1
|
Jaruan O, Promsan S, Thongnak L, Pengrattanachot N, Phengpol N, Sutthasupha P, Lungkaphin A. Pyridoxine exerts antioxidant effects on kidney injury manifestations in high-fat diet-induced obese rats. Chem Biol Interact 2025; 415:111513. [PMID: 40239886 DOI: 10.1016/j.cbi.2025.111513] [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: 11/12/2024] [Revised: 01/21/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025]
Abstract
The modern diet contains a substantial level of fat which is believed to be one of the leading causes of the progression of kidney disease. Several studies have already demonstrated that consumption of a high-fat diet (HFD) induces inflammation and oxidative stress, causing activation of upstream mechanisms associated with kidney injury. For the prevention of such pathological events, a change in diet or the taking of nutritional supplements are recommended as alternative treatments. One of the forms of vitamin B6, pyridoxine (PN), has been shown to be an effective antioxidant and can also inhibit the formation of advanced-glycation end products (AGEs). In this study, the protective effects of PN (100 mg/kg/day for a period of eight weeks) against HFD-induced complications in obese rats were investigated. Rats fed on a HFD developed obesity which promoted inflammation, glucose intolerance, AGE receptor upregulation, oxidative stress, and kidney dysfunction. Intervention using PN mitigated obesity-related events and the impairment of kidney function by markedly reducing oxidative stress and also restoring the activity of antioxidant enzymes. Other studies have shown that some vitamin B6 derivatives inhibit the formation of AGEs but our study shows for the first time that PN exerted an antiglycative effect in this HFD-induced obesity model. Consequently, PN could potentially be a novel supplement for obese individuals to avoid kidney injury.
Collapse
Affiliation(s)
- Onanong Jaruan
- Department of Physiology, Faculty of Medicine Chiang Mai University, Chiang Mai, Thailand
| | - Sasivimon Promsan
- Department of Physiology, Faculty of Medicine Chiang Mai University, Chiang Mai, Thailand
| | - Laongdao Thongnak
- Department of Physiology, Faculty of Medicine Chiang Mai University, Chiang Mai, Thailand; Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | - Nichakorn Phengpol
- Department of Physiology, Faculty of Medicine Chiang Mai University, Chiang Mai, Thailand
| | - Prempree Sutthasupha
- Department of Physiology, Faculty of Medicine Chiang Mai University, Chiang Mai, Thailand
| | - Anusorn Lungkaphin
- Department of Physiology, Faculty of Medicine Chiang Mai University, Chiang Mai, Thailand; Functional Foods for Health and Disease, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Functional Food Research Center for Well-Being, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
2
|
Singh A, Khushboo, Pandey M, Mattoo S, Pore SK, Bhattacharyya J. A glucose-responsive alginate-based hydrogel laden with modified GLP-1 and telmisartan ameliorates type 2 diabetes and reduces liver and kidney toxicities. J Mater Chem B 2025; 13:4419-4432. [PMID: 40095672 DOI: 10.1039/d4tb02261k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The pathophysiology associated with type 2 diabetes mellitus (T2DM) includes insulin resistance, increased oxidative stress, a pro-inflammatory macrophage population, and dysfunction of pancreatic β cells in the islets of Langerhans, along with hepato- and nephro-toxicity. In this study, an injectable glucose-responsive hydrogel (Diabogel) was developed using alginate and 3-aminophenyl boronic acid to deliver modified glucagon-like peptide-1, insulinoma cell-derived extracellular vesicles, and telmisartan. Diabogel demonstrated cytocompatibility, decreased reactive oxygen species, enhanced insulin synthesis, and improved glucose uptake in vitro. In a high-fat diet/streptozotocin-induced murine model of T2DM, Diabogel lowered blood glucose levels, maintained body weight, and increased insulin expression. Furthermore, it promoted an anti-inflammatory microenvironment in the pancreas by regulating macrophage phenotype and the expression of NF-κB, supported cellular proliferation, and restored the pancreatic islets. In addition, Diabogel treatment significantly lowered the serum levels of pro-inflammatory cytokines and enhanced anti-inflammatory cytokines. Interestingly, Diabogel treatment also lowered diabetes-associated hepato- and nephro-toxicity. Taken together, Diabogel may serve as a potential approach for the treatment of T2DM, regulating blood glucose levels, restoring pancreatic β cell function, and reducing hepatic and renal toxicities.
Collapse
Affiliation(s)
- Anjali Singh
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, New Delhi 110016, India.
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, New Delhi 110016, India
| | - Khushboo
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, New Delhi 110016, India.
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, New Delhi 110016, India
| | - Monu Pandey
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, New Delhi 110016, India.
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, New Delhi 110016, India
| | - Shria Mattoo
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Subrata Kumar Pore
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| | - Jayanta Bhattacharyya
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi, New Delhi 110016, India.
- Department of Biomedical Engineering, All India Institute of Medical Science, Delhi, New Delhi 110016, India
| |
Collapse
|
3
|
Sá JM, Fernandes MV, Pontes RB, Colombari E, Menani JV, Colombari DSA. Mechanisms involved in cardiovascular and hydroelectrolytic changes in dehydrated high-fat-diet-fed rats. Am J Physiol Regul Integr Comp Physiol 2025; 328:R481-R491. [PMID: 40059655 DOI: 10.1152/ajpregu.00171.2024] [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: 07/01/2024] [Revised: 07/26/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025]
Abstract
Obesity is increasingly prevalent worldwide, and climate change is exacerbating water shortages, leading to dehydration. Both obesity and dehydration cause increased arterial pressure (AP), fluid electrolytic imbalance, and neuroinflammation. Thus, the present study aimed to verify the changes in the cardiovascular system, hydroelectrolytic balance, and microglia and neuronal activation in rats fed with a high-fat diet (HFD) in response to 24 h of water deprivation (WD) and the possible mechanisms involved. Male Holtzman rats (290-310 g) were fed with a standard diet (SD, 10% calories from fat) or HFD (46% calories from fat) for 6 wk before the WD experiments. Compared with WD SD rats, WD HFD rats presented a greater c-Fos immunolabeling in the subfornical organ (SFO) and supraoptic nucleus and greater microglial activation in SFO. WD-induced water intake was lower in HFD rats than in SD rats. WD HFD rats presented greater antidiuresis and lesser natriuresis than WD SD rats. Renal denervation did not change the antidiuresis or natriuresis observed in WD HFD- or SD-fed rats. The lower water intake in WD HFD rats might be due to neuroinflammation and/or decreased urinary output. The increase in AP after WD was similar between HFD and SD, but it is more dependent on angiotensin II type 1 (AT1) receptor activation in HFD rats. Overall, HFD rats seem less responsive to fluid and electrolyte balance responses to WD, highlighting the need for strategies to prevent dehydration in individuals with obesity, particularly during rising drought conditions worldwide.NEW & NOTEWORTHY Obesity and dehydration are common worldwide. Our study with an animal model found that changes in arterial pressure are linked to increased activation of the AT1 receptor in obese, dehydrated rats. The renal nerves appear unrelated to the significant decrease in urinary volume and sodium excretion in these animals. Neuroinflammation and reduced urine output may explain their lower water intake. These findings highlight the need for strategies to prevent dehydration in individuals with obesity.
Collapse
Affiliation(s)
- Jéssica Matheus Sá
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University - UNESP, Araraquara, Brazil
| | - Marcos Vinícius Fernandes
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University - UNESP, Araraquara, Brazil
| | - Roberto Braz Pontes
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University - UNESP, Araraquara, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University - UNESP, Araraquara, Brazil
| | - José Vanderlei Menani
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University - UNESP, Araraquara, Brazil
| | | |
Collapse
|
4
|
Matsui S, Yamamoto T, Takabatake Y, Takahashi A, Namba-Hamano T, Matsuda J, Minami S, Sakai S, Yonishi H, Nakamura J, Maeda S, Matsumoto A, Matsui I, Yanagita M, Isaka Y. Empagliflozin protects the kidney by reducing toxic ALB (albumin) exposure and preventing autophagic stagnation in proximal tubules. Autophagy 2025; 21:583-597. [PMID: 39385699 PMCID: PMC11849939 DOI: 10.1080/15548627.2024.2410621] [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: 03/17/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
The renoprotective effects of SLC5A2/SGLT2 (solute carrier 5 (sodium/glucose cotransporter), member 2) inhibitors have recently been demonstrated in non-diabetic chronic kidney disease (CKD), even without overt albuminuria. However, the mechanism underlying this renoprotection is largely unclear. We investigated the renoprotective mechanisms of the SLC5A2 inhibitor empagliflozin with a focus on ALB (albumin) reabsorption and macroautophagy/autophagy in proximal tubules using wild-type or drug-inducible lrp2/Megalin or atg5 knockout mice with high-fat diet (HFD)-induced obesity or 5/6 nephrectomy that elevated intraglomerular pressure without overt albuminuria. Empagliflozin treatment of HFD-fed mice reduced several hallmarks of lipotoxicity in the proximal tubules, such as phospholipid accumulation in the lysosome, inflammation and fibrosis. Empagliflozin, which decreases intraglomerular pressure, not only reduced the HFD-induced increase in ALB reabsorption via LRP2 in the proximal tubules (i.e. total nephron ALB filtration), as assessed by urinary ALB excretion caused by genetic ablation of Lrp2, but also ameliorated the HFD-induced imbalance in circulating ALB-bound fatty acids. Empagliflozin alleviated the HFD-induced increase in autophagic demand and successfully prevented autophagic stagnation in the proximal tubules. Similarly, empagliflozin decreased ALB exposure and autophagic demand in 5/6 nephrectomized mice. Finally, empagliflozin reduced HFD-induced vulnerability to ischemia-reperfusion injury, whereas LRP2 blockade and atg5 ablation separately diminished this effect. Our findings indicate that empagliflozin reduces ALB exposure and prevents autophagic stagnation in the proximal tubules even without overt albuminuria. Autophagy improvement may be critical for the renoprotection mediated by SLC5A2 inhibition.
Collapse
Affiliation(s)
- Sho Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitsugu Takabatake
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Takahashi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Namba-Hamano
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Matsuda
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Minami
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinsuke Sakai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Yonishi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Nakamura
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shihomi Maeda
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ayumi Matsumoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
5
|
Hupa-Breier KL, Schenk H, Campos-Murguia A, Wellhöner F, Heidrich B, Dywicki J, Hartleben B, Böker C, Mall J, Terkamp C, Wilkens L, Becker F, Rudolph KL, Manns MP, Mederacke YS, Marhenke S, Redeker H, Lieber M, Iordanidis K, Taubert R, Wedemeyer H, Noyan F, Hardtke-Wolenski M, Jaeckel E. Novel translational mouse models of metabolic dysfunction-associated steatotic liver disease comparable to human MASLD with severe obesity. Mol Metab 2025; 93:102104. [PMID: 39855563 PMCID: PMC11815970 DOI: 10.1016/j.molmet.2025.102104] [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: 11/27/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common cause of chronic liver disease, especially in patients with severe obesity. However, current mouse models for MASLD do not reflect the polygenetic background nor the metabolic changes in this population. Therefore, we investigated two novel mouse models of MASLD with a polygenetic background for the metabolic syndrome. METHODS TALLYHO/JngJ mice and NONcNZO10/LtJ mice were fed a high-fat- high-carbohydrate (HF-HC) diet with a surplus of cholesterol diet. A second group of TH mice was additional treated with empagliflozin. RESULTS After sixteen weeks of feeding, both strains developed metabolic syndrome with severe obesity and histological manifestation of steatohepatitis, which was associated with significantly increased intrahepatic CD8+cells, CD4+cells and Tregs, contributing to a significant increase in pro-inflammatory and pro-fibrotic gene activation as well as ER stress and oxidative stress. In comparison with the human transcriptomic signature, we could demonstrate a good metabolic similarity, especially for the TH mouse model. Furthermore, TH mice also developed signs of kidney injury as an extrahepatic comorbidity of MASLD. Additional treatment with empagliflozin in TH mice attenuates hepatic steatosis and improves histological manifestation of MASH. CONCLUSIONS Overall, we have developed two promising new mouse models that are suitable for preclinical studies of MASLD as they recapitulate most of the key features of MASLD.
Collapse
Affiliation(s)
- Katharina L Hupa-Breier
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Heiko Schenk
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Alejandro Campos-Murguia
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Freya Wellhöner
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Benjamin Heidrich
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Janine Dywicki
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Björn Hartleben
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Clara Böker
- Department of General, Visceral, Vascular and Bariatric Surgery, Klinikum Nordstadt, 30167, Hannover, Germany
| | - Julian Mall
- Department of General, Visceral, Vascular and Bariatric Surgery, Klinikum Nordstadt, 30167, Hannover, Germany
| | - Christoph Terkamp
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ludwig Wilkens
- Department of Pathology, Nordstadt Hospital Hannover, 30167, Hannover, Germany
| | - Friedrich Becker
- Research Group on Stem Cell and Metabolism Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Karl Lenhard Rudolph
- Research Group on Stem Cell and Metabolism Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Michael Peter Manns
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Young-Seon Mederacke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Silke Marhenke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hanna Redeker
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Maren Lieber
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Konstantinos Iordanidis
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Fatih Noyan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Ajmera Transplant Centre, Toronto General Hospital, United Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
6
|
Pati P, De Miguel C, Paul JR, Zhang D, Colson J, Allan JM, Edell CJ, Rhoads MK, Dunaway LS, Biswal SN, Zhong Y, Sedaka R, Millender-Swain T, Bailey SM, Gamble KL, Pollock DM, Pollock JS. Time-restricted feeding reduces cardiovascular disease risk in obese mice. JCI Insight 2025; 10:e160257. [PMID: 39812779 PMCID: PMC11949066 DOI: 10.1172/jci.insight.160257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Disrupted feeding and fasting cycles as well as chronic high-fat diet-induced (HFD-induced) obesity are associated with cardiovascular disease risk factors. We designed studies that determined whether 2 weeks of time-restricted feeding (TRF) intervention in mice fed a chronic HFD would reduce cardiovascular disease risk factors. Mice were fed a normal diet (ND; 10% fat) ad libitum or HFD (45% fat) for 18 weeks ad libitum to establish diet-induced obesity. ND or HFD mice were continued on ad libitum diet or subjected to TRF (limiting food availability to 12 hours only during the dark phase) during the final 2 weeks of the feeding protocol. TRF improved whole-body metabolic diurnal rhythms without a change in body weight. HFD mice showed reduced blood pressure dipping compared with ND, which was restored by TRF. Further, TRF reduced aortic wall thickness, decreased aortic stiffness, as well as increased kidney tubular brush border integrity, decreased renal medullary fibrosis, and reduced renal medullary T cell inflammation in HFD mice. These findings indicate that TRF may be an effective intervention for improving vascular and kidney health in a model of established diet-induced obesity.
Collapse
Affiliation(s)
- Paramita Pati
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Jodi R. Paul
- Division of Behavioral Neurobiology, Department of Psychiatry; and
| | - Dingguo Zhang
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Jackson Colson
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - John Miller Allan
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Claudia J. Edell
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Megan K. Rhoads
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Luke S. Dunaway
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Sara N. Biswal
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Yihan Zhong
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Randee Sedaka
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Telisha Millender-Swain
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shannon M. Bailey
- Division of Molecular and Cellular Pathology, Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Karen L. Gamble
- Division of Behavioral Neurobiology, Department of Psychiatry; and
| | - David M. Pollock
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| | - Jennifer S. Pollock
- Section of Cardio-Renal Physiology & Medicine, Division of Nephrology, Department of Medicine
| |
Collapse
|
7
|
Nakamura J, Yamamoto T, Takabatake Y, Namba-Hamano T, Takahashi A, Matsuda J, Minami S, Sakai S, Yonishi H, Maeda S, Matsui S, Kawai H, Matsui I, Yamamuro T, Edahiro R, Takashima S, Takasawa A, Okada Y, Yoshimori T, Ballabio A, Isaka Y. Age-related TFEB downregulation in proximal tubules causes systemic metabolic disorders and occasional apolipoprotein A4-related amyloidosis. JCI Insight 2024; 10:e184451. [PMID: 39699959 PMCID: PMC11948592 DOI: 10.1172/jci.insight.184451] [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: 07/02/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024] Open
Abstract
With the aging of society, the incidence of chronic kidney disease (CKD), a common cause of death, has been increasing. Transcription factor EB (TFEB), the master transcriptional regulator of the autophagy/lysosomal pathway, is regarded as a promising candidate for preventing various age-related diseases. However, whether TFEB in the proximal tubules plays a significant role in elderly patients with CKD remains unknown. First, we found that nuclear TFEB localization in proximal tubular epithelial cells (PTECs) declined with age in both mice and humans. Next, we generated PTEC-specific Tfeb-deficient mice and bred them for up to 24 months. We found that TFEB deficiency in the proximal tubules caused metabolic disorders and occasionally led to apolipoprotein A4 (APOA4) amyloidosis. Supporting this result, we identified markedly decreased nuclear TFEB localization in the proximal tubules of elderly patients with APOA4 amyloidosis. The metabolic disturbances were accompanied by mitochondrial dysfunction due to transcriptional changes involved in fatty acid oxidation and oxidative phosphorylation pathways, as well as decreased mitochondrial clearance. This decreased clearance was reflected by the accumulation of mitochondria-lysosome-related organelles, which depended on lysosomal function. These results shed light on the presumptive mechanisms of APOA4 amyloidosis pathogenesis and provide a therapeutic strategy for CKD-related metabolic disorders and APOA4 amyloidosis.
Collapse
Affiliation(s)
- Jun Nakamura
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitsugu Takabatake
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Namba-Hamano
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Takahashi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jun Matsuda
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Minami
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinsuke Sakai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Yonishi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shihomi Maeda
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sho Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideaki Kawai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tadashi Yamamuro
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Ryuya Edahiro
- Department of Statistical Genetics and
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akira Takasawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yukinori Okada
- Department of Statistical Genetics and
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe) and
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Tamotsu Yoshimori
- Health Promotion System Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Via Pansini 5, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, USA
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
8
|
Giardini E, Moore D, Sadlier D, Godson C, Brennan E. The dual role of lipids in chronic kidney disease: Pathogenic culprits and therapeutic allies. Atherosclerosis 2024; 398:118615. [PMID: 39370307 DOI: 10.1016/j.atherosclerosis.2024.118615] [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: 06/10/2024] [Revised: 08/23/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Chronic kidney disease (CKD) is a significant health burden, with rising incidence and prevalence, attributed in part to increasing obesity and diabetes rates. Lipid accumulation in the kidney parenchyma and chronic, low-grade inflammation are believed to significantly contribute to the development and progression of CKD. The effect of dysregulated kidney lipid metabolism in CKD progression, including altered cholesterol and fatty acid metabolism contribute to glomerular and tubular cell injury through the activation of oxidative stress and inflammatory signalling cascades. In contrast, classes of endogenous specialized pro-resolving lipid mediators (SPMs) have been described that act to limit the inflammatory response and promote the resolution of inflammation. This review highlights our current understanding of how lipids can cause damage within the kidney, and classes of protective lipid metabolites that offer therapeutic benefits.
Collapse
Affiliation(s)
- Elena Giardini
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Dean Moore
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Denise Sadlier
- Mater Misericordiae University Hospital, Eccles Street, Dublin 7, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Benjamin JI, Pati P, Luong T, Liu X, De Miguel C, Pollock JS, Pollock DM. Chronic mistimed feeding results in renal fibrosis and disrupted circadian blood pressure rhythms. Am J Physiol Renal Physiol 2024; 327:F683-F696. [PMID: 39205662 PMCID: PMC11563648 DOI: 10.1152/ajprenal.00047.2024] [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: 02/13/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Circadian disruption is a disturbance in biological timing, which can occur within or between different organizational levels, ranging from molecular rhythms within specific cells to the misalignment of behavioral and environmental cycles. Previous work from our group showed that less than 1 wk of food restriction to the light (inactive) period is sufficient to invert diurnal blood pressure rhythms in mice. However, kidney excretory rhythms and functions remained aligned with the light-dark cycle. Shift workers have an increased risk of cardiovascular disease that may different between sexes and often have irregular mealtimes, making the possibility of mistimed feeding as a potential contributor to the development of kidney disease. Thus, we hypothesized that chronic mistimed food intake would result in adverse cardiorenal effects, with sex differences in severity. Here, we show that chronic circadian disruption via mistimed feeding results in renal fibrosis and aortic stiffness in a sex-dependent manner. Our results indicate the importance of meal timing for the maintenance of blood pressure rhythms and kidney function, particularly in males. Our results also demonstrate that females are better able to acclimate to circadian-related behavioral change. NEW & NOTEWORTHY Circadian disruption through mistimed feeding resulted in nondipping blood pressure, renal fibrosis, and arterial stiffness that were less severe in females versus males. Mice fed exclusively during the daytime maintain their circadian rhythms of locomotor activity regardless of their loss of blood pressure rhythms. Although these mice ate less food, they maintained body weight, suggesting inefficiencies in overall metabolism. These findings demonstrate the importance of maintaining optimal food intake patterns to prevent cardiorenal pathophysiology.
Collapse
Affiliation(s)
- Jazmine I Benjamin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Paramita Pati
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tha Luong
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Xiaofen Liu
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
10
|
Yang Y, Wang J, Shi Y, Cao H, Wei L, Gao L, Liu M. Oxidation enhances the toxicity of polyethylene microplastics to mouse eye: Perspective from in vitro and in vivo. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 360:124633. [PMID: 39074689 DOI: 10.1016/j.envpol.2024.124633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 07/31/2024]
Abstract
Microplastics (MPs) are ubiquitously dispersed in the environment, and undergoing the process of oxidation that alters their physical and chemical properties. Eyes, which directly interface with the external milieu, inevitably encounter MPs. Nonetheless, the ophthalmic toxicity of MPs towards organisms remains unclear. In this study, primary mouse corneal epithelial cells (MCECs), C57BL/6 mice, and CX3CrlGFP/+ mice were utilized to evaluate the toxicity and differences between oxidized low-density polyethylene MPs (modified-MPs) and low-density polyethylene MPs (virgin-MPs) on eyes. The results manifested that virgin-MPs and modified-MPs could be endocytosed by primary MCECs, resulting in a range of cellular damage. Furthermore, they could diminish tear secretion, increase intraocular pressure, and could be internalized into cornea and retina in mice, instigating a series of detrimental reactions. Importantly, modified-MPs exhibited heightened toxicity towards mouse eyes, seemingly due to oxidation enhances the interaction between virgin-MPs/modified-MPs and tissues/cells, and leading to the release of toxic substances increased. In conclusion, our discoveries demonstrate that oxidation exacerbates the harm of virgin-MPs to eyes, and are of great significance for evaluating the risk of MPs to ocular health.
Collapse
Affiliation(s)
- Ying Yang
- School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Ji Wang
- School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Yongpeng Shi
- School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Hanwen Cao
- School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China
| | - Li Wei
- School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China; NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Lan Gao
- School of Life Sciences, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China.
| | - Mingxin Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
11
|
Kanbay M, Copur S, Guldan M, Ozbek L, Hatipoglu A, Covic A, Mallamaci F, Zoccali C. Proximal tubule hypertrophy and hyperfunction: a novel pathophysiological feature in disease states. Clin Kidney J 2024; 17:sfae195. [PMID: 39050867 PMCID: PMC11267238 DOI: 10.1093/ckj/sfae195] [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: 05/12/2024] [Indexed: 07/27/2024] Open
Abstract
The role of proximal tubules (PTs), a major component of the renal tubular structure in the renal cortex, has been examined extensively. Along with its physiological role in the reabsorption of various molecules, including electrolytes, amino acids and monosaccharides, transcellular transport of different hormones and regulation of homeostasis, pathological events affecting PTs may underlie multiple disease states. PT hypertrophy or a hyperfunctioning state, despite being a compensatory mechanism at first in response to various stimuli or alterations at tubular transport proteins, have been shown to be critical pathophysiological events leading to multiple disorders, including diabetes mellitus, obesity, metabolic syndrome and congestive heart failure. Moreover, pharmacotherapeutic agents have primarily targeted PTs, including sodium-glucose cotransporter 2, urate transporters and carbonic anhydrase enzymes. In this narrative review, we focus on the physiological role of PTs in healthy states and the current understanding of the PT pathologies leading to disease states and potential therapeutic targets.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Department of Internal Medicine, Division of Nephrology, Koç University School of Medicine, Istanbul, Turkey
| | - Sidar Copur
- Department of Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Mustafa Guldan
- Department of Internal Medicine, Division of Internal Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Lasin Ozbek
- Department of Internal Medicine, Division of Internal Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Alper Hatipoglu
- Department of Internal Medicine, Division of Internal Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Adrian Covic
- Nephrology, Dialysis and Transplantation, University Grigore T Popa, Iasi, Romania
| | - Francesca Mallamaci
- Nephrology, Dialysis and Transplantation Unit, Grande Ospedale Metropolitano, Reggio Calabria, Italy
- CNR-IFC, Research Unit of Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, Institute of Clinical Physiology, Reggio Calabria, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, NY, USA
- Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale, Grande Ospedale Metropolitano, c/o Nefrologia, Reggio Calabria, Italy
| |
Collapse
|
12
|
Cho S, Jo H, Hwang YJ, Kim C, Jo YH, Yun JW. Potential impact of underlying diseases influencing ADME in nonclinical safety assessment. Food Chem Toxicol 2024; 188:114636. [PMID: 38582343 DOI: 10.1016/j.fct.2024.114636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
Nonclinical studies involve in vitro, in silico, and in vivo experiments to assess the toxicokinetics, toxicology, and safety pharmacology of drugs according to regulatory requirements by a national or international authority. In this review, we summarize the potential effects of various underlying diseases governing the absorption, distribution, metabolism, and excretion (ADME) of drugs to consider the use of animal models of diseases in nonclinical trials. Obesity models showed alterations in hepatic metabolizing enzymes, transporters, and renal pathophysiology, which increase the risk of drug-induced toxicity. Diabetes models displayed changes in hepatic metabolizing enzymes, transporters, and glomerular filtration rates (GFR), leading to variability in drug responses and susceptibility to toxicity. Animal models of advanced age exhibited impairment of drug metabolism and kidney function, thereby reducing the drug-metabolizing capacity and clearance. Along with changes in hepatic metabolic enzymes, animal models of metabolic syndrome-related hypertension showed renal dysfunction, resulting in a reduced GFR and urinary excretion of drugs. Taken together, underlying diseases can induce dysfunction of organs involved in the ADME of drugs, ultimately affecting toxicity. Therefore, the use of animal models of representative underlying diseases in nonclinical toxicity studies can be considered to improve the predictability of drug side effects before clinical trials.
Collapse
Affiliation(s)
- Sumin Cho
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Harin Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeon Jeong Hwang
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Changuk Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yong Hyeon Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
13
|
Raphael H, Klang E, Konen E, Inbar Y, Leibowitz A, Frenkel-Nir Y, Apter S, Grossman E. Obesity Is Associated with Fatty Liver and Fat Changes in the Kidneys in Humans as Assessed by MRI. Nutrients 2024; 16:1387. [PMID: 38732633 PMCID: PMC11085048 DOI: 10.3390/nu16091387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Obesity is associated with metabolic syndrome and fat accumulation in various organs such as the liver and the kidneys. Our goal was to assess, using magnetic resonance imaging (MRI) Dual-Echo phase sequencing, the association between liver and kidney fat deposition and their relation to obesity. METHODS We analyzed MRI scans of individuals who were referred to the Chaim Sheba Medical Center between December 2017 and May 2020 to perform a study for any indication. For each individual, we retrieved from the computerized charts data on sex, and age, weight, height, body mass index (BMI), systolic and diastolic blood pressure (BP), and comorbidities (diabetes mellitus, hypertension, dyslipidemia). RESULTS We screened MRI studies of 399 subjects with a median age of 51 years, 52.4% of whom were women, and a median BMI 24.6 kg/m2. We diagnosed 18% of the participants with fatty liver and 18.6% with fat accumulation in the kidneys (fatty kidneys). Out of the 67 patients with fatty livers, 23 (34.3%) also had fatty kidneys, whereas among the 315 patients without fatty livers, only 48 patients (15.2%) had fatty kidneys (p < 0.01). In comparison to the patients who did not have a fatty liver or fatty kidneys (n = 267), those who had both (n = 23) were more obese, had higher systolic BP, and were more likely to have diabetes mellitus. In comparison to the patients without a fatty liver, those with fatty livers had an adjusted odds ratio of 2.91 (97.5% CI; 1.61-5.25) to have fatty kidneys. In total, 19.6% of the individuals were obese (BMI ≥ 30), and 26.1% had overweight (25 < BMI < 30). The obese and overweight individuals were older and more likely to have diabetes mellitus and hypertension and had higher rates of fatty livers and fatty kidneys. Fat deposition in both the liver and the kidneys was observed in 15.9% of the obese patients, in 8.3% of the overweight patients, and in none of those with normal weight. Obesity was the only risk factor for fatty kidneys and fatty livers, with an adjusted OR of 6.3 (97.5% CI 2.1-18.6). CONCLUSIONS Obesity is a major risk factor for developing a fatty liver and fatty kidneys. Individuals with a fatty liver are more likely to have fatty kidneys. MRI is an accurate modality for diagnosing fatty kidneys. Reviewing MRI scans of any indication should include assessment of fat fractions in the kidneys in addition to that of the liver.
Collapse
Affiliation(s)
- Hadar Raphael
- Arrow Projects for Medical Research Education, The Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel;
- Department of Imaging, The Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel; (E.K.); (E.K.); (Y.I.); (S.A.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 5266202, Israel; (A.L.); (Y.F.-N.)
| | - Eyal Klang
- Department of Imaging, The Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel; (E.K.); (E.K.); (Y.I.); (S.A.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 5266202, Israel; (A.L.); (Y.F.-N.)
| | - Eli Konen
- Department of Imaging, The Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel; (E.K.); (E.K.); (Y.I.); (S.A.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 5266202, Israel; (A.L.); (Y.F.-N.)
| | - Yael Inbar
- Department of Imaging, The Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel; (E.K.); (E.K.); (Y.I.); (S.A.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 5266202, Israel; (A.L.); (Y.F.-N.)
| | - Avshalom Leibowitz
- Faculty of Medicine, Tel Aviv University, Tel Aviv 5266202, Israel; (A.L.); (Y.F.-N.)
- Internal Medicine D the Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel
| | - Yael Frenkel-Nir
- Faculty of Medicine, Tel Aviv University, Tel Aviv 5266202, Israel; (A.L.); (Y.F.-N.)
- Medical Management Department, The Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel
| | - Sara Apter
- Department of Imaging, The Chaim Sheba Medical Center, Tel Hashomer 5266202, Israel; (E.K.); (E.K.); (Y.I.); (S.A.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 5266202, Israel; (A.L.); (Y.F.-N.)
| | - Ehud Grossman
- Adelson Medical School, Ariel University, Ariel 4077625, Israel
| |
Collapse
|
14
|
Leon G, Preston RJS. Peptidylarginine deiminase 4: casting the NET over obesity? J Thromb Haemost 2024; 22:1316-1318. [PMID: 38670685 DOI: 10.1016/j.jtha.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 04/28/2024]
Affiliation(s)
- Gemma Leon
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland.
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons Ireland University of Medicine and Health Sciences, Dublin, Ireland; National Children's Research Centre, Children's Health Ireland Crumlin, Dublin, Ireland
| |
Collapse
|
15
|
Lipovšek S, Dolenšek J, Dariš B, Valladolid-Acebes I, Vajs T, Leitinger G, Stožer A, Skelin Klemen M. Western diet-induced ultrastructural changes in mouse pancreatic acinar cells. Front Cell Dev Biol 2024; 12:1380564. [PMID: 38550379 PMCID: PMC10972872 DOI: 10.3389/fcell.2024.1380564] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 12/18/2024] Open
Abstract
Mouse models of diet-induced type 2 diabetes mellitus provide powerful tools for studying the structural and physiological changes that are related to the disease progression. In this study, diabetic-like glucose dysregulation was induced in mice by feeding them a western diet, and light and transmission electron microscopy were used to study the ultrastructural changes in the pancreatic acinar cells. Acinar necrosis and vacuolization of the cytoplasm were the most prominent features. Furthermore, we observed intracellular and extracellular accumulation of lipid compounds in the form of lipid droplets, structural enlargement of the cisternae of the rough endoplasmic reticulum (RER), and altered mitochondrial morphology, with mitochondria lacking the typical organization of the inner membrane. Last, autophagic structures, i.e., autophagosomes, autolysosomes, and residual bodies, were abundant within the acinar cells of western diet-fed mice, and the autolysosomes contained lipids and material of varying electron density. While diets inducing obesity and type 2 diabetes are clearly associated with structural changes and dysfunction of the endocrine pancreas, we here demonstrate the strong effect of dietary intervention on the structure of acinar cells in the exocrine part of the organ before detectable changes in plasma amylase activity, which may help us better understand the development of non-alcoholic fatty pancreas disease and its association with endo- and exocrine dysfunction.
Collapse
Affiliation(s)
- Saška Lipovšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Barbara Dariš
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tanja Vajs
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Gerd Leitinger
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | |
Collapse
|
16
|
Eritja À, Caus M, Belmonte T, de Gonzalo-Calvo D, García-Carrasco A, Martinez A, Martínez M, Bozic M. microRNA Expression Profile in Obesity-Induced Kidney Disease Driven by High-Fat Diet in Mice. Nutrients 2024; 16:691. [PMID: 38474819 DOI: 10.3390/nu16050691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Obesity is one of the main causes of chronic kidney disease; however, the precise molecular mechanisms leading to the onset of kidney injury and dysfunction in obesity-associated nephropathy remain unclear. The present study aimed to unveil the kidney microRNA (miRNA) expression profile in a model of obesity-induced kidney disease in C57BL/6J mice using next-generation sequencing (NGS) analysis. High-fat diet (HFD)-induced obesity led to notable structural alterations in tubular and glomerular regions of the kidney, increased renal expression of proinflammatory and profibrotic genes, as well as an elevated renal expression of genes involved in cellular lipid metabolism. The miRNA sequencing analysis identified a set of nine miRNAs differentially expressed in the kidney upon HFD feeding, with miR-5099, miR-551b-3p, miR-223-3p, miR-146a-3p and miR-21a-3p showing the most significant differential expression between standard diet (STD) and HFD mice. A validation analysis showed that the expression levels of miR-5099, miR-551b-3p and miR-146a-3p were consistent with NGS results, while Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses revealed that these three validated miRNAs modulated target genes involved in metabolic and adipocytokine pathways, fatty acid and lipid metabolism, and inflammatory, senescence and profibrotic pathways. Our results suggest that differentially expressed miRNAs play pivotal roles in the intricate pathophysiology of obesity-associated kidney disease and could potentially create novel treatment strategies to counteract the deleterious effects of obesity on kidney function.
Collapse
Affiliation(s)
- Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain
| | - Maite Caus
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain
| | - Thalia Belmonte
- Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 28029 Madrid, Spain
| | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, Biomedical Research Institute of Lleida (IRBLleida), 25198 Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Alicia García-Carrasco
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain
| | - Ana Martinez
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain
| | - Montserrat Martínez
- Biostatistics Unit (Biostat), Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain
| | - Milica Bozic
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLleida), 25196 Lleida, Spain
| |
Collapse
|
17
|
Jhuo JY, Tong ZJ, Ku PH, Cheng HW, Wang HT. Acrolein induces mitochondrial dysfunction and insulin resistance in muscle and adipose tissues in vitro and in vivo. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 336:122380. [PMID: 37625774 DOI: 10.1016/j.envpol.2023.122380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/05/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023]
Abstract
Type 2 diabetes mellitus (DM) is a common chronic condition characterized by persistent hyperglycemia and is associated with insulin resistance (IR) in critical glucose-consuming tissues, including skeletal muscle and adipose tissue. Oxidative stress and mitochondrial dysfunction are known to play key roles in IR. Acrolein is a reactive aldehyde found in the diet and environment that is generated as a fatty acid product through the glucose autooxidation process under hyperglycemic conditions. Our previous studies have shown that acrolein impairs insulin sensitivity in normal and diabetic mice, and this effect can be reversed by scavenging acrolein. This study demonstrated that acrolein increased oxidative stress and inhibited mitochondrial respiration in differentiated C2C12 myotubes and differentiated 3T3-L1 adipocytes. As a result, insulin signaling pathways were inhibited, leading to reduced glucose uptake. Treatment with acrolein scavengers, N-acetylcysteine, or carnosine ameliorated mitochondrial dysfunction and inhibited insulin signaling. Additionally, an increase in acrolein expression correlated with mitochondrial dysfunction in the muscle and adipose tissues of diabetic mice. These findings suggest that acrolein-induced mitochondrial dysfunction contributes to IR, and scavenging acrolein is a potential therapeutic approach for treating IR.
Collapse
Affiliation(s)
- Jia-Yu Jhuo
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Zhen-Jie Tong
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Pei-Hsuan Ku
- Department of Life Sciences and the Institute of Genome Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Hsiao-Wei Cheng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC; Doctor Degree Program in Toxicology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
18
|
Zhang Z, Zhang B, Jiang X, Yu Y, Cui Y, Luo H, Wang B. Hyocholic acid retards renal fibrosis by regulating lipid metabolism and inflammatory response in a sheep model. Int Immunopharmacol 2023; 122:110670. [PMID: 37481851 DOI: 10.1016/j.intimp.2023.110670] [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: 05/06/2023] [Revised: 07/15/2023] [Accepted: 07/16/2023] [Indexed: 07/25/2023]
Abstract
The kidneys are vital organs that regulate metabolic homeostasis in the body, filter waste products from the blood, and remove extrahepatic bile acids. We previously found that the dietary supplementation of hyocholic acid alleviated the sheep body lipid deposition and decreased kidney weight. This study evaluated hyocholic acid's (HCA) roles and mechanisms on lipid metabolism and anti-inflammatory function in the kidney under a high-energy diet. Histomicrograph showing the apparent improvement by HCA by attenuating structural damage. The HCA treatment reduced the renal accumulation of cholesterol. Bile acid receptors such as LXR and FXR were activated at the protein level. HCA significantly altered several genes related to immune response (NF-κB, IL-6, and MCP1) and fibrosis (TGF-β, Col1α1, and α-SMA). These significant changes correlated with renal lipid accumulation. The KEGG pathways including non-alcoholic fatty liver disease, insulin resistance, TNF signaling pathway, and Th17 cell differentiation were enriched and NF-κB, IL-6, and TGF-β were identified as the core interconnected genes. This study revealed that HCA plays an efficient role in alleviating kidney lipids accumulation and inflammatory response through crucial genes such as FXR, LXR, HMGCR, NF-κB, IL-6, MCP1, and TGF-β, and expand our understanding of HCA's role in kidney function. In conclusion, HCA mitigated kidney fibrosis, lipid metabolism disorders and immune responses induced by a high-energy diet by regulating a potential LXR/SREBP2/TGF-β-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zeping Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Boyan Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xianzhe Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yue Yu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yimeng Cui
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Bing Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China.
| |
Collapse
|
19
|
DiKun KM, Gudas LJ. Vitamin A and retinoid signaling in the kidneys. Pharmacol Ther 2023; 248:108481. [PMID: 37331524 PMCID: PMC10528136 DOI: 10.1016/j.pharmthera.2023.108481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
Vitamin A (VA, retinol) and its metabolites (commonly called retinoids) are required for the proper development of the kidney during embryogenesis, but retinoids also play key roles in the function and repair of the kidney in adults. Kidneys filter 180-200 liters of blood per day and each kidney contains approximately 1 million nephrons, which are often referred to as the 'functional units' of the kidney. Each nephron consists of a glomerulus and a series of tubules (proximal tubule, loop of Henle, distal tubule, and collecting duct) surrounded by a network of capillaries. VA is stored in the liver and converted to active metabolites, most notably retinoic acid (RA), which acts as an agonist for the retinoic acid receptors ((RARs α, β, and γ) to regulate gene transcription. In this review we discuss some of the actions of retinoids in the kidney after injury. For example, in an ischemia-reperfusion model in mice, injury-associated loss of proximal tubule (PT) differentiation markers occurs, followed by re-expression of these differentiation markers during PT repair. Notably, healthy proximal tubules express ALDH1a2, the enzyme that metabolizes retinaldehyde to RA, but transiently lose ALDH1a2 expression after injury, while nearby myofibroblasts transiently acquire RA-producing capabilities after injury. These results indicate that RA is important for renal tubular injury repair and that compensatory mechanisms exist for the generation of endogenous RA by other cell types upon proximal tubule injury. ALDH1a2 levels also increase in podocytes, epithelial cells of the glomeruli, after injury, and RA promotes podocyte differentiation. We also review the ability of exogenous, pharmacological doses of RA and receptor selective retinoids to treat numerous kidney diseases, including kidney cancer and diabetic kidney disease, and the emerging genetic evidence for the importance of retinoids and their receptors in maintaining or restoring kidney function after injury. In general, RA has a protective effect on the kidney after various types of injuries (eg. ischemia, cytotoxic actions of chemicals, hyperglycemia related to diabetes). As more research into the actions of each of the three RARs in the kidney is carried out, a greater understanding of the actions of vitamin A is likely to lead to new insights into the pathology of kidney disorders and the development of new therapies for kidney diseases.
Collapse
Affiliation(s)
- Krysta M DiKun
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; Department of Urology, Weill Cornell Medicine, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
20
|
Bae J, Lee BW. Significance of Diabetic Kidney Disease Biomarkers in Predicting Metabolic-Associated Fatty Liver Disease. Biomedicines 2023; 11:1928. [PMID: 37509567 PMCID: PMC10377561 DOI: 10.3390/biomedicines11071928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) and diabetic kidney disease (DKD) share various pathophysiological factors, and epidemiological evidence suggests that these two diseases are associated. Albuminuria and the estimated glomerular filtration rate, which are conventional biomarkers of DKD, are reportedly associated with the risk or severity of MAFLD. Recently, novel DKD biomarkers reflecting renal tubular injury have been introduced to complement conventional DKD markers. In this article, we looked at previous studies that showed an association between MAFLD and DKD, and also reviewed the significance of DKD biomarkers as predictive risk factors for MAFLD.
Collapse
Affiliation(s)
- Jaehyun Bae
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Catholic Kwandong University College of Medicine, International St. Mary's Hospital, Incheon 22711, Republic of Korea
| | - Byung-Wan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
21
|
Pereira PG, Alves LL, Ciambarella BT, Rabelo K, Nascimento ALR, Moraes ACN, Bernardi A, Guimarães FV, Carvalho GM, da Silva JFR, de Carvalho JJ. Capybara Oil Improves Renal Pathophysiology and Inflammation in Obese Mice. Nutrients 2023; 15:2925. [PMID: 37447251 DOI: 10.3390/nu15132925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 07/15/2023] Open
Abstract
Obesity is an inflammatory disease associated with secondary diseases such as kidney disease, which can cause lipotoxicity, inflammation and loss of organ function. Polyunsaturated fatty acids act in the production of lipid mediators and have anti-inflammatory characteristics. In this work, the objective was to evaluate renal histopathology in obese mice and the effects of treatment with capybara oil (CO) (5000 mg/kg/day for 4 weeks). Parameters such as body mass, lipid profile, systolic blood pressure, urinary creatinine and protein excretion, structure and ultrastructure of the renal cortex, fibrosis, tissue inflammation and oxidative stress were analyzed. CO treatment in obese mice showed improvement in the lipid profile and reduction in systolic blood pressure levels, in addition to beneficial remodeling of the renal cortex. Our data demonstrated that CO decreased inflammation, oxidative stress and renal fibrosis, as evidenced by quantifying the expression of TNF-α, IL-10, CAT, SOD, α-SMA and TGF-β. Although treatment with CO did not show improvement in renal function, ultrastructural analysis showed that the treatment was effective in restoring podocytes and pedicels, with restructuring of the glomerular filtration barrier. These results demonstrate, for the first time, that treatment with CO is effective in reducing kidney damage, being considered a promising treatment for obesity.
Collapse
Affiliation(s)
- Priscila G Pereira
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| | - Luciana L Alves
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| | - Bianca T Ciambarella
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| | - Kíssila Rabelo
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| | - Ana Lúcia R Nascimento
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| | - Alan Cesar N Moraes
- Electron Microscopy Laboratory of Biology Institute, University of Federal Fluminense, Rio de Janeiro 21040-900, RJ, Brazil
| | - Andressa Bernardi
- Inflammation Laboratory, Fiocruz, Rio de Janeiro 21040-900, RJ, Brazil
| | | | - Gabriela M Carvalho
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| | - Jemima F R da Silva
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| | - Jorge J de Carvalho
- Ultrastructure and Tissue Biology Laboratory, Institute of Biology, Rio de Janeiro State University, Boulevard Vinte e Oito de Setembro, 87 Fundos, 3° Andar Vila Isabel, Rio de Janeiro 20551-030, RJ, Brazil
| |
Collapse
|
22
|
Nyrén R, Scherman H, Axelsson J, Chang CL, Olivecrona G, Ericsson M. Visualizing increased uptake of [18F]FDG and [18F]FTHA in kidneys from obese high-fat diet fed C57BL/6J mice using PET/CT ex vivo. PLoS One 2023; 18:e0281705. [PMID: 36787333 PMCID: PMC9928095 DOI: 10.1371/journal.pone.0281705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
It is known that high-fat diet (HFD) and/or diabetes may influence substrate preferences and energy demands in the heart preceding diabetic cardiomyopathy. They may also induce structural glomerular changes causing diabetic nephropathy. PET/CT has been utilized to examine uptake of energy substrates, and to study metabolic changes or shifts before onset of metabolic disorders. However, conventional PET/CT scanning of organs with relatively low uptake, such as the kidney, in small animals in vivo may render technical difficulties. To address this issue, we developed a PET/CT ex vivo protocol with radiolabeled glucose and fatty acid analouges, [18F]FDG and [18F]FTHA,to study substrate uptake in mouse kidneys. We also aimed to detect a possible energy substrate shift before onset of diabetic nephropathy. The ex vivo protocol reduced interfering background as well as interindividual variances. We found increased uptake of [18F]FDG and [18F]FTHA in kidneys after HFD, compared to kidneys from young mice on standard chow. Levels of kidney triglycerides also increased on HFD. Lipoprotein lipase (LPL) activity, the enzyme responsible for release of fatty acids from circulating lipoproteins, is normally increased in postprandial mice kidneys. After long-term HFD, we found that LPL activity was suppressed, and could therefore not explain the increased levels of stored triglycerides. Suppressed LPL activity was associated with increased expression of angiopoietin-like protein4, an inhibitor of LPL. HFD did not alter the transcriptional control of some common glucose and fatty acid transporters that may mediate uptake of [18F]FDG and [18F]FTHA. Performing PET/CT ex vivo reduced interfering background and interindividual variances. Obesity and insulin resistance induced by HFD increased the uptake of [18F]FDG and [18F]FTHA and triglyceride accumulation in mouse kidneys. Increased levels of [18F]FDG and [18F]FTHA in obese insulin resistant mice could be used clinically as an indicator of poor metabolic control, and a complementary test for incipient diabetic nephropathy.
Collapse
Affiliation(s)
- Rakel Nyrén
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, Umeå, Sweden
- Department of Medical Biosciences/Pathology, Umeå University, Umeå, Sweden
| | - Henrik Scherman
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Jan Axelsson
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Chuchun L. Chang
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Gunilla Olivecrona
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, Umeå, Sweden
| | - Madelene Ericsson
- Department of Medical Biosciences/Physiological Chemistry, Umeå University, Umeå, Sweden
- Umeå Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
23
|
Endoplasmic reticulum stress inhibition ameliorated WFS1 expression alterations and reduced pancreatic islets' insulin secretion induced by high-fat diet in rats. Sci Rep 2023; 13:1860. [PMID: 36725880 PMCID: PMC9892558 DOI: 10.1038/s41598-023-28329-1] [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: 08/14/2022] [Accepted: 01/17/2023] [Indexed: 02/03/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is involved in the development of glucose homeostasis impairment. When ER stress occurs, the unfolded protein response (UPR) is activated to cope with it. One of the UPR components is WFS1 (Wolfram syndrome 1), which plays important roles in ER homeostasis and pancreatic islets glucose-stimulated insulin secretion (GSIS). Accordingly and considering that feeding high-fat food has a major contribution in metabolic disorders, this study aimed to investigate the possible involvement of pancreatic ER stress in glucose metabolism impairment induced by feeding high-fat diet (HFD) in male rats. After weaning, the rats were divided into six groups, and fed on normal diet and HFD for 20 weeks, then 4-phenyl butyric acid (4-PBA, an ER stress inhibitor) was administered. Subsequently, in all groups, after performing glucose tolerance test, the animals were dissected and their pancreases were removed to extract ER, islets isolation and assessment of GSIS. Moreover, the pancreatic ER stress [binding of immunoglobulin protein (BIP) and enhancer-binding protein homologous protein (CHOP)] and oxidative stress [malondialdehyde (MDA), glutathione (GSH) and catalase] biomarkers as well as WFS1 expression level were evaluated. HFD decreased pancreatic WFS1 protein and GSH levels, and enhanced pancreatic catalase activity, MDA content, BIP and CHOP protein and mRNA levels as well as Wfs1 mRNA amount. Accordingly, it increased BIP, CHOP and WFS1 protein levels in the extracted ER of pancreas. In addition, the HFD caused glucose intolerance, and decreased the islets' GSIS and insulin content. However, 4-PBA administration restored the alterations. It seems that, HFD consumption through inducing pancreatic ER stress, altered WFS1 expression levels, reduced the islets' GSIS and insulin content and finally impaired glucose homeostasis.
Collapse
|
24
|
Park YS, Han JH, Park JH, Choi JS, Kim SH, Kim HS. Pyruvate Kinase M2: A New Biomarker for the Early Detection of Diabetes-Induced Nephropathy. Int J Mol Sci 2023; 24:ijms24032683. [PMID: 36769016 PMCID: PMC9916947 DOI: 10.3390/ijms24032683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/25/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Diabetic nephropathy (DN) is a common complication of diabetes. DN progresses to end-stage renal disease, which has a high mortality rate. Current research is focused on identifying non-invasive potential biomarkers in the early stage of DN. We previously indicated that pyruvate kinase M2 (PKM2) is excreted in the urine of rats after cisplatin-induced acute kidney injury (AKI). However, it has not been reported whether PKM2 can be used as a biomarker to diagnose DN. Therefore, we try to compare whether the protein PKM2 can be detected in the urine samples from diabetic patients as shown in the results of DN models. In this study, high-fat diet (HFD)-induced Zucker diabetic fatty (ZDF) rats were used for DN phenotyping. After 19 weeks of receiving a HFD, the DN model's blood glucose, blood urea nitrogen, and serum creatinine levels were significantly increased; severe tubular and glomerular damages were also noted. The following protein-based biomarkers were increased in the urine of these models: kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and PKM2. PKM2 had the earliest detection rate. In the urine samples of patients, PKM2 protein was highly detected in the urine of diabetic patients but was not excreted in the urine of normal subjects. Therefore, PKM2 was selected as the new biomarker for the early diagnosis of DN. Our results reflect current knowledge on the role of PKM2 in DN.
Collapse
Affiliation(s)
- Yeon Su Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Joo Hee Han
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Soo Choi
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seung Hyeon Kim
- St. Mark’s School, 25 Marlboro Rd, Southborough, MA 01772, USA
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Correspondence: ; Tel.:+82-31-290-7789
| |
Collapse
|
25
|
de Souza VR, Lima TPB, Bedê TP, Faria SBA, Alves R, Louzada A, de Moraes BPT, Silva AR, Gonçalves de Albuquerque CF, de Azeredo VB, Teodoro AJ. Murici ( Byrsonima crassifolia (L.) Kunth and verbascifolia (L.)) and Tapereba ( Spondias mombin) Improve Hepatic and Inflammatory Biomarkers in High-Fat-Diet Rats. Foods 2023; 12:foods12020255. [PMID: 36673347 PMCID: PMC9857676 DOI: 10.3390/foods12020255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/27/2022] [Accepted: 12/30/2022] [Indexed: 01/08/2023] Open
Abstract
The present study investigated the effects of murici and tapereba on improving hepatic and inflammatory biomarkers in high-fat-diet rats. Female Wistar rats were divided into five groups (n = 10/group): control (CON), high-fat diet (HF), murici drink + high-fat diet (Mu-HF), tapereba drink + high-fat diet (Tap-HF), and murici and tapereba blend drink + high-fat diet (MT-HF). Drinks were offered daily for 60 days, following which body and liver weights, hepatosomatic indexes, serum parameters, inflammatory profile, and antioxidant activity (DPPH and ORAC) were analyzed. The cell death of hepatic cells was evaluated using flow cytometry. It was observed that weight gain was similar among the groups, while glycemia was lower in the MT-HF group. A high-fat diet increased the concentration of cholesterol total, ALT, IL-1β (in plasma and liver), and TNF-α (in the liver), and this was reduced by treatment with the fruit-based beverages. The other evaluated parameters showed no statistically significant difference. Compared to the CON and HF groups, the groups that received the drinks had higher cellular antioxidant activity and reduced oxidative stress, lipid oxidation, and development of pro-inflammatory cytokines, such as IL-1β. A high-fat diet induced higher cell death in hepatic tissue, which was prevented by the murici, tapereba, and the fruit-blend drinks. The consumption of murici, tapereba, and fruit-blend-based beverages showed beneficial effects on liver metabolism; therefore, they may serve as a nutritional approach for preventing and treating non-alcoholic liver disease.
Collapse
Affiliation(s)
- Vanessa Rosse de Souza
- Food and Nutrition Program, Functional Foods Laboratory, Federal University of the State of Rio de Janeiro, Rio de Janeiro 22290-240, Brazil
| | - Thuane Passos Barbosa Lima
- Food and Nutrition Program, Functional Foods Laboratory, Federal University of the State of Rio de Janeiro, Rio de Janeiro 22290-240, Brazil
| | - Teresa Palmiciano Bedê
- Department of Nutrition and Dietetics, Fluminense Federal University, Rio de Janeiro 24020-140, Brazil
| | | | - Renata Alves
- Membrane Transport Laboratory, State University of Rio de Janeiro, Rio de Janeiro 24020-140, Brazil
| | - Alana Louzada
- Department of Nutrition and Dietetics, Fluminense Federal University, Rio de Janeiro 24020-140, Brazil
| | - Bianca Portugal Tavares de Moraes
- Immunopharmacology Laboratory, Biomedical Institute, Federal University of the State of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Adriana Ribeiro Silva
- Immunopharmacology Laboratory, Oswaldo Cruz, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| | | | - Vilma Blondet de Azeredo
- Department of Nutrition and Dietetics, Fluminense Federal University, Rio de Janeiro 24020-140, Brazil
| | - Anderson Junger Teodoro
- Food and Nutrition Program, Functional Foods Laboratory, Federal University of the State of Rio de Janeiro, Rio de Janeiro 22290-240, Brazil
- Department of Nutrition and Dietetics, Fluminense Federal University, Rio de Janeiro 24020-140, Brazil
- Correspondence:
| |
Collapse
|
26
|
Tukhovskaya EA, Shaykhutdinova ER, Pakhomova IA, Slashcheva GA, Goryacheva NA, Sadovnikova ES, Rasskazova EA, Kazakov VA, Dyachenko IA, Frolova AA, Brovkin AN, Kaluzhsky VE, Beburov MY, Murashev AN. AICAR Improves Outcomes of Metabolic Syndrome and Type 2 Diabetes Induced by High-Fat Diet in C57Bl/6 Male Mice. Int J Mol Sci 2022; 23:ijms232415719. [PMID: 36555360 PMCID: PMC9778872 DOI: 10.3390/ijms232415719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/16/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The aim of the study was to investigate the effect of AMP-activated protein kinase activator 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) on the consequences of metabolic syndrome and type 2 diabetes induced by the consumption of a high-fat diet (HFD) in male C57Bl/6 mice. Additionally, the animals from group 6 were administered Methotrexate (MTX) at a dose of 1 mg/kg in parallel with AICAR, which slows down the metabolism of AICAR. The animals were recorded with signs of metabolic syndrome and type 2 diabetes mellitus by recording their body weights, glucose and insulin levels, and the calculating HOMA-IRs. At the end of the study, at the end of the 13th week, during necropsy, the internal organs were assessed, the masses of the organs were recorded, and special attention was paid to visceral fat, assessing its amount and the mass of the fat surrounding epididymis. The biochemical parameters and histology of the internal organs and tissues were assessed. The animals showed signs of metabolic syndrome and type 2 diabetes, namely, weight gain, hyperglycemia, hyperinsulinemia, an increase in the amount and mass of abdominal fat, and metabolic disorders, all expressed in a pathological change in biochemical parameters and pathological changes in internal organs. The AICAR treatment led to a decrease in body weight, a decrease in the amount and mass of abdominal fat, and an improvement in the pathomorphological picture of internal organs. However, some hepatotoxic effects were observed when the animals, on a received standard diet (STD), were treated with AICAR starting from the first day of the study. The additional administration of MTX, an AICAR metabolic inhibitor, did not improve its efficacy. Thus, AICAR has therapeutic potential for the treatment of metabolic syndrome and type 2 diabetes.
Collapse
Affiliation(s)
- Elena A. Tukhovskaya
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
- Correspondence:
| | - Elvira R. Shaykhutdinova
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Irina A. Pakhomova
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Gulsara A. Slashcheva
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Natalya A. Goryacheva
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Elena S. Sadovnikova
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Ekaterina A. Rasskazova
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Vitaly A. Kazakov
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Igor A. Dyachenko
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| | - Alina A. Frolova
- LLC “OKA-BIOTECH”, Novatorov St., d. 34, bldg. 7, apt. 42, 119421 Moscow, Russia
| | - Alexey N. Brovkin
- LLC “OKA-BIOTECH”, Novatorov St., d. 34, bldg. 7, apt. 42, 119421 Moscow, Russia
| | - Vasiliy E. Kaluzhsky
- LLC “OKA-BIOTECH”, Novatorov St., d. 34, bldg. 7, apt. 42, 119421 Moscow, Russia
| | - Mikhail Yu. Beburov
- LLC “OKA-BIOTECH”, Novatorov St., d. 34, bldg. 7, apt. 42, 119421 Moscow, Russia
| | - Arkady N. Murashev
- Biological Testing Laboratory, Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Prospekt Nauki, 6, Pushchino, 142290 Moscow, Russia
| |
Collapse
|
27
|
Chua D, Low ZS, Cheam GX, Ng AS, Tan NS. Utility of Human Relevant Preclinical Animal Models in Navigating NAFLD to MAFLD Paradigm. Int J Mol Sci 2022; 23:14762. [PMID: 36499091 PMCID: PMC9737809 DOI: 10.3390/ijms232314762] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Fatty liver disease is an emerging contributor to disease burden worldwide. The past decades of work established the heterogeneous nature of non-alcoholic fatty liver disease (NAFLD) etiology and systemic contributions to the pathogenesis of the disease. This called for the proposal of a redefinition in 2020 to that of metabolic dysfunction-associated fatty liver disease (MAFLD) to better reflect the current understanding of the disease. To date, several clinical cohort studies comparing NAFLD and MAFLD hint at the relevancy of the new nomenclature in enriching for patients with more severe hepatic injury and extrahepatic comorbidities. However, the underlying systemic pathogenesis is still not fully understood. Preclinical animal models have been imperative in elucidating key biological mechanisms in various contexts, including intrahepatic disease progression, interorgan crosstalk and systemic dysregulation. Furthermore, they are integral in developing novel therapeutics against MAFLD. However, substantial contextual variabilities exist across different models due to the lack of standardization in several aspects. As such, it is crucial to understand the strengths and weaknesses of existing models to better align them to the human condition. In this review, we consolidate the implications arising from the change in nomenclature and summarize MAFLD pathogenesis. Subsequently, we provide an updated evaluation of existing MAFLD preclinical models in alignment with the new definitions and perspectives to improve their translational relevance.
Collapse
Affiliation(s)
- Damien Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Zun Siong Low
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
| | - Guo Xiang Cheam
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Aik Seng Ng
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 11 Mandalay Road, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
28
|
Tanisha, Venkategowda S, Majumdar M. Amelioration of hyperglycemia and hyperlipidemia in a high-fat diet-fed mice by supplementation of a developed optimized polyherbal formulation. 3 Biotech 2022; 12:251. [PMID: 36060893 PMCID: PMC9428098 DOI: 10.1007/s13205-022-03309-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022] Open
Abstract
This study evaluated in vivo anti-diabetic and anti-obesity activity of a polyherbal formulation's methanolic extract containing an optimized ratio of edible seeds (Salvia hispanica, Chenopodium quinoa, Nelumbo nucifera). Diet-induced obese mice model (C57BL/6) was developed by feeding the mice a high-fat diet for 10 weeks resulting in hyperglycemia and obesity. Different doses (125, 250 and 500 mg/kg of body weight) of formulation were administered orally daily for 6 weeks. Fasting blood glucose and body weight were monitored throughout the study. At the end of the study, serum parameters were analyzed and histological examinations were performed. There was a significant reduction in fasting blood glucose levels and body weight in animal groups receiving polyherbal formulation. Lipid profile was improved as revealed by a reduction in serum triglycerides and total cholesterol. Histological study showed an improvement in liver, kidney and pancreatic sections of treated mice. High-performance thin layer chromatography was performed to identify the phytochemicals responsible for the above-mentioned bioactivities. The results revealed the presence of flavonoid (rutin) in seeds of N.nucifera and in the polyherbal formulation. For the first time, this study demonstrated the anti-diabetic and anti-obesity potential of the optimized formulation. The formulation can be used as a potential therapy for management of diabesity.
Collapse
Affiliation(s)
- Tanisha
- Jain (Deemed-to-be University), School of Sciences, #18/3, 9th Main, Jayanagar, 3rd Block, Bangalore, 560011 India
| | - Sunil Venkategowda
- Jain (Deemed-to-be University), School of Sciences, #18/3, 9th Main, Jayanagar, 3rd Block, Bangalore, 560011 India
| | - Mala Majumdar
- Jain (Deemed-to-be University), School of Sciences, #18/3, 9th Main, Jayanagar, 3rd Block, Bangalore, 560011 India
| |
Collapse
|
29
|
Dahl Salt-Resistant Rat Is Protected against Hypertension during Diet-Induced Obesity. Nutrients 2022; 14:nu14183843. [PMID: 36145220 PMCID: PMC9506364 DOI: 10.3390/nu14183843] [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: 08/12/2022] [Revised: 09/07/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
A high-fat diet (HFD) frequently causes obesity-induced hypertension. Because Dahl salt-resistant rats are protected against hypertension after high-salt or high-fructose intake, it is of interest whether this model also protects against hypertension after diet-induced obesity. We tested the hypothesis that Dahl salt-resistant rat protects against hypertension during diet-induced obesity. Dahl salt-sensitive (SS) and Dahl salt-resistant (SR) rats were fed a HFD (60% fat) or a chow diet (CD; 8% fat) for 12 weeks. We measured blood pressure using the tail-cuff method. The paraffin sections of thoracic perivascular adipose tissue (tPVAT) were stained with hematoxylin/eosin and trichrome. The expression of genes in the tPVAT and kidneys were measured by reverse transcription-quantitative polymerase chain reaction. The HFD induced hypertension in SS (p < 0.01) but not SR rats, although it increased body weight gain (p < 0.05) and tPVAT weight (p < 0.01) in both rats. The HFD did not affect the expression of genes related to any of the adipocyte markers in both rats, although SR rats had reduced beige adipocyte marker Tmem26 levels (p < 0.01) and increased anti-inflammatory cytokine adiponectin (p < 0.05) as compared with SS rat. The HFD did not affect the mRNA expression of contractile factors in the tPVAT of SS and SR rats. SR rats are protected against hypertension during diet-induced obesity. This result implies that the genetic trait determining salt sensitivity may also determine fructose and fat sensitivity and that it is associated with the prevention of hypertension.
Collapse
|
30
|
Control of Obesity, Blood Glucose, and Blood Lipid with Olax imbricata Roxb. Root Extract in High-Fat Diet-Induced Obese Mice. J Toxicol 2022; 2022:7781723. [PMID: 36091101 PMCID: PMC9463018 DOI: 10.1155/2022/7781723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/23/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022] Open
Abstract
Mice were used in in vivo experiments to evaluate the effects of doses of n-hexane extract (from 100 to 1,300 mg/kg body weight/day) on the ability to control obesity, blood glucose, and blood lipid. In this study, body weight gain, caloric intake, glucose tolerance, blood lipid, histopathological study, and locomotion activity were examined. Furthermore, this study evaluated the lethality of the extract in extremely high doses in the tested mice. After 3 months of use with an extremely high dose of 5,000 mg/kg body weight/day (equivalent to 350 g/day for a 70 kg person), no animals with abnormal conditions or death were observed. This initially demonstrated the safety of the extract. In addition, after 6 weeks of testing on high-fat diet-induced obese mice, n-hexane extract at a dose of 500 mg/kg body weight/day (equivalent to 35 g/day for a 70 kg person) demonstrated a positive effect on the ability to control obesity, blood glucose, and blood lipid through the results of body weight, blood lipids, glucose tolerance, and histopathology (white fat, liver, and kidney tissues). In this study, n-hexane extract from the roots of Duong-dau tree has proven to be strongly biologically active in preventing and supporting the treatment of diseases related to overweight and obesity, helping to control blood glucose levels thereby reducing the risk of type 2 diabetes.
Collapse
|
31
|
Nazari S, Moosavi SMS. Temporal patterns of alterations in obesity index, lipid profile, renal function and blood pressure during the development of hypertension in male, but not female, rats fed a moderately high-fat diet. Arch Physiol Biochem 2022; 128:897-909. [PMID: 32195603 DOI: 10.1080/13813455.2020.1739713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Male Sprague-Dawley rats consuming a moderately high-fat (MHF)-diet diverge into obesity-prone (OP) with hypertension and obesity-resistant. OBJECTIVES To study the temporal inter-relationships between body-weight, obesity-index, plasma lipid-profile, renal functional parameters and systolic-pressure alterations during 10-weeks feeding MHF or normal diet to male and female rats. METHODS Body-weight, obesity-index and systolic-pressure were measured weekly, while metabolic-cage and blood-sampling protocols were performed every other week. After 10-weeks, renal excretory responses to acute salt-loading and renal autoregulation were examined. RESULTS The male-OP group had progressively increased body-weight, plasma-triglyceride and systolic-pressure from Weeks 2, 4 and 5, respectively, lower renal sodium-excretion at weeks 4-8 and finally, delayed excretory response to salt-loading and rightward and downward shifts in renal autoregulatory curves compared to all other groups. CONCLUSION Feeding the MHF-diet in male-OP rats led to a greater weight-gain and adiposity followed by the development of atherogenic-hyperlipidaemia and persistently impaired pressure-natriuresis to induce hypertension.
Collapse
Affiliation(s)
- Somayeh Nazari
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mostafa Shid Moosavi
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
Locatelli M, Macconi D, Corna D, Cerullo D, Rottoli D, Remuzzi G, Benigni A, Zoja C. Sirtuin 3 Deficiency Aggravates Kidney Disease in Response to High-Fat Diet through Lipotoxicity-Induced Mitochondrial Damage. Int J Mol Sci 2022; 23:ijms23158345. [PMID: 35955472 PMCID: PMC9368634 DOI: 10.3390/ijms23158345] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 02/05/2023] Open
Abstract
Sirtuin 3 (SIRT3) is the primary mitochondrial deacetylase that controls the antioxidant pathway and energy metabolism. We previously found that renal Sirt3 expression and activity were reduced in mice with type 2 diabetic nephropathy associated with oxidative stress and mitochondrial abnormalities and that a specific SIRT3 activator improved renal damage. SIRT3 is modulated by diet, and to assess whether Sirt3 deficiency aggravates mitochondrial damage and accelerates kidney disease in response to nutrient overloads, wild-type (WT) and Sirt3−/− mice were fed a high-fat-diet (HFD) or standard diet for 8 months. Sirt3−/− mice on HFD exhibited earlier and more severe albuminuria compared to WT mice, accompanied by podocyte dysfunction and glomerular capillary rarefaction. Mesangial matrix expansion, tubular vacuolization and inflammation, associated with enhanced lipid accumulation, were more evident in Sirt3−/− mice. After HFD, kidneys from Sirt3−/− mice showed more oxidative stress than WT mice, mitochondria ultrastructural damage in tubular cells, and a reduction in mitochondrial mass and energy production. Our data demonstrate that Sirt3 deficiency renders mice more prone to developing oxidative stress and mitochondrial abnormalities in response to HFD, resulting in more severe kidney diseases, and this suggests that mitochondria protection may be a method to prevent HFD-induced renal injury.
Collapse
|
33
|
Narciso L, Martinelli A, Torriani F, Frassanito P, Bernardini R, Chiarotti F, Marianelli C. Natural Mineral Waters and Metabolic Syndrome: Insights From Obese Male and Female C57BL/6 Mice on Caloric Restriction. Front Nutr 2022; 9:886078. [PMID: 35685873 PMCID: PMC9172593 DOI: 10.3389/fnut.2022.886078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/04/2022] [Indexed: 01/01/2023] Open
Abstract
Metabolic syndrome (MetS) represents one of the greatest challenges to public health given its serious consequences on cardiovascular diseases and type 2 diabetes. A carbohydrate-restricted, low-fat diet is the current therapy for MetS. Natural mineral waters (NMWs) are known to exert beneficial effects on human health. Our primary objective was to shed light on the potential therapeutic properties of NMWs in MetS. A total of 125 C57BL/6 male and female mice were included in the study. Of these, 10 were left untreated. They were fed a standard diet with tap water throughout the study period, and stayed healthy. The remaining 115 mice were initially fed a high-calorie diet (HCD) consisting of a high-fat feed (60% of energy from fat) with 10% fructose in tap water, served ad libitum over a period of 4 months to induce MetS (the MetS induction phase). Mice were then randomly divided into six treatment groups and a control group, all of which received a low-calorie diet (LCD), but with a different kind of drinking water, for 2 months (the treatment phase). Five groups were each treated with a different kind of NMW, one group by alternating the five NMWs, and one group – the control group – was given tap water. Body weight and blood biochemistry were monitored over the 6-month trial. After 4 months, male and female mice on HCD developed obesity, hypercholesterolaemia and hyperglycaemia, although gains in body weight, total cholesterol, and blood glucose in males were greater than those observed in females (P < 0.0001). When combined with an LCD, the NMWs rich in sulphate, magnesium and bicarbonate, and the minimally mineralised one were the most effective in reducing the blood levels of total cholesterol, high-density lipoprotein (HDL) cholesterol, and glucose. Sex differences emerged during both the MetS induction phase and the treatment phase. These results suggest that NMWs rich in specific macronutrients, such as bicarbonate, sulphate and magnesium, and minimally mineralised water, in combination with an LCD, may contribute to controlling blood lipid and glucose levels in subjects with MetS. Further studies are needed to confirm these results and to extend them to humans.
Collapse
Affiliation(s)
- Laura Narciso
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Andrea Martinelli
- Centre for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, Rome, Italy
| | - Flavio Torriani
- Centre for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, Rome, Italy
| | - Paolo Frassanito
- Centre for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta Bernardini
- Interdepartmental Center for Comparative Medicine, Alternative Techniques and Aquaculture, University of Rome “Tor Vergata,”Rome, Italy
| | - Flavia Chiarotti
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Cinzia Marianelli
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
- *Correspondence: Cinzia Marianelli,
| |
Collapse
|
34
|
Hamada S, Takata T, Yamada K, Yamamoto M, Mae Y, Iyama T, Ikeda S, Kanda T, Sugihara T, Isomoto H. Steatosis is involved in the progression of kidney disease in a high-fat-diet-induced non-alcoholic steatohepatitis mouse model. PLoS One 2022; 17:e0265461. [PMID: 35294499 PMCID: PMC8926260 DOI: 10.1371/journal.pone.0265461] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/02/2022] [Indexed: 01/03/2023] Open
Abstract
Chronic kidney disease (CKD) and non-alcoholic steatohepatitis (NASH) are major health issues associated with the metabolic syndrome. Although NASH is a known risk factor of CKD, the mechanisms linking these two diseases remain poorly understood. We aimed to investigate alterations in the kidney complicated with dyslipidemia in an established NASH mouse model. Male C57BL6/J mice were fed with control diet or high-fat diet (HFD), containing 40% fat, 22% fructose, and 2% cholesterol for 16 weeks. Metabolic characteristics, histological changes in the kidney, endoplasmic reticulum (ER) stress, apoptosis, and fibrosis were evaluated by histological analysis, immunoblotting, and quantitative reverse transcription-polymerase chain reaction. Levels of serum aspartate aminotransferase, alanine aminotransferase, alkali-phosphatase, total cholesterol, and urinary albumin were significantly higher in mice fed with HFD. Remarkable steatosis, glomerular hypertrophy, and interstitial fibrosis were also shown in in the kidney by leveraging HFD. Furthermore, HFD increased the mRNA expression levels of Casp3, Tgfb1, and Nfe2l2 and the protein level of BiP. We observed the early changes of CKD and speculate that the underlying mechanisms that link CKD and NASH are the induction of ER stress and apoptosis. Further, we observed the activation of Nfe2l2 in the steatosis-induced CKD mouse model. This NASH model holds implications in investigating the mechanisms linking dyslipidemia and CKD.
Collapse
Affiliation(s)
- Shintaro Hamada
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tomoaki Takata
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
- * E-mail:
| | - Kentaro Yamada
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Marie Yamamoto
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yukari Mae
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Takuji Iyama
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Suguru Ikeda
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tsutomu Kanda
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Takaaki Sugihara
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
35
|
Zeitler EM, Jennette JC, Flythe JE, Falk RJ, Poulton JS. High-calorie diet results in reversible obesity-related glomerulopathy in adult zebrafish regardless of dietary fat. Am J Physiol Renal Physiol 2022; 322:F527-F539. [PMID: 35224994 PMCID: PMC8977181 DOI: 10.1152/ajprenal.00018.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/10/2022] [Accepted: 02/22/2022] [Indexed: 11/22/2022] Open
Abstract
Obesity is a risk factor for the development of kidney disease. The role of diet in this association remains undetermined, in part due to practical limitations in studying nutrition in humans. In particular, the relative importance of calorie excess versus dietary macronutrient content is poorly understood. For example, it is unknown if calorie restriction modulates obesity-related kidney pathology. To study the effects of diet-induced obesity in a novel animal model, we treated zebrafish for 8 wk with diets varied in both calorie and fat content. Kidneys were evaluated by light and electron microscopy. We evaluated glomerular filtration barrier function using a dextran permeability assay. We assessed the effect of diet on podocyte sensitivity to injury using an inducible podocyte injury model. We then tested the effect of calorie restriction on the defects caused by diet-induced obesity. Fish fed a high-calorie diet developed glomerulomegaly (mean: 1,211 vs. 1,010 µm2 in controls, P = 0.007), lower podocyte density, foot process effacement, glomerular basement membrane thickening, tubular enlargement (mean: 1,038 vs. 717 µm2 in controls, P < 0.0001), and ectopic lipid deposition. Glomerular filtration barrier dysfunction and increased susceptibility to podocyte injury were observed with high-calorie feeding regardless of dietary fat content. These pathological changes resolved with 4 wk of calorie restriction. Our findings suggest that calorie excess rather than dietary fat drives obesity-related kidney dysfunction and that inadequate podocyte proliferation in response to glomerular enlargement may cause podocyte dysfunction. We also demonstrate the value of zebrafish as a novel model for studying diet in obesity-related kidney disease.NEW & NOTEWORTHY Obesity is a risk factor for kidney disease. The role of diet in this association is difficult to study in humans. In this study, zebrafish fed a high-calorie diet, regardless of fat macronutrient composition, developed glomerulomegaly, foot process effacement, and filtration barrier dysfunction, recapitulating the changes seen in humans with obesity. Calorie restriction reversed the changes. This work suggests that macronutrient composition may be less important than total calories in the development of obesity-related kidney disease.
Collapse
Affiliation(s)
- Evan M Zeitler
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - J Charles Jennette
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Nephropathology Division, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jennifer E Flythe
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ronald J Falk
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - John S Poulton
- Division of Nephrology and Hypertension, Department of Medicine, UNC Kidney Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
36
|
Monu SR, Wang H, Potter DL, Liao TD, Ortiz PA. Decreased tubuloglomerular feedback response in high-fat diet-induced obesity. Am J Physiol Renal Physiol 2022; 322:F429-F436. [PMID: 35224993 PMCID: PMC9169969 DOI: 10.1152/ajprenal.00307.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 12/17/2022] Open
Abstract
Obesity increases the risk of renal damage, but the mechanisms are not clear. Normally, kidneys autoregulate to keep the glomerular capillary pressure (PGC), renal blood flow, and glomerular filtration rate in a steady state. However, in obesity, higher PGC, renal blood flow, and glomerular filtration rate are noted. Together, these may lead to glomerular damage. PGC is controlled mainly by afferent arteriole resistance, which, in turn, is regulated by tubuloglomerular feedback (TGF), a vasoconstrictor mechanism. High fat-induced obesity causes renal damage, and this may be related to increased PGC. However, there are no studies as to whether high-fat diet (HFD)-induced obesity affects TGF. We hypothesized that TGF would be attenuated in obesity caused by HFD feeding (60% fat) in Sprague-Dawley rats. Sprague-Dawley rats fed a normal-fat diet (NFD; 12% fat) served as the control. We studied 4 and 16 wk of HFD feeding using in vivo renal micropuncture of individual rat nephrons. We did not observe significant differences in body weight, TGF response, and mean arterial pressure at 4 wk of HFD feeding, but after 16 wk of HFD, rats were heavier and hypertensive. The maximal TGF response was smaller in HFD-fed rats than in NFD-fed rats, indicating an attenuation of TGF in HFD-induced obesity. Baseline PGC was higher in HFD-fed rats than in NFD-fed rats and was associated with higher glomerulosclerosis. We conclude that attenuated TGF and higher PGC along with hypertension in HFD-fed obese Sprague-Dawley rats could explain the higher propensity of glomerular damage observed in obesity.NEW & NOTEWORTHY Reduced tubuloglomerular feedback, higher glomerular capillary pressure, and hypertension in combination may explain the higher glomerular damage observed in high-fat diet-induced obesity.
Collapse
Affiliation(s)
- Sumit R Monu
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Hong Wang
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - D'Anna L Potter
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Tang-Dong Liao
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Pablo A Ortiz
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| |
Collapse
|
37
|
Dozio E, Maffioli E, Vianello E, Nonnis S, Grassi Scalvini F, Spatola L, Roccabianca P, Tedeschi G, Corsi Romanelli MM. A Wide-Proteome Analysis to Identify Molecular Pathways Involved in Kidney Response to High-Fat Diet in Mice. Int J Mol Sci 2022; 23:ijms23073809. [PMID: 35409168 PMCID: PMC8999052 DOI: 10.3390/ijms23073809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022] Open
Abstract
The etiopathogenesis of obesity-related chronic kidney disease (CKD) is still scarcely understood. To this aim, we assessed the effect of high-fat diet (HF) on molecular pathways leading to organ damage, steatosis, and fibrosis. Six-week-old male C57BL/6N mice were fed HF diet or normal chow for 20 weeks. Kidneys were collected for genomic, proteomic, histological studies, and lipid quantification. The main findings were as follows: (1) HF diet activated specific pathways leading to fibrosis and increased fatty acid metabolism; (2) HF diet promoted a metabolic shift of lipid metabolism from peroxisomes to mitochondria; (3) no signs of lipid accumulation and/or fibrosis were observed, histologically; (4) the early signs of kidney damage seemed to be related to changes in membrane protein expression; (5) the proto-oncogene MYC was one of the upstream transcriptional regulators of changes occurring in protein expression. These results demonstrated the potential usefulness of specific selected molecules as early markers of renal injury in HF, while histomorphological changes become visible later in obesity-related CDK. The integration of these information with data from biological fluids could help the identification of biomarkers useful for the early detection and prevention of tissue damage in clinical practice.
Collapse
Affiliation(s)
- Elena Dozio
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
| | - Elisa Maffioli
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
| | - Elena Vianello
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
- Correspondence: ; Tel.: +39-02-50315342
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
- CRC “Innovation for Well-Being and Environment” (I-WE), Università degli Studi di Milano, 29133 Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
| | - Leonardo Spatola
- Division of Nephrology, Dialysis and Renal Transplantation, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Paola Roccabianca
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
- CRC “Innovation for Well-Being and Environment” (I-WE), Università degli Studi di Milano, 29133 Milan, Italy
| | - Massimiliano Marco Corsi Romanelli
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
- Service of Laboratory Medicine1-Clinical Pathology, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| |
Collapse
|
38
|
Pitere RR, van Heerden MB, Pepper MS, Ambele MA. Slc7a8 Deletion Is Protective against Diet-Induced Obesity and Attenuates Lipid Accumulation in Multiple Organs. BIOLOGY 2022; 11:311. [PMID: 35205177 PMCID: PMC8869389 DOI: 10.3390/biology11020311] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 11/22/2022]
Abstract
Adipogenesis, through adipocyte hyperplasia and/or hypertrophy, leads to increased adiposity, giving rise to obesity. A genome-wide transcriptome analysis of in vitro adipogenesis in human adipose-derived stromal/stem cells identified SLC7A8 (Solute Carrier Family 7 Member 8) as a potential novel mediator. The current study has investigated the role of SLC7A8 in adipose tissue biology using a mouse model of diet-induced obesity. slc7a8 knockout (KO) and wildtype (WT) C57BL/6J mice were fed either a control diet (CD) or a high-fat diet (HFD) for 14 weeks. On the HFD, both WT and KO mice (WTHFD and KOHFD) gained significantly more weight than their CD counterparts. However, KOHFD gained significantly less weight than WTHFD. KOHFD had significantly reduced levels of glucose intolerance compared with those observed in WTHFD. KOHFD also had significantly reduced adipocyte mass and hypertrophy in inguinal, mesenteric, perigonadal, and brown adipose depots, with a corresponding decrease in macrophage infiltration. Additionally, KOHFD had decreased lipid accumulation in the liver, heart, gastrocnemius muscle, lung, and kidney. This study demonstrates that targeting slc7a8 protects against diet-induced obesity by reducing lipid accumulation in multiple organs and suggests that if targeted, has the potential to mitigate the development of obesity-associated comorbidities.
Collapse
Affiliation(s)
- Reabetswe R. Pitere
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
| | - Marlene B. van Heerden
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
| | - Melvin A. Ambele
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (R.R.P.); (M.S.P.)
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| |
Collapse
|
39
|
Zhang W, Rong G, Gu J, Fan C, Guo T, Jiang T, Deng W, Xie J, Su Z, Yu Q, Mai J, Zheng R, Chen X, Tang X, Zhang J. Nicotinamide N-methyltransferase ameliorates renal fibrosis by its metabolite 1-methylnicotinamide inhibiting the TGF-β1/Smad3 pathway. FASEB J 2022; 36:e22084. [PMID: 35107844 DOI: 10.1096/fj.202100913rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD), a disease involving damage to the kidney structure and function, is a global public health problem. Tubulointerstitial fibrosis (TIF) is both an inevitable pathological change in individuals with CKD and a driving force in the progression of renal fibrosis. Nicotinamide N-methyltransferase (NNMT) and its metabolite 1-methylnicotinamide (MNAM) have been shown to protect against lipotoxicity-induced kidney tubular injury. However, the biological roles of NNMT and MNAM in regulating TIF remain elusive. This study aimed to investigate the protective effect of NNMT and MNAM on TIF and the mechanisms involved. We explored the functions and mechanisms of NNMT and MNAM in TIF, as well as the interaction between NNMT and MNAM, using unilateral ureteral obstruction (UUO) mice and cultured mouse tubular epithelial cells (mTECs) stimulated with transforming growth factor-β1 (TGF-β1). Several important findings were obtained as follows: (1) NNMT expression was upregulated in the kidneys of UUO mice and TGF-β1-induced mTECs, and this upregulation was proposed to be a protective compensatory response to TIF. (2) MNAM was a potentially effective antifibrotic and anti-inflammatory medication in UUO mice. (3) The antifibrotic effect of NNMT overexpression was exerted by increasing the concentration of MNAM. (4) The renoprotective role of MNAM depended on the selective blockade of the interaction of Smad3 with TGFβ receptor I. Overall, our study shows that NNMT is involved in the development and progression of CKD and that its metabolite MNAM may be a novel inhibitor of the TGF-β1/Smad3 pathway with great therapeutic potential for CKD.
Collapse
Affiliation(s)
- Wenying Zhang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Nephrology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guang Rong
- Department of Nephrology, SSL Central Hospital of Dongguan City, Dongguan, China
| | - Jinge Gu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cuiling Fan
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Guo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tingting Jiang
- Department of Nephrology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Weiqian Deng
- Department of Nephrology, Fifth Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jiayu Xie
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, China
| | - Zhihua Su
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qimin Yu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyi Mai
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Rinan Zheng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xingling Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xun Tang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Zhang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
40
|
Guo X, Sunil C, Qian G. Obesity and the Development of Lung Fibrosis. Front Pharmacol 2022; 12:812166. [PMID: 35082682 PMCID: PMC8784552 DOI: 10.3389/fphar.2021.812166] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Obesity is an epidemic worldwide and the obese people suffer from a range of respiratory complications including fibrotic changes in the lung. The influence of obesity on the lung is multi-factorial, which is related to both mechanical injury and various inflammatory mediators produced by excessive adipose tissues, and infiltrated immune cells. Adiposity causes increased production of inflammatory mediators, for example, cytokines, chemokines, and adipokines, both locally and in the systemic circulation, thereby rendering susceptibility to respiratory diseases, and altered responses. Lung fibrosis is closely related to chronic inflammation in the lung. Current data suggest a link between lung fibrosis and diet-induced obesity, although the mechanism remains incomplete understood. This review summarizes findings on the association of lung fibrosis with obesity, highlights the role of several critical inflammatory mediators (e.g., TNF-α, TGF-β, and MCP-1) in obesity related lung fibrosis and the implication of obesity in the outcomes of idiopathic pulmonary fibrosis patients.
Collapse
Affiliation(s)
- Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Christudas Sunil
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| |
Collapse
|
41
|
Endothelial ADAM17 Expression in the Progression of Kidney Injury in an Obese Mouse Model of Pre-Diabetes. Int J Mol Sci 2021; 23:ijms23010221. [PMID: 35008648 PMCID: PMC8745741 DOI: 10.3390/ijms23010221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Disintegrin and metalloproteinase domain 17 (ADAM17) activates inflammatory and fibrotic processes through the shedding of various molecules such as Tumor Necrosis Factor-α (TNF-α) or Transforming Growht Factor-α (TGF-α). There is a well-recognised link between TNF-α, obesity, inflammation, and diabetes. In physiological situations, ADAM17 is expressed mainly in the distal tubular cell while, in renal damage, its expression increases throughout the kidney including the endothelium. The aim of this study was to characterize, for the first time, an experimental mouse model fed a high-fat diet (HFD) with a specific deletion of Adam17 in endothelial cells and to analyse the effects on different renal structures. Endothelial Adam17 knockout male mice and their controls were fed a high-fat diet, to induce obesity, or standard rodent chow, for 22 weeks. Glucose tolerance, urinary albumin-to-creatinine ratio, renal histology, macrophage infiltration, and galectin-3 levels were evaluated. Results showed that obese mice presented higher blood glucose levels, dysregulated glucose homeostasis, and higher body weight compared to control mice. In addition, obese wild-type mice presented an increased albumin-to-creatinine ratio; greater glomerular size and mesangial matrix expansion; and tubular fibrosis with increased galectin-3 expression. Adam17 deletion decreased the albumin-to-creatinine ratio, glomerular mesangial index, and tubular galectin-3 expression. Moreover, macrophage infiltration in the glomeruli of obese Adam17 knockout mice was reduced as compared to obese wild-type mice. In conclusion, the expression of ADAM17 in endothelial cells impacted renal inflammation, modulating the renal function and histology in an obese pre-diabetic mouse model.
Collapse
|
42
|
Kotsis V, Martinez F, Trakatelli C, Redon J. Impact of Obesity in Kidney Diseases. Nutrients 2021; 13:nu13124482. [PMID: 34960033 PMCID: PMC8703549 DOI: 10.3390/nu13124482] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 01/14/2023] Open
Abstract
The clinical consequences of obesity on the kidneys, with or without metabolic abnormalities, involve both renal function and structures. The mechanisms linking obesity and renal damage are well understood, including several effector mechanisms with interconnected pathways. Higher prevalence of urinary albumin excretion, sub-nephrotic syndrome, nephrolithiasis, increased risk of developing CKD, and progression to ESKD have been identified as being associated with obesity and having a relevant clinical impact. Moreover, renal replacement therapy and kidney transplantation are also influenced by obesity. Losing weight is key in limiting the impact that obesity produces on the kidneys by reducing albuminuria/proteinuria, declining rate of eGFR deterioration, delaying the development of CKD and ESKD, and improving the outcome of a renal transplant. Weight reduction may also contribute to appropriate control of cardiometabolic risk factors such as hypertension, metabolic syndrome, diabetes, and dyslipidemia which may be protective not only in renal damage but also cardiovascular disease. Lifestyle changes, some drugs, and bariatric surgery have demonstrated the benefits.
Collapse
Affiliation(s)
- Vasilios Kotsis
- 3rd Department of Internal Medicine, Hypertension-24h ABPM ESH Center of Excellence, Papageorgiou Hospital, Aristotle University of Thessaloniki, 564 29 Pavlos Melas, Greece; (V.K.); (C.T.)
| | - Fernando Martinez
- Internal Medicine Hospital Clínico de Valencia, 46010 Valencia, Spain;
| | - Christina Trakatelli
- 3rd Department of Internal Medicine, Hypertension-24h ABPM ESH Center of Excellence, Papageorgiou Hospital, Aristotle University of Thessaloniki, 564 29 Pavlos Melas, Greece; (V.K.); (C.T.)
| | - Josep Redon
- Internal Medicine Hospital Clínico de Valencia, 46010 Valencia, Spain;
- Cardiovascular and Renal Research Group, INCLIVA Research Institute, University of Valencia, 46010 Valencia, Spain
- CIBERObn Carlos III Institute, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
43
|
Palau V, Villanueva S, Jarrín J, Benito D, Márquez E, Rodríguez E, Soler MJ, Oliveras A, Gimeno J, Sans L, Crespo M, Pascual J, Barrios C, Riera M. Redefining the Role of ADAM17 in Renal Proximal Tubular Cells and Its Implications in an Obese Mouse Model of Pre-Diabetes. Int J Mol Sci 2021; 22:ijms222313093. [PMID: 34884897 PMCID: PMC8657896 DOI: 10.3390/ijms222313093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 12/14/2022] Open
Abstract
Acute and chronic kidney lesions induce an increase in A Disintegrin And Metalloproteinase domain 17 (ADAM17) that cleaves several transmembrane proteins related to inflammatory and fibrotic pathways. Our group has demonstrated that renal ADAM17 is upregulated in diabetic mice and its inhibition decreases renal inflammation and fibrosis. The purpose of the present study was to analyze how Adam17 deletion in proximal tubules affects different renal structures in an obese mice model. Tubular Adam17 knockout male mice and their controls were fed a high-fat diet (HFD) for 22 weeks. Glucose tolerance, urinary albumin-to-creatinine ratio, renal histology, and pro-inflammatory and pro-fibrotic markers were evaluated. Results showed that wild-type mice fed an HFD became obese with glucose intolerance and renal histological alterations mimicking a pre-diabetic condition; consequently, greater glomerular size and mesangial expansion were observed. Adam17 tubular deletion improved glucose tolerance and protected animals against glomerular injury and prevented podocyte loss in HFD mice. In addition, HFD mice showed more glomerular macrophages and collagen accumulation, which was prevented by Adam17 deletion. Galectin-3 expression increased in the proximal tubules and glomeruli of HFD mice and ameliorated with Adam17 deletion. In conclusion, Adam17 in proximal tubules influences glucose tolerance and participates in the kidney injury in an obese pre-diabetic murine model. The role of ADAM17 in the tubule impacts on glomerular inflammation and fibrosis.
Collapse
Affiliation(s)
- Vanesa Palau
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Sofia Villanueva
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Josué Jarrín
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - David Benito
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Eva Márquez
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Eva Rodríguez
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - María José Soler
- Nephrology Research Group, Vall d’Hebron Research Institute (VHIR), Nephrology Department, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Anna Oliveras
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Javier Gimeno
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain;
| | - Laia Sans
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
| | - Clara Barrios
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
- Correspondence: (C.B.); (M.R.); Tel.: +34-65-004-2149 (C.B.); +34-93-316-0626 (M.R.)
| | - Marta Riera
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), 08003 Barcelona, Spain; (V.P.); (S.V.); (J.J.); (D.B.); (E.M.); (E.R.); (A.O.); (L.S.); (M.C.); (J.P.)
- Correspondence: (C.B.); (M.R.); Tel.: +34-65-004-2149 (C.B.); +34-93-316-0626 (M.R.)
| |
Collapse
|
44
|
Castro BBA, Foresto-Neto O, Saraiva-Camara NO, Sanders-Pinheiro H. Renal lipotoxicity: Insights from experimental models. Clin Exp Pharmacol Physiol 2021; 48:1579-1588. [PMID: 34314523 DOI: 10.1111/1440-1681.13556] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 07/13/2021] [Accepted: 07/23/2021] [Indexed: 11/29/2022]
Abstract
In recent decades, there has been a progressive increase in the prevalence of obesity and chronic kidney disease. Renal lipotoxicity has been associated with obesity. Although lipids play fundamental physiological roles, the accumulation of lipids in kidney cells may cause dysfunction and/or renal fibrosis. Adipose tissue that exceeds their lipid storage capacity begins to release triglycerides into the bloodstream that can get stored in several organs, including the kidneys. The mechanisms underlying renal lipotoxicity involve intracellular lipid accumulation and organelle dysfunction, which trigger oxidative stress and inflammation that consequently result in insulin resistance and albuminuria. However, the specific pathways involved in renal lipotoxicity have not yet been fully understood. We aimed to summarize the current knowledge on the mechanisms by which lipotoxicity affects the renal morphology and function in experimental models of obesity. The accumulation of fatty acids in tubular cells has been described as the main mechanism of lipotoxicity; however, lipids and their metabolism also affect the function and the survival of podocytes. In this review, we presented indication of mitochondrial, lysosomal and endoplasmic reticulum alterations involved in kidney damage caused by obesity. The kidney is vulnerable to lipotoxicity, and studies of the mechanisms underlying renal injury caused by obesity can help identify therapeutic targets to control renal dysfunction.
Collapse
Affiliation(s)
- Barbara Bruna Abreu Castro
- Laboratory of Experimental Nephrology, Nucleus of Animal Experimentation (NIDEAL), Centre of Reproductive Biology (CBR), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
- Nephrology Division and Interdisciplinary Nucleus of Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
| | - Orestes Foresto-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP, São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva-Camara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP, São Paulo, São Paulo, Brazil
| | - Helady Sanders-Pinheiro
- Laboratory of Experimental Nephrology, Nucleus of Animal Experimentation (NIDEAL), Centre of Reproductive Biology (CBR), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
- Nephrology Division and Interdisciplinary Nucleus of Studies and Research in Nephrology (NIEPEN), Federal University of Juiz de Fora (UFJF, Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
45
|
Abstract
Obesity is one of the risk factors for the development and progression of chronic kidney disease (CKD). Several studies have shown the association between increased body mass index and kidney function decline. Obesity leads to CKD directly by acting as an independent risk factor and indirectly through increasing risks for diabetes, hypertension, and atherosclerosis, a group of well-established independent risk factors for CKD. Alterations in renal hemodynamics, inflammation, and in hormones and growth factors results in hyperfiltration injury and focal segmental glomerulosclerosis. In recent years, many studies have shown that the gut microbiome may play a role in the pathogenesis of obesity. Dysbiosis has been noted in obese subjects in both human and animal studies. Changes in the gut microbiome in obese patients promote weight gain by effectively extracting energy from diet, and induction of low-grade inflammation. Evidence also points to the role of inflammation within the adipose tissue in obesity as a key factor in the pathogenesis of obesity-related complications. Thus, obesity is the net result of complex interactions between behavioral, genetic, and environmental factors. In terms of management, conservative approaches are often the first option, but they often are unsuccessful in achieving and/or maintaining weight loss, particularly in severe obesity. Consequently, nonmedical management with bariatric surgery is the most effective treatment option for morbid obesity and has shown mitigation of multiple risk factors for the progression of CKD. The most frequently performed interventions are vertical sleeve gastrectomy and Roux-en-Y gastric bypass. Studies have shown that bariatric surgery is associated with beneficial effects on CKD by mitigating its risk factors by weight loss, reducing insulin resistance, hemoglobin A1c, and proteinuria, in addition to positive long-term outcomes. Because of the epidemic of obesity, the prevalence of obesity in kidney transplant recipients also is increasing. The maximal body mass index (BMI) threshold for kidney transplantation is not clear. The Organ Procurement Transplant Network/Scientific Registry of Transplant Recipients 2019 annual data report showed that the proportion of kidney transplant recipient candidates with a BMI of 30 kg/m2 or greater is increasing steadily. Morbid obesity is linked to adverse graft outcomes including delayed graft function, primary nonfunction, and decreased graft survival. Obesity is also an independent risk factor for cardiovascular death in kidney transplant recipients, suggesting that these patients should not be excluded from transplantation based on their BMI because transplantation is associated with lower mortality compared with dialysis. However, many centers exclude obese patients (with different BMI cut-off values) from transplantation to avoid postoperative complications. To minimize the surgical complications of kidney transplantation in obese patients, our center has adopted the robot-assisted kidney transplantation procedure. Our data show that this approach is comparable with historical nonobese controls in the United Network for Organ Sharing database in terms of patient and graft survival. Another surgical option for this group of patients at our center is a combined robotic sleeve gastrectomy and robotic-assisted kidney transplant. In a recent study, this approach showed promising results in terms of weight loss, patient survival, and graft survival, and might become more common in the future.
Collapse
|
46
|
Caus M, Eritja À, Bozic M. Role of microRNAs in Obesity-Related Kidney Disease. Int J Mol Sci 2021; 22:ijms222111416. [PMID: 34768854 PMCID: PMC8583993 DOI: 10.3390/ijms222111416] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity is a major global health problem and is associated with a significant risk of renal function decline. Obesity-related nephropathy, as one of the complications of obesity, is characterized by a structural and functional damage of the kidney and represents one of the important contributors to the morbidity and mortality worldwide. Despite increasing data linking hyperlipidemia and lipotoxicity to kidney injury, the apprehension of molecular mechanisms leading to a development of kidney damage is scarce. MicroRNAs (miRNAs) are endogenously produced small noncoding RNA molecules with an important function in post-transcriptional regulation of gene expression. miRNAs have been demonstrated to be important regulators of a vast array of physiological and pathological processes in many organs, kidney being one of them. In this review, we present an overview of miRNAs, focusing on their functional role in the pathogenesis of obesity-associated renal pathologies. We explain novel findings regarding miRNA-mediated signaling in obesity-related nephropathies and highlight advantages and future perspectives of the therapeutic application of miRNAs in renal diseases.
Collapse
|
47
|
Young SL, Ryan L, Mullins TP, Flint M, Steane SE, Walton SL, Bielefeldt-Ohmann H, Carter DA, Reichelt ME, Gallo LA. Sotagliflozin, a Dual SGLT1/2 Inhibitor, Improves Cardiac Outcomes in a Normoglycemic Mouse Model of Cardiac Pressure Overload. Front Physiol 2021; 12:738594. [PMID: 34621187 PMCID: PMC8490778 DOI: 10.3389/fphys.2021.738594] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Selective SGLT2 inhibition reduces the risk of worsening heart failure and cardiovascular death in patients with existing heart failure, irrespective of diabetic status. We aimed to investigate the effects of dual SGLT1/2 inhibition, using sotagliflozin, on cardiac outcomes in normal diet (ND) and high fat diet (HFD) mice with cardiac pressure overload. Five-week-old male C57BL/6J mice were randomized to receive a HFD (60% of calories from fat) or remain on ND for 12 weeks. One week later, transverse aortic constriction (TAC) was employed to induce cardiac pressure-overload (50% increase in right:left carotid pressure versus sham surgery), resulting in left ventricular hypertrophic remodeling and cardiac fibrosis, albeit preserved ejection fraction. At 4 weeks post-TAC, mice were treated for 7 weeks by oral gavage once daily with sotagliflozin (10 mg/kg body weight) or vehicle (0.1% tween 80). In ND mice, treatment with sotagliflozin attenuated cardiac hypertrophy and histological markers of cardiac fibrosis induced by TAC. These benefits were associated with profound diuresis and glucosuria, without shifts toward whole-body fatty acid utilization, increased circulating ketones, nor increased cardiac ketolysis. In HFD mice, sotagliflozin reduced the mildly elevated glucose and insulin levels but did not attenuate cardiac injury induced by TAC. HFD mice had vacuolation of proximal tubular cells, associated with less profound sotagliflozin-induced diuresis and glucosuria, which suggests dampened drug action. We demonstrate the utility of dual SGLT1/2 inhibition in treating cardiac injury induced by pressure overload in normoglycemic mice. Its efficacy in high fat-fed mice with mild hyperglycemia and compromised renal morphology requires further study.
Collapse
Affiliation(s)
- Sophia L Young
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.,Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Lydia Ryan
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.,Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Thomas P Mullins
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.,Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Melanie Flint
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Sarah E Steane
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Sarah L Walton
- Cardiovascular Disease Program, Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | - David A Carter
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Linda A Gallo
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia.,Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
48
|
Liu C, Tian J, Jose MD, Dwyer T, Venn AJ. BMI Trajectories from Childhood to Midlife are Associated with Subclinical Kidney Damage in Midlife. Obesity (Silver Spring) 2021; 29:1058-1066. [PMID: 33864358 DOI: 10.1002/oby.23145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 01/22/2023]
Abstract
OBJECTIVE This study aimed to investigate the relationship of BMI trajectories from childhood with subclinical kidney damage (SKD) in midlife, a surrogate measure for chronic kidney disease. METHODS The study followed up 1,442 participants from the 1985 Australian Schools Health and Fitness Survey who were between 7 and 15 years old at the time the survey was conducted and who had BMI measurements in childhood and at least two follow-ups in adulthood. Measures of kidney function for participants 36 to 50 years old were also included. Latent class growth mixture modeling was used to identify the BMI trajectories. Log-binomial regression determined the associations of BMI trajectories with SKD defined as either 1) an estimated glomerular filtration rate (eGFR) of 30 to 60 mL/min/1.73 m2 or 2) an eGFR > 60 mL/min/1.73 m2 with a urine albumin-creatinine ratio ≥ 2.5 mg/mmol (males) or 3.5 mg/mmol (females), adjusting for childhood age, sex, and duration of follow-up. RESULTS Relative to the persistently low trajectory (n = 534, 37.0%), being in higher BMI trajectories was associated with greater risk of SKD in midlife (relative risk [RR] = 1.89, 95% CI = 1.10-3.25 for progressing to moderate [n = 633, 43.9%]; RR = 1.91, 95% CI = 0.95-3.81 for progressing to moderate/high [n = 194, 13.5%]; RR = 2.86, 95% CI = 1.03-7.99 for progressing to high/very high [n = 39, 2.7%]; and RR = 2.47, 95% CI = 0.77-7.94 for adult-onset high [n = 35, 2.4%]). CONCLUSIONS Participants with increasing BMI trajectories from childhood had an increased risk of SKD in midlife.
Collapse
Affiliation(s)
- Conghui Liu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jing Tian
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Matthew D Jose
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Terence Dwyer
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- George Institute for Global Health, University of Oxford, Oxford, UK
| | - Alison J Venn
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
49
|
Ugbaja RN, Ogungbemi K, James AS, Peter Folorunsho A, Abolade SO, Ajamikoko SO, Atayese EO, Adedeji OV. Chitosan from Crabs (Scylla serrata) Represses Hyperlipidemia-Induced Hepato-Renal Dysfunctions in Rats: Modulation of CD43 and p53 Expression. PATHOPHYSIOLOGY 2021; 28:224-237. [PMID: 35366259 PMCID: PMC8830478 DOI: 10.3390/pathophysiology28020015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/29/2022] Open
Abstract
Hepato-renal dysfunctions associated with hyperlipidemia necessitates a continuous search for natural remedies. This study thus evaluated the effect of dietary chitosan on diet-induced hyperlipidemia in rats. A total of 30 male Wistar rats (90 ± 10) g were randomly allotted into six (6) groups (n = 5): Normal diet, High-fat diet (HFD), and Normal diet + 5% chitosan. The three other groups received HFD, supplemented with 1%, 3%, and 5% of chitosan. The feeding lasted for 6 weeks, after which the rats were sacrificed. The liver and kidneys were harvested for analyses. Hepatic alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) activity, and renal biomarkers (ALT, AST, urea, and creatinine) were assayed spectrophotometrically. Additionally, expression of hepatic and renal CD43 and p53 was estimated immunohistochemically. The HFD group had elevated bodyweight compared to normal which was reversed in the chitosan-supplemented groups. Hyperlipidemia caused a significant (p < 0.05) decrease in the hepatic (AST, ALT, and ALP) and renal (AST and ALT) activities, while renal urea and creatinine increased. Furthermore, the HFD group showed an elevated level of hepatic and renal CD43 while p53 expression decreased. However, groups supplemented with chitosan showed improved hepatic and renal biomarkers, as well as corrected the aberrations in the expressions of p53 and CD43. Conclusively, dietary chitosan inclusion in the diet (between 3% and 5%) could effectively improve kidney and liver functionality via abatement of inflammatory responses.
Collapse
Affiliation(s)
- Regina Ngozi Ugbaja
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
- Department of Chemistry/Biochemistry, Nigerian Stored Product Research Institute, P.M.B. 5044 Ibadan, Nigeria
- Correspondence: or ; Tel.: +234-(0)7066050043
| | - Kunle Ogungbemi
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
- Biochemistry Program, Department of Chemical Sciences, Faculty of Science, Augustine University, P.M.B. 1010 Ilara-Epe, Nigeria
| | - Adewale Segun James
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Ayodele Peter Folorunsho
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Samuel Olanrewaju Abolade
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Stella Onajite Ajamikoko
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Eniola Olapeju Atayese
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| | - Omowunmi Victoria Adedeji
- Department of Biochemistry, College of Bioscience, Federal University of Agriculture, P.M.B. 2240 Abeokuta, Nigeria; (K.O.); (A.S.J.); (A.P.F.); (S.O.A.); (S.O.A.); (E.O.A.); (O.V.A.)
| |
Collapse
|
50
|
Dragoljevic D, Veiga CB, Michell DL, Shihata WA, Al-Sharea A, Head GA, Murphy AJ, Kraakman MJ, Lee MKS. A spontaneously hypertensive diet-induced atherosclerosis-prone mouse model of metabolic syndrome. Biomed Pharmacother 2021; 139:111668. [PMID: 34243630 DOI: 10.1016/j.biopha.2021.111668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic Syndrome (MetS) is a complex and multifactorial condition often characterised by obesity, hypertension, hyperlipidaemia, insulin resistance, glucose intolerance and fasting hyperglycaemia. Collectively, MetS can increase the risk of atherosclerotic-cardiovascular disease, which is the leading cause of death worldwide. However, no animal model currently exists to study MetS in the context of atherosclerosis. In this study we developed a pre-clinical mouse model that recapitulates the spectrum of MetS features while developing atherosclerosis. When BPHx mice were placed on a western type diet for 16 weeks, all the classical characteristics of MetS were observed. Comprehensive metabolic analyses and atherosclerotic imaging revealed BPHx mice to be obese and hypertensive, with elevated total plasma cholesterol and triglyceride levels, that accelerated atherosclerosis. Altogether, we demonstrate that the BPHx mouse has all the major components of MetS, and accelerates the development of atherosclerosis.
Collapse
Affiliation(s)
- Dragana Dragoljevic
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia; Monash University, Melbourne, VIC, Australia
| | - Camilla Bertuzzo Veiga
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia
| | | | - Waled A Shihata
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Annas Al-Sharea
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geoffrey A Head
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia; Monash University, Melbourne, VIC, Australia
| | | | - Man K S Lee
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; The University of Melbourne, Melbourne, VIC, Australia; Monash University, Melbourne, VIC, Australia.
| |
Collapse
|