldosterone system overactivation, and adipokines leading to inflammation and fibrosis. Obesity-related kidney disease comprises both obesity-related glomerulopathy and fatty kidney disease. Obesity-related glomerulopathy is a consequence of glomerular hyperfiltration and often presents clinically with subnephrotic proteinuria and pathologically with glomerulomegaly with or without focal glomerulosclerosis. Fatty kidney disease is the effect of renal ectopic fat contributing to chronic kidney disease. Whether the renal ectopic fat is a distinct clinical entity or a pathologic mechanism contributing to obesity-related glomerulopathy, the treatment paradigm of weight and proteinuria reduction remains the same. We present the pathophysiology behind obesity-related kidney disease, clinical outcomes, and treatment strategies, which include lifestyle interventions, use of renin-angiotensin-aldosterone system inhibitors, glucagon-like peptide 1 receptor agonists, sodium-glucose co-transporter-2 inhibitors, and bariatric surgery. With old and novel therapeutics, we are attempting to stave off the silent epidemic that obesity-related kidney disease is becoming.
Collapse
Affiliation(s)
- Nang San Hti Lar Seng
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine Bronx, NY
| | - Petras Lohana
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine Bronx, NY
| | - Shruti Chandra
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine Bronx, NY
| | - Belinda Jim
- Division of Nephrology, Department of Medicine, Jacobi Medical Center at Albert Einstein College of Medicine Bronx, NY.
| |
Collapse
|
9
|
Kreiner FF, Schytz PA, Heerspink HJL, von Scholten BJ, Idorn T. Obesity-Related Kidney Disease: Current Understanding and Future Perspectives. Biomedicines 2023; 11:2498. [PMID: 37760939 PMCID: PMC10526045 DOI: 10.3390/biomedicines11092498] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Obesity is a serious chronic disease and an independent risk factor for the new onset and progression of chronic kidney disease (CKD). CKD prevalence is expected to increase, at least partly due to the continuous rise in the prevalence of obesity. The concept of obesity-related kidney disease (OKD) has been introduced to describe the still incompletely understood interplay between obesity, CKD, and other cardiometabolic conditions, including risk factors for OKD and cardiovascular disease, such as diabetes and hypertension. Current therapeutics target obesity and CKD individually. Non-pharmacological interventions play a major part, but the efficacy and clinical applicability of lifestyle changes and metabolic surgery remain debatable, because the strategies do not benefit everyone, and it remains questionable whether lifestyle changes can be sustained in the long term. Pharmacological interventions, such as sodium-glucose co-transporter 2 inhibitors and the non-steroidal mineralocorticoid receptor antagonist finerenone, provide kidney protection but have limited or no impact on body weight. Medicines based on glucagon-like peptide-1 (GLP-1) induce clinically relevant weight loss and may also offer kidney benefits. An urgent medical need remains for investigations to better understand the intertwined pathophysiologies in OKD, paving the way for the best possible therapeutic strategies in this increasingly prevalent disease complex.
Collapse
Affiliation(s)
| | | | - Hiddo J. L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, 9700 RB Groningen, The Netherlands;
| | | | - Thomas Idorn
- Novo Nordisk A/S, DK-2860 Søborg, Denmark; (F.F.K.); (P.A.S.); (B.J.v.S.)
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Chronic kidney disease (CKD) is a common condition and a major cause of morbidity and mortality in adults, but children and adolescents are also at risk for early kidney injury and development of CKD. Obesity contributes both directly and indirectly to the development of CKD. The purpose of this review is to describe obesity-related kidney disease (ORKD) and diabetic kidney disease (DKD) and their impact in the pediatric population. RECENT FINDINGS Although obesity-related CKD in childhood and adolescence is uncommon, nascent kidney damage may magnify the lifetime risk of CKD. Glomerular hyperfiltration is an early phenotype of both ORKD and DKD and typically manifests prior to albuminuria and progressive decline in GFR. Novel treatments for obesity and type 2 diabetes exerting protective effects on the kidneys are being investigated for use in the pediatric population. It is important to understand the impact of obesity on the kidneys more fully in the pediatric population to help detect injury earlier and intervene prior to the onset of irreversible progression of disease and to guide future research in this area.
Collapse
Affiliation(s)
- Alexandra Sawyer
- Department of Pediatrics, Division of Endocrinology, University of Colorado School of Medicine, 13123 East 16Th Avenue, Box 158, Aurora, CO, 80045, USA.
| | - Evan Zeitler
- Department of Medicine, Division of Nephrology and Hypertension, University of North Carolina Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Howard Trachtman
- Department of Pediatrics, Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Petter Bjornstad
- Department of Pediatrics, Division of Endocrinology, University of Colorado School of Medicine, 13123 East 16Th Avenue, Box 158, Aurora, CO, 80045, USA
| |
Collapse
|
11
|
Zhao L, Zou Y, Wu Y, Cai L, Zhao Y, Wang Y, Xiao X, Yang Q, Yang J, Ren H, Tong N, Liu F. Metabolic phenotypes and risk of end-stage kidney disease in patients with type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1103251. [PMID: 37234807 PMCID: PMC10206309 DOI: 10.3389/fendo.2023.1103251] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/13/2023] [Indexed: 05/28/2023] Open
Abstract
Background Obesity often initiates or coexists with metabolic abnormalities. This study aimed to investigate the pathological characteristics and the independent or mutual relations of obesity and metabolic abnormalities with end-stage kidney disease (ESKD) in patients with type 2 diabetes (T2D) and associated diabetic kidney disease (DKD). Methods A total of 495 Chinese patients with T2D and biopsy-confirmed DKD between 2003 and 2020 were enrolled in this retrospective study. The metabolic phenotypes were based on the body weight index (BMI)-based categories (obesity, BMI ≥ 25.0 kg/m2) and metabolic status (metabolically unhealthy status, ≥ 1 criterion National Cholesterol Education Program Adult Treatment Panel III (NCEP/ATP III) excluding waist circumference and hyperglycemia) and were categorized into four types: metabolically healthy non-obesity (MHNO), metabolically healthy obesity (MHO), metabolically unhealthy non-obesity (MUNO), and metabolically unhealthy obesity (MUO). The pathological findings were defined by the Renal Pathology Society classification. Cox proportional hazards models were used to estimate hazard ratios (HRs) for ESKD. Results There are 56 (11.3%) MHNO patients, 28 (5.7%) MHO patients, 176 (35.6%) MUNO patients, and 235 (47.5%) MUO patients. The high prevalence of the Kimmelstiel-Wilson nodule and severe mesangial expansion were associated with obesity, whereas severe IFTA was related to metabolically unhealthy status. In the multivariate analysis, the adjusted HR (aHR) was 2.09 [95% confidence interval (CI) 0.99-4.88] in the MHO group, 2.16 (95% CI 1.20-3.88) in the MUNO group, and 2.31 (95% CI 1.27-4.20) in the MUO group compared with the MHNO group. Furthermore, the presence of obesity was insignificantly associated with ESKD compared with non-obese patients (aHR 1.22, 95% CI 0.88-1.68), while the metabolically unhealthy status was significantly associated with ESKD compared to the metabolically healthy status in the multivariate analysis (aHR 1.69, 95% CI 1.10-2.60). Conclusion Obesity itself was insignificantly associated with ESKD; however, adding a metabolically unhealthy status to obesity increased the risk for progression to ESKD in T2D and biopsy-proven DKD.
Collapse
Affiliation(s)
- Lijun Zhao
- Department of General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yutong Zou
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yucheng Wu
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Linli Cai
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuancheng Zhao
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yiting Wang
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiang Xiao
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qing Yang
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jia Yang
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Honghong Ren
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Nanwei Tong
- Division of Endocrinology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Fang Liu
- Department of Nephrology, Laboratory of Diabetic Kidney Disease, Centre of Diabetes and Metabolism Research, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Song D, Zhang A, Hu X, Zeng M, Zhou H. Wen-Shen-Jian-Pi-Hua-Tan decoction protects against early obesity-related glomerulopathy by improving renal bile acid composition and suppressing lipogenesis, inflammation, and fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154861. [PMID: 37167823 DOI: 10.1016/j.phymed.2023.154861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND Obesity is an independent predictor of chronic kidney disease (CKD) development and may directly lead to kidney lesions such as obesity-related glomerulopathy (ORG) which might play a vital pathogenic role in obese patients with CKD. Wen-Shen-Jian-Pi-Hua-Tan decoction (WSHT) has been clinically used for the treatment of obesity and obesity-related metabolic diseases for years. However, the renoprotective effects and potential mechanism of action of WSHT against ORG remain unknown. PURPOSE This study aimed to explore the potential effect of WSHT on ORG and reveal its mechanisms in high-fat diet (HFD)-induced obese rats. METHODS An animal model of early stage ORG was established using HFD-induced obese rats. After treatment with WSHT for 6 weeks, an integrated metabolomics and molecular biology strategy was utilized to illustrate the effects and mechanism of WSHT on ORG. First, UPLC-ESI-MS/MS-based targeted metabolomics was used to analyze renal bile acid (BA) levels. Biochemical, histological, and immunofluorescence assays; electron microscopy; and western blotting were performed to evaluate the efficacy of WSHT against ORG and its underlying mechanisms in vivo. RESULTS Our results showed that an HFD led to hyperlipidemia, proteinuria, renal lipid deposition, effacement of podocyte foot processes, and increased expression of proinflammatory factors and profibrotic growth factors in ORG rats. In addition, an HFD decreased the levels of renal BAs such as cholic acid, chenodeoxycholic acid, and lithocholic acid. After 6 weeks of treatment, WSHT markedly attenuated dyslipidemia and reduced body, kidney and epididymal fat weights in ORG rats. WSHT also significantly increased BA levels, suggesting that it altered BA composition; the effects of BAs are closely associated with farnesoid X receptor (FXR) activation. WSHT alleviated fat accumulation, podocyte loss and proteinuria, and reduced the expression of proinflammatory cytokines and profibrotic growth factors in the kidneys of ORG rats. Finally, WSHT remarkably upregulated the renal expression of FXR and salt-induced kinase 1 and blocked the renal expression of sterol regulatory element-binding protein-1c and its target genes. CONCLUSION WSHT attenuated early renal lesions in ORG rats by improving renal BA composition and suppressing lipogenesis, inflammation and fibrosis. This study develops a new way to alleviate obesity-induced renal damages.
Collapse
Affiliation(s)
- Daofei Song
- Department of Endocrinology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan 430015, China
| | - Aijie Zhang
- Department of Gynaecology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, 430015, China
| | - Xu Hu
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - MingXing Zeng
- Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Huimin Zhou
- Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
13
|
Verde L, Lucà S, Cernea S, Sulu C, Yumuk VD, Jenssen TG, Savastano S, Sarno G, Colao A, Barrea L, Muscogiuri G. The Fat Kidney. Curr Obes Rep 2023:10.1007/s13679-023-00500-9. [PMID: 36933154 DOI: 10.1007/s13679-023-00500-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/28/2023] [Indexed: 03/19/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current evidence on the role of obesity in the development and progression of chronic kidney disease and the current evidence on nutritional, pharmacological, and surgical strategies for the management of individuals with obesity and chronic kidney disease. RECENT FINDINGS Obesity can hurt the kidney via direct pathways, through the production of pro-inflammatory adipocytokines, and indirectly due to systemic complications of obesity, including type 2 diabetes mellitus and hypertension. In particular, obesity can damage the kidney through alterations in renal hemodynamics resulting in glomerular hyperfiltration, proteinuria and, finally, impairment in glomerular filtratation rate. Several strategies are available for weight loss and maintenance, such as the modification of lifestyle (diet and physical activity), anti-obesity drugs, and surgery therapy, but there are no clinical practice guidelines to manage subjects with obesity and chronic kidney disease. Obesity is an independent risk factor for the progression of chronic kidney disease. In subjects with obesity, weight loss can slow down the progression of renal failure with a significant reduction in proteinuria and improvement in glomerular filtratation rate. Specifically, in the management of subjects with obesity and chronic renal disease, it has been shown that bariatric surgery can prevent the decline in renal function, while further clinical studies are needed to evaluate the efficacy and safety on the kidney of weight reducing agents and the very low-calorie ketogenic diet.
Collapse
Affiliation(s)
- Ludovica Verde
- Department of Public Health, University of Naples Federico II, Naples, Italy
- Centro Italiano Per La Cura E il Benessere del Paziente con Obesità (C.I.B.O), University of Naples Federico II, Naples, Italy
| | - Stefania Lucà
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy
| | - Simona Cernea
- Technology of Târgu Mures/Internal Medicine I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureş, Romania
- Diabetes, Nutrition and Metabolic Diseases Outpatient Unit, Emergency County Clinical Hospital, Târgu Mureş, Romania
| | - Cem Sulu
- Division of Endocrinology, Metabolism and Diabetes, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Volkan Demirhan Yumuk
- Division of Endocrinology, Metabolism and Diabetes, Cerrahpaşa Medical Faculty, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Trond Geir Jenssen
- Department of Transplantation Medicine, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Silvia Savastano
- Centro Italiano Per La Cura E il Benessere del Paziente con Obesità (C.I.B.O), University of Naples Federico II, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Diabetologia e Andrologia, Unità di Endocrinologia, Università degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Gerardo Sarno
- General Surgery and Kidney Transplantation Unit, d'Aragona University Hospital, San Giovanni di Dio e Ruggid, 84131, Salerno, Italy
| | - Annamaria Colao
- Centro Italiano Per La Cura E il Benessere del Paziente con Obesità (C.I.B.O), University of Naples Federico II, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Diabetologia e Andrologia, Unità di Endocrinologia, Università degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco Educazione alla salute e allo sviluppo sostenibile, University Federico II, Naples, Italy
| | - Luigi Barrea
- Centro Italiano Per La Cura E il Benessere del Paziente con Obesità (C.I.B.O), University of Naples Federico II, Naples, Italy
- Dipartimento di Scienze Umanistiche, Centro Direzionale, Università Telematica Pegaso, Via Porzio, Isola F2, 80143, Naples, Italy
| | - Giovanna Muscogiuri
- Centro Italiano Per La Cura E il Benessere del Paziente con Obesità (C.I.B.O), University of Naples Federico II, Naples, Italy.
- Dipartimento di Medicina Clinica e Chirurgia, Diabetologia e Andrologia, Unità di Endocrinologia, Università degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Cattedra Unesco Educazione alla salute e allo sviluppo sostenibile, University Federico II, Naples, Italy.
| |
Collapse
|
14
|
Abstract
The prevalence of obesity has increased dramatically during the past decades, which has been a major health problem. Since 1975, the number of people with obesity worldwide has nearly tripled. An increasing number of studies find obesity as a driver of chronic kidney disease (CKD) progression, and the mechanisms are complex and include hemodynamic changes, inflammation, oxidative stress, and activation of the renin-angiotensin-aldosterone system (RAAS). Obesity-related kidney disease is characterized by glomerulomegaly, which is often accompanied by localized and segmental glomerulosclerosis lesions. In these patients, the early symptoms are atypical, with microproteinuria being the main clinical manifestation and nephrotic syndrome being rare. Weight loss and RAAS blockers have a protective effect on obesity-related CKD, but even so, a significant proportion of patients eventually progress to end-stage renal disease despite treatment. Thus, it is critical to comprehend the mechanisms underlying obesity-related CKD to create new tactics for slowing or stopping disease progression. In this review, we summarize current knowledge on the mechanisms of obesity-related kidney disease, its pathological changes, and future perspectives on its treatment.
Collapse
Affiliation(s)
- Zongmiao Jiang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Yao Wang
- Department of Orthopedics, The Second Hospital Jilin University, Changchun, China
| | - Xue Zhao
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Haiying Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Mingyue Han
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Xinhua Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Satoh K, Nagasawa K, Takebe N, Kinno H, Shozushima M, Onodera K, Oda T, Hasegawa Y, Satoh J, Ishigaki Y. Adiponectin Paradox More Evident in Non-Obese Than in Obese Patients with Diabetic Microvascular Complications. Diabetes Metab Syndr Obes 2023; 16:201-212. [PMID: 36760589 PMCID: PMC9882416 DOI: 10.2147/dmso.s387744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023] Open
Abstract
AIMS/INTRODUCTION Adiponectin is generally regarded as a beneficial molecule, protecting against insulin resistance and atherosclerosis, and its serum levels are low in individuals with obesity as well as in those with type 2 diabetes (T2DM). However, several clinical studies have shown associations between high adiponectin values and major health concerns. These conflicting findings are termed the "adiponectin paradox". Similarly, these paradoxical adiponectin elevations were observed in patients with diabetic microvascular complications. This cross-sectional study aimed to identify differences in factors, including adiponectin, related to diabetic vascular complications between non-obese and obese patients. MATERIALS AND METHODS Study patients with T2DM were non-obese (n=197) or obese (n=197), matched by a propensity score model adjusted with age and gender. Independent factors for each of the microvascular complications were determined using multivariate logistic regression analyses. RESULTS The prevalence of nephropathy was high in obese T2DM patients. In addition to long diabetes duration, elevated adiponectin was a common characteristic of patients with microvascular complications. Logistic regression analyses for microvascular complications revealed adiponectin to be highly related to retinopathy (odds ratio [OR], 1.138; 95%confidence intervals [CI], 1.004-1.289, p<0.001), nephropathy (OR, 1.192; CI, 1.077-1.319, p<0.001) and neuropathy (OR, 1.217; CI, 1.071-1.384, p<0.001), in non-obese patients. In contrast, the association between adiponectin values and complications was modest in obese patients. CONCLUSION Adiponectin regulation in response to vascular damage differed between non-obese and obese patients, suggesting that adiponectin regulation is compromised by fat accumulation. Assuming that paradoxical elevation of adiponectin in vascular damage is a compensatory response, we speculate that responsive upregulation might be insufficient in obese patients. These newly-recognized differences in adiponectin values might lead to novel insights into adiponectin regulation and our understanding of the adiponectin paradox.
Collapse
Affiliation(s)
- Ken Satoh
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Kan Nagasawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Noriko Takebe
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Hirofumi Kinno
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Masaharu Shozushima
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Ken Onodera
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Tomoyasu Oda
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Yutaka Hasegawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
| | - Jo Satoh
- Department of Diabetes, Tohoku Medical and Pharmaceutical University, Wakabayashi Hospital, Sendai, Japan
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Yahaba, Japan
- Correspondence: Yasushi Ishigaki, Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, 2-1-1 Idai-Dori, Yahaba, Iwate, 028-3695, Japan, Tel +81 19 613 7111, Fax +81 19 907 8270, Email
| |
Collapse
|
16
|
Wen W, Huang B, Ye S. Metformin Ameliorates Epithelial-Mesenchymal Transition of Renal Tubular Epithelial Cells in Diabetes by Increasing Vitamin D Receptor Expression. Diabetes Metab Syndr Obes 2022; 15:4001-4010. [PMID: 36582506 PMCID: PMC9792813 DOI: 10.2147/dmso.s389918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Metformin is used as a first-line drug for the treatment of type 2 diabetes. Epithelial-mesenchymal transition (EMT) plays a significant role in the development of renal tubular damage in diabetic kidney disease. However, the underlying mechanisms of EMT in diabetic kidney disease are unclear and how to inhibit this process remains to be explored. METHODS C57 mice were randomly divided into four groups, including the normal control group (NC group), the Type 2 diabetes group (T2DM group), the metformin group (MET group), and glibenclamide group (GLIB). Fasting blood glucose (FBG), glycated hemoglobin (HbA1c), urinary albumin, RBP, PCX, and creatinine were measured. Renal pathology was observed with HE staining. Molecular mechanism of VDR expression are regulated by metformin through wound healing assay, and Western blot analysis of VDR, Ecad, and SMA in HK2 cells. RESULTS In animal experiments, compared with the NC group, the T2DM group showed decreased body weight, increased levels of FBG, HbA1c, UAlb/UCR, URBP/UCR, and UPCX/UCR, decreased levels of VDR protein and mRNA expression in renal tissues (P < 0.05), and significantly increased renal pathological damage in mice in the T2DM group. Compared with the T2DM group, mice in the GLIB and MET groups had higher body weight and lower FBG, HbA1c, UAlb/UCR, URBP/UCR, and UPCX/UCR (P < 0.05). In addition, renal pathological damage was significantly reduced in the MET group compared to the GLIB group. In HK2 cells, high glucose promoted the reduction of VDR and the development of EMT compared to the NC group. In addition, we found that Metformin can up-regulate VDR and inhibit EMT. CONCLUSION Our study shows that the renoprotective effect of metformin is independent of glycemic control and metformin is involved in the progression of EMT by regulating VDR expression.
Collapse
Affiliation(s)
- Wenjie Wen
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of University of Science and Technology of China (USTC), Department of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People’s Republic of China
- Department of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People’s Republic of China
| | - Bin Huang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of University of Science and Technology of China (USTC), Department of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People’s Republic of China
- Department of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People’s Republic of China
| | - Shandong Ye
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of University of Science and Technology of China (USTC), Department of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People’s Republic of China
- Department of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People’s Republic of China
- Correspondence: Shandong Ye, University of Science and Technology of China, No. 96 Jinzhai Road, Hefei, Anhui Province, 230026, People’s Republic of China, Email
| |
Collapse
|
17
|
Xie J, Zhong F, Guo Z, Li X, Wang J, Gao Z, Chang B, Yang J. Hyperinsulinemia impairs the metabolic switch to ketone body utilization in proximal renal tubular epithelial cells under energy crisis via the inhibition of the SIRT3/SMCT1 pathway. Front Endocrinol (Lausanne) 2022; 13:960835. [PMID: 36237185 PMCID: PMC9551351 DOI: 10.3389/fendo.2022.960835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/31/2022] [Indexed: 12/05/2022] Open
Abstract
OBJECTIVE To investigate the effects and mechanism of hyperinsulinemia on the metabolic switch to β-hydroxybutyrate (BHB) absorption and utilization under a starvation or hypoxic environment in proximal tubular epithelial cells. METHODS A high-fat diet-induced hyperinsulinemia model in ZDF rats was used to test the expression of key enzymes/proteins of ketone body metabolism in the kidney. Notably, 12-week-old renal tubule SMCT1 specific knockout mice (SMCT1 flox/floxCre+) and control mice (SMCT1 flox/floxCre-) were used to confirm the roles of SMCT1 in kidney protection under starvation. The changes of key enzymes/proteins of energy metabolism, mitochondrial function, and albumin endocytosis in HK2 cells under low glucose/hypoxic environments with or without 50 ng/mL insulin were studied. Silent information regulation 2 homolog 3 (SIRT3) was overexpressed to evaluate the effect of hyperinsulinemia on the metabolic switch to BHB absorption and utilization through the SIRT3/SMCT1 pathway in HK2 cells. RESULTS In ZDF rats, the expression of HMGCS2 increased, the SMCT1 expression decreased, while SCOT remained unchanged. In renal tubule SMCT1 gene-specific knockout mice, starvation for 48 h induced an increase in the levels of urine retinol-binding protein, N-acetyl-β-glucosaminidase, and transferrin, which reflected tubular damages. In HK2 cells under an environment of starvation and hypoxia, the levels of key enzymes related to fatty acid oxidation and ketone body metabolism were increased, whereas glucose glycolysis did not change. The addition of 2 mmol/l BHB improved ATP production, mitochondrial biosynthesis, and endocytic albumin function, while cell apoptosis was reduced in HK2 cells. The addition of 50 ng/ml insulin resulted in the decreased expression of SMCT1 along with an impaired mitochondrial function, decreased ATP production, and increased apoptosis. The overexpression of SIRT3 or SMCT1 reversed these alterations induced by a high level of insulin both in low-glucose and hypoxic environments. CONCLUSIONS The increased absorption and utilization of BHB is part of the metabolic flexibility of renal tubular epithelial cells under starvation and hypoxic environments, which exhibits a protective effect on renal tubular epithelial cells by improving the mitochondrial function and cell survival. Moreover, hyperinsulinemia inhibits the absorption of BHB through the inhibition of the SIRT3/SMCT1 pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Juhong Yang
- *Correspondence: Juhong Yang, ; Baocheng Chang,
| |
Collapse
|
18
|
Ye T, Zhang J, Wu D, Shi J, Kuang Z, Ma Y, Xu Q, Chen B, Kan C, Sun X, Han F. Empagliflozin Attenuates Obesity-Related Kidney Dysfunction and NLRP3 Inflammasome Activity Through the HO-1-Adiponectin Axis. Front Endocrinol (Lausanne) 2022; 13:907984. [PMID: 35784553 PMCID: PMC9248377 DOI: 10.3389/fendo.2022.907984] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/12/2022] [Indexed: 01/17/2023] Open
Abstract
Empagliflozin (EMPA) is a novel sodium-glucose cotransporter 2 inhibitor (SGLT2i) that produces protective cardiovascular-renal outcomes in patients with diabetes. However, the effects of EMPA on obesity-related kidney disease have not been determined. The heme oxygenase-1 (HO-1)-adiponectin axis is an essential antioxidant system with anti-apoptotic and anti-inflammatory properties. This study explored whether EMPA improves obesity-related kidney disease through regulation of the renal HO-1-mediated adiponectin axis. C57BL/6J mice were assigned to control, high-fat diet (HFD) groups, and EMPA (10 mg/kg) groups. HFD mice showed metabolic abnormality and renal injury, including increased urinary albumin excretion, morphologic changes, and lipid accumulation. EMPA treatment improved metabolic disorders and attenuated lipotoxicity-induced renal injury. Furthermore, EMPA treatment ameliorated renal NLRP3 inflammasome activity and upregulated the HO-1-adiponectin axis. Our findings indicate that EMPA improves obesity-related kidney disease through reduction of NLRP3 inflammasome activity and upregulation of the HO-1-adiponectin axis, suggesting a novel mechanism for SGLT2i-mediated renal protection in obesity.
Collapse
Affiliation(s)
- Tongtong Ye
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jingwen Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Di Wu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Junfeng Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zengguang Kuang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yuting Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Qian Xu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Bing Chen
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Xiaodong Sun, ; Fang Han,
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Xiaodong Sun, ; Fang Han,
| |
Collapse
|