1
|
Chen S, Lu H, Yin G, Zhang X, Meng D, Yu W, Wang L, Liu H, Zhang F. Hesperitin prevents non-alcoholic steatohepatitis by modulating mitochondrial dynamics and mitophagy via the AMPKα-Drp1/PINK1-Parkin signaling pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159570. [PMID: 39454819 DOI: 10.1016/j.bbalip.2024.159570] [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: 11/20/2023] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is becoming a global public health burden, yet effective therapeutic strategies are notably lacking. NAFLD development may be mediated by mitochondrial dysfunction, according to new research. Producing mitochondrial regulators from plant-based substances to treat mitochondrial dysfunction is an appealing approach to treating NAFLD. Hesperetin (HES) is a flavonoid that is found naturally and is a member of the flavanone family. This study aims to clarify the mechanism of HES in preventing NAFLD which is caused by a high-fat diet (HFD). Serum and liver biochemical parameters, liver histology, lipid profiles, and mitochondrial function were evaluated in HFD-induced NAFLD Sprague-Dawley (SD) rats. HES treatment significantly reduced body weight gain, liver weight, and the liver index, while also improving hepatic steatosis, lipid metabolism disorders, and mitochondrial dysfunction in rats with NAFLD. The mechanism was investigated and confirmed using western blot and real-time quantitative polymerase chain reaction (RT-qPCR). We showed that in the liver of NAFLD rats, HES decreased the expression of dynamic-related protein 1 (Drp1), phosphorylated Drp1 at serine-616 (Drp1-pS616) and induced phosphorylated Drp1 at serine-637 (Drp1-pS637), PTEN-induced kinase 1 (PINK1), and E3 Ubiquitin-Protein Ligase Parkin (Parkin) via an AMP-activated protein kinase alpha (AMPKα)-dependent mechanism. Moreover, HES increased the expression of the mitochondrial fusion proteins mitofusin-2 (Mfn2) and optic atrophy 1 (Opa1) while suppressing the expression of fission protein 1 (Fis1). In this work, we identify a unique mechanism by which HES prevents NAFLD from developing. HES may be an attractive potential therapeutic agent to cure NAFLD.
Collapse
Affiliation(s)
- Suwen Chen
- Shandong University of Traditional Chinese Medicine, Jinan 250013, China; Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100032, China
| | - Haifei Lu
- Huanggang Hospital of Chinese Medicine, Affiliated to Hubei University of Chinese Medicine, Huanggang 438000, China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan 250013, China
| | - Xin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan 250013, China
| | - Decheng Meng
- Shandong University of Traditional Chinese Medicine, Jinan 250013, China
| | - Wenfei Yu
- Shandong University of Traditional Chinese Medicine, Jinan 250013, China
| | - Linya Wang
- Shandong University of Traditional Chinese Medicine, Jinan 250013, China
| | - Hongshuai Liu
- Shandong University of Traditional Chinese Medicine, Jinan 250013, China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250013, China.
| |
Collapse
|
2
|
Bracha M, Jaroch A, Falkowski A, Zwierko B, Szwed M, Michalik M, Borkowska A, Szwed K, Kozakiewicz M. Elevated Interleukin-6 Is Associated with Successful Weight Loss 3 Months Postlaparoscopic Sleeve Gastrectomy. Obes Surg 2024; 34:3824-3832. [PMID: 39180618 PMCID: PMC11481637 DOI: 10.1007/s11695-024-07468-y] [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: 05/11/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE Bariatric surgery poses an ever-increasing importance in the effective and long-lasting treatment of obesity, a condition strongly associated with inflammation and increased risk of other diseases and health problems. In obesity-related inflammation, maintaining a balance between pro-inflammatory and anti-inflammatory cytokines is crucial. In this study, we examined early effects of laparoscopic sleeve gastrectomy (LSG) on inflammatory and anti-inflammatory cytokines in obese patients, and assessed their effect on postoperative weight loss. MATERIALS AND METHODS This prospective cohort study was conducted from September 2022 till June 2023. Fifty obese adults were enrolled for LSG. All patients underwent assessments of body measurements, as well as levels of interleukin-6 (IL-6), interleukin-10 (IL-10), and TNF-alpha at baseline and 3 months postsurgery. We developed a decision tree model to predict the success of weight loss. RESULTS At 3 months postsurgery, patients lost 18.9 ± 6.9 kg of excess body weight. A significant decrease was observed for IL-10 (p < 0.0001), simultaneously with a significant increase in IL-6 (p < 0.0001). We found that high IL-6 (> 1.169 pg/mL) levels could contribute to an effective weight loss among patients with a baseline BMI less than 47.46 kg/m2. CONCLUSION Study revealed that 3 months after bariatric surgery, inflammation persists, and its markers significantly influence postoperative weight loss, as indicated by BMI range. Distinct behaviors of IL-10 and IL-6 in relation to obesity underline the necessity of considering individual cytokine profiles when evaluating bariatric surgery outcomes.
Collapse
Affiliation(s)
- Marietta Bracha
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85626, Bydgoszcz, Poland.
| | - Alina Jaroch
- Department of Clinical Neuropsychology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85094, Bydgoszcz, Poland
| | - Adrian Falkowski
- Department of Probability Theory and Stochastic Analysis, Faculty of Mathematics and Computer Science, Nicolaus Copernicus University in Torun, 87100, Torun, Poland
| | - Beata Zwierko
- Department of Clinical Neuropsychology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85094, Bydgoszcz, Poland
| | - Magdalena Szwed
- Department of Clinical Neuropsychology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85094, Bydgoszcz, Poland
| | - Maciej Michalik
- Department of General and Minimally Invasive Surgery, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85168, Bydgoszcz, Poland
| | - Alina Borkowska
- Department of Clinical Neuropsychology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85094, Bydgoszcz, Poland
| | - Krzysztof Szwed
- Department of Clinical Neuropsychology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85094, Bydgoszcz, Poland
| | - Mariusz Kozakiewicz
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85626, Bydgoszcz, Poland
- The Mazovian Academy in Plock, 09402, Plock, Poland
| |
Collapse
|
3
|
Engin A. Bariatric Surgery in Obesity: Metabolic Quality Analysis and Comparison of Surgical Options. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:697-726. [PMID: 39287870 DOI: 10.1007/978-3-031-63657-8_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obesity is a constantly growing health problem which reduces quality of life and life expectancy. Bariatric surgery (BS) for obesity is considered when all other conservative treatment modalities have failed. Comparison of the multidisciplinary programs with BS regarding to the weight loss showed that substantial and durable weight reduction have been achieved only with bariatric surgical treatments. Although laparoscopic sleeve gastrectomy is the most popular BS, it has high long-term failure rates, and it is claimed that one of every three patients will undergo another bariatric procedure within a 10-year period. Although BS provides weight loss and improvement of metabolic comorbidities, in long-term follow-up, weight gain is observed in half of the patients, while decrease in bone mass and nutritional deficiencies occur in up to 90%. Moreover, despite significant weight loss, several psychological aspects of patients are worsened in comparison to preoperative levels. Nearly one-fifth of postoperative patients with "Loss-of-eating control" meet food addiction criteria. Therefore, the benefits of weight loss following bariatric procedures alone are still debated in terms of the proinflammatory and metabolic profile of obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
4
|
Srilatha M, Malla R, Adem MP, Foote JB, Nagaraju GP. Obesity associated pancreatic ductal adenocarcinoma: Therapeutic challenges. Semin Cancer Biol 2023; 97:12-20. [PMID: 37926347 DOI: 10.1016/j.semcancer.2023.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/27/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
Obesity is a prominent health issue worldwide and directly impacts pancreatic health, with obese individuals exhibiting a significant risk for increasing pancreatic ductal adenocarcinoma (PDAC). Several factors potentially explain the increased risk for the development of PDAC, including obesity-induced chronic inflammation within and outside of the pancreas, development of insulin resistance and metabolic dysfunction, promotion of immune suppression within the pancreas during inflammation, pre- and malignant stages, variations in hormones levels (adiponectin, ghrelin, and leptin) produced from the adipose tissue, and acquisition of somatic mutations in tumor once- and suppressor proteins critical for pancreatic tumorigenesis. In this manuscript, we will explore the broad impact of these obesity-induced risk factors on the development and progression of PDAC, focusing on changes within the tumor microenvironment (TME) as they pertain to prevention, current therapeutic strategies, and future directions for targeting obesity management as they relate to the prevention of pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | - Ramarao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India
| | - Megha Priya Adem
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam (Women's University), Tirupati, Andhra Pradesh 517502, India
| | - Jeremy B Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | |
Collapse
|
5
|
Zhang LH, Wang J, Tan BH, Yin YB, Kang YM. Roux-en-Y Gastric Bypass Improves Insulin Sensitivity in Obese Rats with Type 2 Diabetes Mellitus by Regulating the Grin3a/AMPK Signal Axis in Hypothalamic Arcuate Nucleus. Diabetes Metab Syndr Obes 2023; 16:3617-3629. [PMID: 38028990 PMCID: PMC10644885 DOI: 10.2147/dmso.s430445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The objective of this study was to explore the effects and related mechanisms of Roux-en-Y gastric bypass (RYGB) on insulin sensitivity in obese rats with type 2 diabetes mellitus (T2DM). Methods The obese T2DM rat model was constructed by feeding a high-fat diet and injecting streptozotocin (STZ), and treated with RYGB. Grin3a shRNA was injected into the bilateral hypothalamic arcuate nucleus (ARC) to knockdown the Grin3a expression on T2DM rats. Eight weeks after operation, the body weight, fasting blood glucose (FBG), fasting serum insulin (FSI), homeostatic model assessment of insulin resistance (HOMA-IR), and plasma triglyceride (TG) levels were assessed. Hematoxylin & eosin (H&E) staining was adopted to observe the white adipose tissue (WAT) of rats. Western blot and qRT-PCR were used to detect the expression of Grin3a, adenosine 5' monophosphate-activated protein kinase (AMPK) and p-AMPK in ARC of rats. Later, the plasmid over-expressing or knocking down Grin3a was transfected into differentiated 3T3-L1 adipocytes, and the TG level and the formation of lipid droplets in adipocyte were assessed by TG kit and oil red O staining. The expression of lipogenic transcription factors in cells was detected by qRT-PCR. Results RYGB reduced FBG, FSI, HOMA-IR and plasma TG levels in T2DM rats while increasing Grin3a expression and p-AMPK/AMPK ratio in ARC. Knockdown of Grin3a not only reversed the decrease of FBG, FSI, HOMA-IR and plasma TG levels in T2DM rats induced by RYGB, but also reversed the up-regulation of p-AMPK/AMPK ratio in ARC affected by RYGB. Moreover, knocking down Grin3a significantly increased the TG level, promoted the formation of lipid droplets and up-regulated the expressions of lipogenic transcription factors in adipocytes. Conclusion RYGB improved the insulin sensitivity, reduced the plasma TG level and lessens the fat accumulation in obese T2DM rats by regulating the Grin3a/AMPK signal in ARC.
Collapse
Affiliation(s)
- Li-Hai Zhang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Xi’an, People’s Republic of China
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Jiao Wang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Xi’an, People’s Republic of China
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Bai-Hong Tan
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Yan-Bin Yin
- Six Wards of General Surgery, The First Affiliated Hospital of Jiamusi University, Jiamusi, People’s Republic of China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University School of Basic Medical Sciences, Xi’an, People’s Republic of China
| |
Collapse
|
6
|
Göransson O, Kopietz F, Rider MH. Metabolic control by AMPK in white adipose tissue. Trends Endocrinol Metab 2023; 34:704-717. [PMID: 37673765 DOI: 10.1016/j.tem.2023.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023]
Abstract
White adipose tissue (WAT) plays an important role in the integration of whole-body metabolism by storing fat and mobilizing triacylglycerol when needed. The released free fatty acids can then be oxidized by other tissues to provide ATP. AMP-activated protein kinase (AMPK) is a key regulator of metabolic pathways, and can be targeted by a new generation of direct, small-molecule activators. AMPK activation in WAT inhibits insulin-stimulated lipogenesis and in some situations also inhibits insulin-stimulated glucose uptake, but AMPK-induced inhibition of β-adrenergic agonist-stimulated lipolysis might need to be re-evaluated in vivo. The lack of dramatic effects of AMPK activation on basal metabolism in WAT could be advantageous when treating type 2 diabetes with pharmacological pan-AMPK activators.
Collapse
Affiliation(s)
- Olga Göransson
- Lund University, Department of Experimental Medical Science, BMC, 221 84 Lund, Sweden.
| | - Franziska Kopietz
- Lund University, Department of Experimental Medical Science, BMC, 221 84 Lund, Sweden
| | - Mark H Rider
- Université catholique de Louvain (UCLouvain) and de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium
| |
Collapse
|
7
|
Steinberg GR, Hardie DG. New insights into activation and function of the AMPK. Nat Rev Mol Cell Biol 2023; 24:255-272. [PMID: 36316383 DOI: 10.1038/s41580-022-00547-x] [Citation(s) in RCA: 306] [Impact Index Per Article: 153.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
The classical role of AMP-activated protein kinase (AMPK) is as a cellular energy sensor activated by falling energy status, signalled by increases in AMP to ATP and ADP to ATP ratios. Once activated, AMPK acts to restore energy homeostasis by promoting ATP-producing catabolic pathways while inhibiting energy-consuming processes. In this Review, we provide an update on this canonical (AMP/ADP-dependent) activation mechanism, but focus mainly on recently described non-canonical pathways, including those by which AMPK senses the availability of glucose, glycogen or fatty acids and by which it senses damage to lysosomes and nuclear DNA. We also discuss new findings on the regulation of carbohydrate and lipid metabolism, mitochondrial and lysosomal homeostasis, and DNA repair. Finally, we discuss the role of AMPK in cancer, obesity, diabetes, nonalcoholic steatohepatitis (NASH) and other disorders where therapeutic targeting may exert beneficial effects.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
8
|
Latteri S, Sofia M, Puleo S, Di Vincenzo A, Cinti S, Castorina S. Mechanisms linking bariatric surgery to adipose tissue, glucose metabolism, fatty liver disease and gut microbiota. Langenbecks Arch Surg 2023; 408:101. [PMID: 36826628 PMCID: PMC9957865 DOI: 10.1007/s00423-023-02821-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 01/25/2023] [Indexed: 02/25/2023]
Abstract
PURPOSE In the last 20 years, bariatric surgery has achieved an important role in translational and clinical research because of obesity comorbidities. Initially, a tool to lose weight, bariatric surgery now has been shown to be involved in several metabolic pathways. METHODS We conducted a narrative review discussing the underlying mechanisms that could explain the impact of bariatric surgery and the relationship between obesity and adipose tissue, T2D, gut microbiota, and NAFLD. RESULTS Bariatric surgery has an impact in the relation between obesity and type 2 diabetes, but in addition it induces the white-to-brown adipocyte trans-differentiation, by enhancing thermogenesis. Another issue is the connection of bariatric surgery with the gut microbiota and its role in the complex mechanism underlying weight gain. CONCLUSION Bariatric surgery modifies gut microbiota, and these modifications influence lipid metabolism, leading to improvement of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Saverio Latteri
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Maria Sofia
- Department of General Surgery, Cannizzaro Hospital, Via Messina 829, 95126, Catania, Italy.
| | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Angelica Di Vincenzo
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, Center of Obesity, Marche Polytechnic University, Via Tronto 10A, 60020, Ancona, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Catania, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
9
|
Kopietz F, Degerman E, Göransson O. AMPKβ isoform expression patterns in various adipocyte models and in relation to body mass index. Front Physiol 2022; 13:928964. [PMID: 35991175 PMCID: PMC9386264 DOI: 10.3389/fphys.2022.928964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
AMP-activated protein kinase (AMPK) activation is considered a useful strategy for the treatment of type 2 diabetes (T2D). It is unclear whether the expression and/or activity of AMPK in adipocytes is dysregulated in obesity. Also, the expression/activity pattern of AMPKβ isoforms, which are targets for AMPK activators, in adipocytes remains elusive. In this study we show that the two AMPKβ isoforms make roughly equal contributions to AMPK activity in primary human and mouse adipocytes, whereas in cultured 3T3-L1 adipocytes of mouse origin and in primary rat adipocytes, β1-associated activity clearly dominates. Additionally, we found that obesity is not associated with changes in AMPK subunit expression or kinase activity in adipocytes isolated from subcutaneous adipose tissue from individuals with various BMI.
Collapse
|
10
|
Preservation of Fat-free Mass in the first year after Bariatric Surgery: A systematic review and meta-analysis of 122 studies and 10758 participants. Surg Obes Relat Dis 2022; 18:964-982. [DOI: 10.1016/j.soard.2022.02.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 02/07/2023]
|
11
|
Abad-Jiménez Z, López-Domènech S, García-Gargallo C, Vezza T, Gómez-Abril SÁ, Morillas C, Díaz-Pozo P, Falcón R, Bañuls C, Víctor VM, Rocha M. Roux-en-Y Gastric Bypass Modulates AMPK, Autophagy and Inflammatory Response in Leukocytes of Obese Patients. Biomedicines 2022; 10:biomedicines10020430. [PMID: 35203639 PMCID: PMC8962362 DOI: 10.3390/biomedicines10020430] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Obesity is characterized by low-grade chronic inflammation, metabolic overload, and impaired endothelial and cardiovascular function. Roux-en-Y gastric bypass (RYGB) results in amelioration of the pro-oxidant status of leukocytes and the metabolic profile. Nevertheless, little is known about the precise mechanism that drives systemic and metabolic improvements following bariatric surgery. In this cohort study, we investigated the effect of RYGB on molecular pathways involving energy homeostasis in leukocytes in 43 obese subjects one year after surgery. In addition to clinical and biochemical parameters, we determined protein expression of systemic proinflammatory cytokines by Luminex®, different markers of inflammation, endoplasmic reticulum (ER) stress, autophagy/mitophagy by western blot, and mitochondrial membrane potential by fluorescence imaging. Bariatric surgery induced an improvement in metabolic outcomes that was accompanied by a systemic drop in hsCRP, IL6, and IL1β levels, and a slowing down of intracellular inflammatory pathways in leukocytes (NF-κB and MCP-1), an increase in AMPK content, a reduction of ER stress (ATF6 and CHOP), augmented autophagy/mitophagy markers (Beclin 1, ATG5, LC3-I, LC3-II, NBR1, and PINK1), and a decrease of mitochondrial membrane potential. These findings shed light on the specific molecular mechanisms by which RYGB facilitates metabolic improvements, highlighting the relevance of pathways involving energy homeostasis as key mediators of these outcomes. In addition, since leukocytes are particularly exposed to physiological changes, they could be used in routine clinical practice as a good sensor of the whole body’s responses.
Collapse
Affiliation(s)
- Zaida Abad-Jiménez
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
| | - Sandra López-Domènech
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
- Correspondence: (S.L.-D.); (V.M.V.); (M.R.); Tel.: +34-96-318-91-32 (M.R.)
| | - Celia García-Gargallo
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
| | - Teresa Vezza
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
| | - Segundo Ángel Gómez-Abril
- Department of General and Digestive System Surgery, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain;
- Department of Surgery, Faculty of Medicine and Dentistry, University of Valencia, Av Blasco Ibáñez 13, 46010 Valencia, Spain
| | - Carlos Morillas
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
| | - Pedro Díaz-Pozo
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
| | - Rosa Falcón
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
| | - Celia Bañuls
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
| | - Víctor M. Víctor
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
- CIBERehd-Department of Pharmacology, University of Valencia, Av Blasco Ibáñez 15, 46010 Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Av Blasco Ibáñez 13, 46010 Valencia, Spain
- Correspondence: (S.L.-D.); (V.M.V.); (M.R.); Tel.: +34-96-318-91-32 (M.R.)
| | - Milagros Rocha
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (C.G.-G.); (T.V.); (C.M.); (P.D.-P.); (R.F.); (C.B.)
- CIBERehd-Department of Pharmacology, University of Valencia, Av Blasco Ibáñez 15, 46010 Valencia, Spain
- Correspondence: (S.L.-D.); (V.M.V.); (M.R.); Tel.: +34-96-318-91-32 (M.R.)
| |
Collapse
|
12
|
Mirhosseini Dehabadi S, Sayadi Shahraki M, Mahmoudieh M, Kalidari B, Melali H, Mousavi M, Ghourban Abadi M, Mirhosseini S. Bone health after bariatric surgery: Consequences, prevention, and treatment. Adv Biomed Res 2022; 11:92. [DOI: 10.4103/abr.abr_182_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/05/2022] Open
|
13
|
Palmer TM, Salt IP. Nutrient regulation of inflammatory signalling in obesity and vascular disease. Clin Sci (Lond) 2021; 135:1563-1590. [PMID: 34231841 DOI: 10.1042/cs20190768] [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: 02/10/2021] [Revised: 06/10/2021] [Accepted: 06/28/2021] [Indexed: 11/17/2022]
Abstract
Despite obesity and diabetes markedly increasing the risk of developing cardiovascular diseases, the molecular and cellular mechanisms that underlie this association remain poorly characterised. In the last 20 years it has become apparent that chronic, low-grade inflammation in obese adipose tissue may contribute to the risk of developing insulin resistance and type 2 diabetes. Furthermore, increased vascular pro-inflammatory signalling is a key event in the development of cardiovascular diseases. Overnutrition exacerbates pro-inflammatory signalling in vascular and adipose tissues, with several mechanisms proposed to mediate this. In this article, we review the molecular and cellular mechanisms by which nutrients are proposed to regulate pro-inflammatory signalling in adipose and vascular tissues. In addition, we examine the potential therapeutic opportunities that these mechanisms provide for suppression of inappropriate inflammation in obesity and vascular disease.
Collapse
Affiliation(s)
- Timothy M Palmer
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, United Kingdom
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
14
|
Turner L, Santosa S. Putting ATM to BED: How Adipose Tissue Macrophages Are Affected by Bariatric Surgery, Exercise, and Dietary Fatty Acids. Adv Nutr 2021; 12:1893-1910. [PMID: 33979430 PMCID: PMC8483961 DOI: 10.1093/advances/nmab011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/17/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
With increasing adiposity in obesity, adipose tissue macrophages contribute to adipose tissue malfunction and increased circulating proinflammatory cytokines. The chronic low-grade inflammation that occurs in obesity ultimately gives rise to a state of metainflammation that increases the risk of metabolic disease. To date, only lifestyle and surgical interventions have been shown to be somewhat effective at reversing the negative consequences of obesity and restoring adipose tissue homeostasis. Exercise, dietary interventions, and bariatric surgery result in immunomodulation, and for some individuals their effects are significant with or without weight loss. Robust evidence suggests that these interventions reduce chronic inflammation, in part, by affecting macrophage infiltration and promoting a phenotypic switch from the M1- to M2-like macrophages. The purpose of this review is to discuss the impact of dietary fatty acids, exercise, and bariatric surgery on cellular characteristics affecting adipose tissue macrophage presence and phenotypes in obesity.
Collapse
Affiliation(s)
- Laurent Turner
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Montreal, Quebec, Canada,Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Montreal, Quebec, Canada
| | | |
Collapse
|
15
|
Ferraz-Bannitz R, Welendorf CR, Coelho PO, Salgado W, Nonino CB, Beraldo RA, Foss-Freitas MC. Bariatric surgery can acutely modulate ER-stress and inflammation on subcutaneous adipose tissue in non-diabetic patients with obesity. Diabetol Metab Syndr 2021; 13:19. [PMID: 33593418 PMCID: PMC7887793 DOI: 10.1186/s13098-021-00623-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bariatric surgery, especially Roux-en-Y gastric bypass (RYGB), is the most effective and durable treatment option for severe obesity. The mechanisms involving adipose tissue may be important to explain the effects of surgery. METHODS We aimed to identify the genetic signatures of adipose tissue in patients undergoing RYGB. We evaluated 13 obese, non-diabetic patients (mean age 37 years, 100% women, Body mass index (BMI) 42.2 kg/m2) one day before surgery, 3 and 6 months (M) after RYGB. RESULTS Analysis of gene expression in adipose tissue collected at surgery compared with samples collected at 3 M and 6 M Post-RYGB showed that interleukins [Interleukin 6, Tumor necrosis factor-α (TNF-α), and Monocyte chemoattractant protein-1(MCP1)] and endoplasmic reticulum stress (ERS) genes [Eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3) and Calreticulin (CALR)] decreased during the follow-up (P ≤ 0.01 for all). Otherwise, genes involved in energy homeostasis [Adiponectin and AMP-activated protein kinase (AMPK)], cellular response to oxidative stress [Sirtuin 1, Sirtuin 3, and Nuclear factor erythroid 2-related factor 2 (NRF2)], mitochondrial biogenesis [Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α)] and amino acids metabolism [General control nonderepressible 2 (GCN2)] increased from baseline to all other time points evaluated (P ≤ 0.01 for all). Also, expression of Peroxisome proliferator-activated receptor gamma (PPARϒ) (adipogenesis regulation) was significantly decreased after RYGB (P < 0.05). Additionally, we observed that PGC1α, SIRT1 and AMPK strongly correlated to BMI at 3 M (P ≤ 0.01 for all), as well as ADIPOQ and SIRT1 to BMI at 6 M (P ≤ 0.01 for all). CONCLUSIONS Our findings demonstrate that weight loss is associated with amelioration of inflammation and ERS and increased protection against oxidative stress in adipose tissue. These observations are strongly correlated with a decrease in BMI and essential genes that control cellular energy homeostasis, suggesting an adaptive process on a gene expression level during the caloric restriction and weight loss period after RYGB. Trial registration CAAE: 73,585,317.0.0000.5440.
Collapse
Affiliation(s)
- Rafael Ferraz-Bannitz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil.
| | - Caroline Rossi Welendorf
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Priscila Oliveira Coelho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Wilson Salgado
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Carla Barbosa Nonino
- Laboratory of Nutrigenomic Studies, Ribeirão Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Rebeca A Beraldo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Maria Cristina Foss-Freitas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil.
| |
Collapse
|
16
|
Mosinski JD, Aminian A, Axelrod CL, Batayyah E, Romero-Talamas H, Daigle C, Mulya A, Scelsi A, Schauer PR, Brethauer SA, Kirwan JP. Roux-en-Y gastric bypass restores islet function and morphology independent of body weight in ZDF rats. Am J Physiol Endocrinol Metab 2021; 320:E392-E398. [PMID: 33427046 PMCID: PMC8260360 DOI: 10.1152/ajpendo.00467.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Reductions in β-cell number and function contribute to the onset type 2 diabetes (T2D). Roux-en-Y gastric bypass (RYGB) surgery can resolve T2D within days of operation, indicating a weight-independent mechanism of glycemic control. We hypothesized that RYGB normalizes glucose homeostasis by restoring β-cell structure and function. Male Zucker Diabetic Fatty (fa/fa; ZDF) rats were randomized to sham surgery (n = 16), RYGB surgery (n = 16), or pair feeding (n = 16). Age-matched lean (fa/+) rats (n = 8) were included as a secondary control. Postprandial metabolism was assessed by oral glucose tolerance testing before and 27 days after surgery. Fasting and postprandial plasma GLP-1 was determined by mixed meal tolerance testing. Fasting plasma glucagon was also measured. β-cell function was determined in isolated islets by a glucose-stimulated insulin secretion assay. Insulin and glucagon positive areas were evaluated in pancreatic sections by immunohistochemistry. RYGB reduced body weight (P < 0.05) and improved glucose tolerance (P < 0.05) compared with sham surgery. RYGB reduced fasting glucose compared with both sham (P < 0.01) and pair-fed controls (P < 0.01). Postprandial GLP-1 (P < 0.05) was elevated after RYGB compared with sham surgery. RYGB islets stimulated with 20 mM glucose had higher insulin secretion than both sham and pair-fed controls (P < 0.01) and did not differ from lean controls. Insulin content was greater after RYGB compared with the sham (P < 0.05) and pair-fed (P < 0.05) controls. RYGB improves insulin secretion and pancreatic islet function, which may contribute to the remission of type 2 diabetes following bariatric surgery.NEW & NOTEWORTHY The onset and progression of type 2 diabetes (T2D) results from failure to secrete sufficient amounts of insulin to overcome peripheral insulin resistance. Here, we demonstrate that Roux-en-Y gastric bypass (RYGB) restores islet function and morphology compared to sham and pair-fed controls in ZDF rats. The improvements in islet function were largely attributable to enhanced insulin content and secretory function in response to glucose stimulation.
Collapse
Affiliation(s)
- J David Mosinski
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, Ohio
| | - Ali Aminian
- Bariatric Metabolic Institute, Cleveland Clinic, Cleveland, Ohio
| | - Christopher L Axelrod
- Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Bariatric and Metabolic Institute, Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Esam Batayyah
- Bariatric Metabolic Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | - Anny Mulya
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Metabolic Translational Research Center, Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, Ohio
| | - Amanda Scelsi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Philip R Schauer
- Bariatric Metabolic Institute, Cleveland Clinic, Cleveland, Ohio
- Bariatric and Metabolic Institute, Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Metabolic Translational Research Center, Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, Ohio
| | - Stacy A Brethauer
- Bariatric Metabolic Institute, Cleveland Clinic, Cleveland, Ohio
- Metabolic Translational Research Center, Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, Ohio
| | - John P Kirwan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Bariatric and Metabolic Institute, Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Metabolic Translational Research Center, Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
17
|
Zhang C, Biehl Rudkjær LC, Cachón MF, Falkenhahn M, Theis S, Schmidt T, Vrang N, Jelsing J, Rigbolt K. Transcriptomic changes in pancreatic islets, adipose and liver after Roux-en-Y gastric bypass in a diet-induced obese rat model. Peptides 2021; 136:170467. [PMID: 33253774 DOI: 10.1016/j.peptides.2020.170467] [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] [Received: 08/27/2020] [Revised: 10/28/2020] [Accepted: 11/21/2020] [Indexed: 12/28/2022]
Abstract
Roux-en-Y gastric bypass (RYGB) is the most efficient intervention in morbid obesity and promotes metabolic improvements in several peripheral tissues. However, the underlying molecular mechanisms are still poorly understood. To further understand the effects of RYGB on peripheral tissues transcriptomes, we determined transcriptome signatures in pancreatic islets, adipose and liver tissue from diet-induced obese (DIO) rats model following RYGB. Whereas RYGB led to discrete gene expression changes in pancreatic islets, substantial transcriptome changes were observed in metabolic and immune signaling pathways in adipose tissue and the liver, indicating major gene adaptive responses in fat-storing tissues. Compared to RYGB DIO rats, peripheral tissue transcriptome signatures were markedly different in caloric restricted weight matching DIO rats, implying that caloric restriction paradigms do not reflect transcriptomic regulations of RYGB induced weight loss. The present gene expression study may serve as a basis for further investigations into molecular regulatory effects in peripheral tissues following RYGB-induced weight loss.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Theis
- Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
18
|
Carlsson ER, Fenger M, Henriksen T, Kjaer LK, Worm D, Hansen DL, Madsbad S, Poulsen HE. Reduction of oxidative stress on DNA and RNA in obese patients after Roux-en-Y gastric bypass surgery-An observational cohort study of changes in urinary markers. PLoS One 2020; 15:e0243918. [PMID: 33315915 PMCID: PMC7735613 DOI: 10.1371/journal.pone.0243918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 11/18/2020] [Indexed: 01/14/2023] Open
Abstract
Increased oxidative stress in obesity and diabetes is associated with morbidity and mortality risks. Levels of oxidative damage to DNA and RNA can be estimated through measurement of 8-oxo-7,8-dihydro-2´-deoxyguanosine (8-oxodG) and 8-oxo-7,8-dihydroguanosine (8-oxoGuo) in urine. Both markers have been associated with type 2 diabetes, where especially 8-oxoGuo is prognostic for mortality risk. We hypothesized that Roux-en-Y gastric bypass (RYGB) surgery that has considerable effects on bodyweight, hyperglycemia and mortality, might be working through mechanisms that reduce oxidative stress, thereby reducing levels of the urinary markers. We used liquid chromatography coupled with tandem mass spectrometry to analyze the content of 8-oxodG and 8-oxoGuo in urinary samples from 356 obese patients treated with the RYGB-procedure. Mean age (SD) was 44.2 (9.6) years, BMI was 42.1 (5.6) kg/m2. Ninety-six (27%) of the patients had type 2 diabetes. Excretion levels of each marker before and after surgery were compared as estimates of the total 24-hour excretion, using a model based on glomerular filtration rate (calculated from cystatin C, age, height and weight), plasma- and urinary creatinine. The excretion of 8-oxodG increased in the first months after RYGB. For 8-oxoGuo, a gradual decrease was seen. Two years after RYGB and a mean weight loss of 35 kg, decreased hyperglycemia and insulin resistance, excretion levels of both markers were reduced by approximately 12% (P < 0.001). For both markers, mean excretion levels were about 30% lower in the female subgroup (P < 0.0001). Also, in this subgroup, excretion of 8-oxodG was significantly lower in patients with than without diabetes. We conclude, that oxidative damage to nucleic acids, reflected in the excretion of 8-oxodG and 8-oxoGuo, had decreased significantly two years after RYGB-indicating that reduced oxidative stress could be contributing to the many long-term benefits of RYGB-surgery in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Elin Rebecka Carlsson
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Clinical Biochemistry, Nordsjaellands Hospital, University of Copenhagen, Hilleroed, Denmark
| | - Mogens Fenger
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Trine Henriksen
- Department of Clinical Pharmacology, Bispebjerg Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Laura Kofoed Kjaer
- Department of Clinical Pharmacology, Bispebjerg Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Dorte Worm
- Department of Medicine, Amager hospital, Copenhagen, Denmark
| | | | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Henrik Enghusen Poulsen
- Department of Clinical Pharmacology, Bispebjerg Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
19
|
More than an Anti-diabetic Bariatric Surgery, Metabolic Surgery Alleviates Systemic and Local Inflammation in Obesity. Obes Surg 2019; 28:3658-3668. [PMID: 30187424 DOI: 10.1007/s11695-018-3400-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Obesity, associated with increased risk of type 2 diabetes (T2D), cardiovascular disease, and hepatic steatosis et al., has become a major global health problem. Recently, obesity has been proven to be under a status of low-grade, chronic inflammation, which contributes to insulin resistance and T2D. Bariatric surgery is currently an effective treatment for the control of morbid obesity and T2D, which impels ongoing efforts to clarify physiological and molecular mechanisms mediating these benefits. The correlation between obesity, inflammation, and T2D has been revealed to a certain extent, and studies have shed light on the effect of bariatric surgery on inflammatory status of subjects with obesity. Based on recent findings, this review focuses on the relationship between inflammation, obesity, and bariatric surgery.
Collapse
|
20
|
Abstract
In recent years the surgical treatment of metabolic diseases has become established as an effective alternative to conservative treatment. The new S3 guidelines address these changes and give clear indications for obesity surgery. One of the core points of the new guidelines is the differentiation between obesity surgery and metabolic surgery. In obesity surgery the primary aim of treatment is weight loss whereas for metabolic indications the aim is an improvement of comorbidities independent of the body mass index (BMI). With respect to the selection of procedures sleeve gastrectomy (SG) and the traditional Roux-en-Y gastric bypass (RYGB) can be used as safe and evidence-based operative procedures. The RYGB has better metabolic effects but higher complication and reintervention rates. More recent procedures, such as the one anastomosis gastric bypass (OAGB) and single anastomosis duodeno-ileal (SADI) bypass possibly have slightly stronger metabolic effects, however, the risk of malnutrition and vitamin deficiency is higher.
Collapse
Affiliation(s)
- A T Billeter
- Klinik für Allgemein‑, Viszeral- und Transplantationschirurgie, Universitätsklinik Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Deutschland
| | - B P Müller-Stich
- Klinik für Allgemein‑, Viszeral- und Transplantationschirurgie, Universitätsklinik Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Deutschland.
| |
Collapse
|
21
|
Muo IM, MacDonald SD, Madan R, Park SJ, Gharib AM, Martinez PE, Walter MF, Yang SB, Rodante JA, Courville AB, Walter PJ, Cai H, Glicksman M, Guerrieri GM, Ben-Dor RR, Ouwerkerk R, Mao S, Chung JH. Early effects of roflumilast on insulin sensitivity in adults with prediabetes and overweight/obesity involve age-associated fat mass loss - results of an exploratory study. Diabetes Metab Syndr Obes 2019; 12:743-759. [PMID: 31213865 PMCID: PMC6542328 DOI: 10.2147/dmso.s182953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Roflumilast (Daliresp, Daxas) is a FDA-approved phosphodiesterase 4 (PDE4) inhibitor for the treatment of moderate-to-severe chronic obstructive pulmonary disease. In mice and in limited human studies, this oral medication can cause weight loss and improve insulin sensitivity. We set out to determine the mechanism of its effect on insulin sensitivity. PATIENTS AND METHODS Eight adults with overweight/obesity and prediabetes received roflumilast for 6 weeks. Before and after roflumilast, subjects underwent tests of insulin sensitivity, mixed meal test, body composition, markers of inflammation, and mitochondria function. Dietary intake and physical activity were also assessed. Our primary outcome was the change in peripheral insulin sensitivity, as assessed by the hyper-insulinemic euglycemic clamp. RESULTS This study was underpowered for the primary outcome. Pre- and post-roflumilast mean peripheral insulin sensitivity were 48.7 and 70.0 mg/g fat free mass/minute, respectively, (P-value=0.18), respectively. Among the mixed meal variables, roflumilast altered glucagon-like peptide 1 (GLP-1) hormone the most, although the average effect was not statistically significant (P=0.18). Roflumilast induced a trend toward significance in 1) decreased energy intake (from 11,095 KJ to 8,4555 KJ, P=0.07), 2) decreased fat mass (from 34.53 to 32.97 kg, P=0.06), 3) decreased total and LDL cholesterol (P=0.06 for both variables), and 4) increased plasma free fatty acids (from 0.40 to 0.50 mEq/L, P=0.09) The interval changes in adiposity and free fatty acid were significantly associated with the subject's age (P-value range= <0.001 to 0.02 for the correlations). Inflammatory and adhesion markers, though unchanged, significantly correlated with one another and with incretin hormones only after roflumilast. CONCLUSION We demonstrate, for the first time in humans, increasing percentage of fat mass loss from roflumilast with increasing age in adults with prediabetes and overweight/obesity. We also demonstrate novel associations among roflumilast-induced changes in incretin hormones, inflammatory markers, peripheral insulin sensitivity, and adiposity. We conclude that roflumilast's early effects on insulin sensitivity is indirect and likely mediated through roflumilast's prioritization of lipid over glucose handling. CLINICAL TRIALS REGISTRATION NCT01862029.
Collapse
Affiliation(s)
- Ijeoma M Muo
- Laboratory of Obesity and Aging Research NHLBI, National Institutes of Health, Bethesda, MD 20892, USA, ,
| | - Sandra D MacDonald
- NHLBI Pulmonary Branch, Laboratory of Chronic Airway Infections, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ritu Madan
- Diabetes Endocrinology and Obesity Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sung-Jun Park
- Laboratory of Obesity and Aging Research NHLBI, National Institutes of Health, Bethesda, MD 20892, USA, ,
| | - Ahmed M Gharib
- Biomedical and Metabolic Imaging Branch NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pedro E Martinez
- Section on Behavioral Endocrinology, NIMH, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mary F Walter
- Diabetes Endocrinology and Obesity Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shanna B Yang
- Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD 20892, USA
| | - Justin A Rodante
- Laboratory of Inflammation and Cardiometabolic Diseases, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amber B Courville
- Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter J Walter
- Mass Spectrometry Clinical Core, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hongyi Cai
- Mass Spectrometry Clinical Core, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Glicksman
- Diabetes Endocrinology and Obesity Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gioia M Guerrieri
- Section on Behavioral Endocrinology, NIMH, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rivka R Ben-Dor
- NIMH, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald Ouwerkerk
- Biomedical and Metabolic Imaging Branch NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephanie Mao
- Laboratory of Obesity and Aging Research NHLBI, National Institutes of Health, Bethesda, MD 20892, USA, ,
| | - Jay H Chung
- Laboratory of Obesity and Aging Research NHLBI, National Institutes of Health, Bethesda, MD 20892, USA, ,
| |
Collapse
|
22
|
Pérez-Pevida B, Escalada J, Miras AD, Frühbeck G. Mechanisms Underlying Type 2 Diabetes Remission After Metabolic Surgery. Front Endocrinol (Lausanne) 2019; 10:641. [PMID: 31608010 PMCID: PMC6761227 DOI: 10.3389/fendo.2019.00641] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/04/2019] [Indexed: 01/19/2023] Open
Abstract
Type 2 diabetes prevalence is increasing dramatically worldwide. Metabolic surgery is the most effective treatment for selected patients with diabetes and/or obesity. When compared to intensive medical therapy and lifestyle intervention, metabolic surgery has shown superiority in achieving glycemic improvement, reducing number of medications and cardiovascular risk factors, which translates in long-term benefits on cardiovascular morbidity and mortality. The mechanisms underlying diabetes improvement after metabolic surgery have not yet been clearly understood but englobe a complex interaction among improvements in beta cell function and insulin secretion, insulin sensitivity, intestinal gluconeogenesis, changes in glucose utilization, and absorption by the gut and changes in the secretory pattern and morphology of adipose tissue. These are achieved through different mediators which include an enhancement in gut hormones release, especially, glucagon-like peptide 1, changes in bile acids circulation, gut microbiome, and glucose transporters expression. Therefore, this review aims to provide a comprehensive appraisal of what is known so far to better understand the mechanisms through which metabolic surgery improves glycemic control facilitating future research in the field.
Collapse
Affiliation(s)
- Belén Pérez-Pevida
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, United Kingdom
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- *Correspondence: Belén Pérez-Pevida
| | - Javier Escalada
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Alexander D. Miras
- Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, United Kingdom
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- Biomedical Research Networking Center for Physiopathology of Obesity and Nutrition (CIBEROBN), ISCIII, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Gema Frühbeck
| |
Collapse
|
23
|
Burhans MS, Hagman DK, Kuzma JN, Schmidt KA, Kratz M. Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus. Compr Physiol 2018; 9:1-58. [PMID: 30549014 DOI: 10.1002/cphy.c170040] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The objective of this comprehensive review is to summarize and discuss the available evidence of how adipose tissue inflammation affects insulin sensitivity and glucose tolerance. Low-grade, chronic adipose tissue inflammation is characterized by infiltration of macrophages and other immune cell populations into adipose tissue, and a shift toward more proinflammatory subtypes of leukocytes. The infiltration of proinflammatory cells in adipose tissue is associated with an increased production of key chemokines such as C-C motif chemokine ligand 2, proinflammatory cytokines including tumor necrosis factor α and interleukins 1β and 6 as well as reduced expression of the key insulin-sensitizing adipokine, adiponectin. In both rodent models and humans, adipose tissue inflammation is consistently associated with excess fat mass and insulin resistance. In humans, associations with insulin resistance are stronger and more consistent for inflammation in visceral as opposed to subcutaneous fat. Further, genetic alterations in mouse models of obesity that reduce adipose tissue inflammation are-almost without exception-associated with improved insulin sensitivity. However, a dissociation between adipose tissue inflammation and insulin resistance can be observed in very few rodent models of obesity as well as in humans following bariatric surgery- or low-calorie-diet-induced weight loss, illustrating that the etiology of insulin resistance is multifactorial. Taken together, adipose tissue inflammation is a key factor in the development of insulin resistance and type 2 diabetes in obesity, along with other factors that likely include inflammation and fat accumulation in other metabolically active tissues. © 2019 American Physiological Society. Compr Physiol 9:1-58, 2019.
Collapse
Affiliation(s)
- Maggie S Burhans
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Derek K Hagman
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Jessica N Kuzma
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Kelsey A Schmidt
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Mario Kratz
- Cancer Prevention Program, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Epidemiology, University of Washington, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
24
|
Impact of Ileal Transposition Surgical Intervention on Antioxidant Status Measured in Liver Tissue of Obese Zucker Rats (Crl:ZUC-Lepr fa). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7342451. [PMID: 30534350 PMCID: PMC6252239 DOI: 10.1155/2018/7342451] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/10/2018] [Indexed: 12/24/2022]
Abstract
Background The main factor characteristic for low-grade systemic inflammation typical for obesity is oxidative stress (OS). Reactive oxygen species (ROS) production is higher and more increased in time in the obese patients than in lean subjects. Aims To assess the effect of ileal transposition (IT) and sham types of bariatric procedures on the antioxidative systems in the liver tissue of Zucker rats (Crl:ZUC Leprfa). Method 21 animals were divided into the experimental groups: control group (n = 7), sham group (n = 7), and IT group (n = 7). Sham and IT animals underwent selected surgery. The concentration of total antioxidant capacity (TAC), total antioxidant status (TOS), and activity of glutathione reductase (glutathione-disulfide reductase, GR, GSR), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), and total superoxide dismutase activity (SOD) were assessed in liver tissue 3 months after surgery. Results IT procedure significantly increased TAC when compared to sham and the control group. Animals after IT showed higher levels of TOS when compared to sham procedure. The total amount of TOS was similar in IT and control groups. GPx activity was increased in the groups submitted to the sham and IT surgery in relation to control. GR and CAT activities were significantly higher after IT in comparison to control and sham procedures. Total SOD and MnSOD were significantly higher in sham-operated animals in comparison to IT intervention and control groups. Conclusions IT procedure had a positive impact on the diminishing of oxidative stress measured by TAC and TOS markers. The dynamic, adaptive, and protective mechanisms of enzymatic antioxidant systems were observed after the IT but not sham procedure. Nevertheless, 3 months after surgery, the midterm effect of bariatric surgery was observed, which might not fully balance the antioxidative response.
Collapse
|
25
|
Oliva-Olivera W, Lhamyani S, Coín-Aragüez L, Alcaide-Torres J, Cardona F, El Bekay R, Tinahones FJ. Involvement of acetyl-CoA-producing enzymes in the deterioration of the functional potential of adipose-derived multipotent cells from subjects with metabolic syndrome. Metabolism 2018; 88:12-21. [PMID: 30172756 DOI: 10.1016/j.metabol.2018.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The expansion capacity of white adipose tissue influences the distribution of fat depots in the body, the visceral accumulation of which is linked to metabolic syndrome, regardless of the degree of obesity of the subjects. Alterations in the adipose tissue-derived mesenchymal stem cells (ASCs) may contribute to the adipose tissue remodeling associated with metabolic syndrome and impact the regional distribution of adipose tissue by generating inherently dysfunctional adipocytes. Here we examine the expression levels of acetyl-CoA-producing enzymes and their relationship with the lipogenic, antioxidant and oxidative potential of adipocytes generated from visceral ASCs (adipo-visASCs) and subcutaneous ASCs (adipo-subASCs) from subjects with different metabolic profiles. MATERIALS/METHODS Paired samples of visceral and subcutaneous adipose tissue were processed to isolate the respective ASCs from normal-weight (Nw) subjects and obese patients with metabolic syndrome (METS) and without METS (NonMETS). qPCR was used to quantify the expression levels of the genes studied in both adipo-ASCs from the patient groups and those generated after silencing by small interfering RNA of acetyl-CoA-producing enzymes. The accumulation of lipids was quantified by absorbance. RESULTS No significant differences in cell yield or CD34+CD31-CD45- ASC percentage were observed between the different patient groups. Unlike adipo-visASCs, adipo-subASCs from METS patients showed a decrease in expression levels of acetyl-CoA-producing enzymes as well as proteins linked to lipogenesis, antioxidant defense and fatty acid oxidation. Transcriptional silencing of acetyl-CoA-producing enzymes in adipo-subASCs reduced lipid accumulation and affected transcription levels of lipogenic and antioxidant defense proteins. CONCLUSIONS Adipo-subASCs may be more susceptible than adipo-visASCs to deterioration of the lipogenic, oxidative and antioxidant potential associated with metabolic syndrome. Intrinsic alterations in transcription levels of acetyl-CoA-producing enzymes may contribute to the metabolic reprogramming of adipo-subASCs from METS patients.
Collapse
Affiliation(s)
- Wilfredo Oliva-Olivera
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Spain; Pathophysiology of Obesity and Nutrition, CIBEROBN, Institute of Health Carlos III, ISCIII, Spain.
| | - Said Lhamyani
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Spain; Pathophysiology of Obesity and Nutrition, CIBEROBN, Institute of Health Carlos III, ISCIII, Spain
| | - Leticia Coín-Aragüez
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Spain; Pathophysiology of Obesity and Nutrition, CIBEROBN, Institute of Health Carlos III, ISCIII, Spain
| | - Juan Alcaide-Torres
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Spain; Pathophysiology of Obesity and Nutrition, CIBEROBN, Institute of Health Carlos III, ISCIII, Spain
| | - Fernando Cardona
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Spain; Pathophysiology of Obesity and Nutrition, CIBEROBN, Institute of Health Carlos III, ISCIII, Spain
| | - Rajaa El Bekay
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Spain; Pathophysiology of Obesity and Nutrition, CIBEROBN, Institute of Health Carlos III, ISCIII, Spain.
| | - Francisco J Tinahones
- Department of Clinical Endocrinology and Nutrition, Institute of Biomedical Research of Málaga (IBIMA), Hospital of Málaga (Virgen de la Victoria), University of Málaga (UMA), Spain; Pathophysiology of Obesity and Nutrition, CIBEROBN, Institute of Health Carlos III, ISCIII, Spain.
| |
Collapse
|
26
|
Gut adaptation after metabolic surgery and its influences on the brain, liver and cancer. Nat Rev Gastroenterol Hepatol 2018; 15:606-624. [PMID: 30181611 DOI: 10.1038/s41575-018-0057-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metabolic surgery is the best treatment for long-term weight loss maintenance and comorbidity control. Metabolic operations were originally intended to change anatomy to alter behaviour, but we now understand that the anatomical changes can modulate physiology to change behaviour. They are no longer considered only mechanically restrictive and/or malabsorptive procedures; rather, they are considered metabolic procedures involving complex physiological changes, whereby gut adaptation influences signalling pathways in several other organs, including the liver and the brain, regulating hunger, satiation, satiety, body weight, glucose metabolism and immune functions. The integrative physiology of gut adaptation after these operations consists of a complex mechanistic web of communication between gut hormones, bile acids, gut microbiota, the brain and both enteric and central nervous systems. The understanding of nutrient sensing via enteroendocrine cells, the enteric nervous system, hypothalamic peptides and adipose tissue and of the role of inflammation has advanced our knowledge of this integrative physiology. In this Review, we focus on the adaptation of gut physiology to the anatomical alterations from Roux-en-Y gastric bypass and vertical sleeve gastrectomy and the influence of these procedures on food intake, weight loss, nonalcoholic fatty liver disease (NAFLD) and cancer. We also aim to demonstrate the underlying mechanisms that could explain how metabolic surgery could be used as a therapeutic option in NAFLD and certain obesity-related cancers.
Collapse
|
27
|
Lu Z, Wei X, Sun F, Zhang H, Gao P, Pu Y, Wang A, Chen J, Tong W, Li Q, Zhou X, Yan Z, Zheng H, Yang G, Huang Y, Liu D, Zhu Z. Non-insulin determinant pathways maintain glucose homeostasis upon metabolic surgery. Cell Discov 2018; 4:58. [PMID: 30275974 PMCID: PMC6155125 DOI: 10.1038/s41421-018-0062-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/13/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022] Open
Abstract
Insulin is critical for glucose homeostasis, and insulin deficiency or resistance leads to the development of diabetes. Recent evidence suggests that diabetes can be remitted independent of insulin. However, the underlying mechanism remains largely elusive. In this study, we utilized metabolic surgery as a tool to identify the non-insulin determinant mechanism. Here, we report that the most common metabolic surgery, Roux-en-Y gastric bypass (RYGB), reduced insulin production but persistently maintained euglycemia in healthy Sprague-Dawley (SD) rats and C57 mice. This reduction in insulin production was associated with RYGB-mediated inhibition of pancreatic preproinsulin and polypyrimidine tract-binding protein 1. In addition, RYGB also weakened insulin sensitivity that was evaluated by hyperinsulinemic-euglycemic clamp test and downregulated signaling pathways in insulin-sensitive tissues. The mechanistic evidence suggests that RYGB predominately shifted the metabolic profile from glucose utilization to fatty acid oxidation, enhanced the energy expenditure and activated multiple metabolic pathways through reducing gut energy uptake. Importantly, the unique effect of RYGB was extended to rats with islet disruption and patients with type 2 diabetes. These results demonstrate that compulsory rearrangement of the gastrointestinal tract can initiate non-insulin determinant pathways to maintain glucose homeostasis. Based on the principle of RYGB action, the development of a noninvasive intervention of the gastrointestinal tract is a promising therapeutic route to combat disorders characterized by energy metabolism dysregulation.
Collapse
Affiliation(s)
- Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Xiao Wei
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Hexuan Zhang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Yunfei Pu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Anlong Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Jing Chen
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Weidong Tong
- Department of Gastrointestinal Metabolic Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Qiang Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Xunmei Zhou
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 China
| | - Yu Huang
- Institute of Vascular Medicine and School of Biomedical Sciences, Chinese University of Hong Kong, BMSB315, Shatin, Hong Kong 00852 China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| |
Collapse
|
28
|
Desjardins EM, Steinberg GR. Emerging Role of AMPK in Brown and Beige Adipose Tissue (BAT): Implications for Obesity, Insulin Resistance, and Type 2 Diabetes. Curr Diab Rep 2018; 18:80. [PMID: 30120579 DOI: 10.1007/s11892-018-1049-6] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The global prevalence of type 2 diabetes (T2D) is escalating at alarming rates, demanding the development of additional classes of therapeutics to further reduce the burden of disease. Recent studies have indicated that increasing the metabolic activity of brown and beige adipose tissue may represent a novel means to reduce circulating glucose and lipids in people with T2D. The AMP-activated protein kinase (AMPK) is a cellular energy sensor that has recently been demonstrated to be important in potentially regulating the metabolic activity of brown and beige adipose tissue. The goal of this review is to summarize recent work describing the role of AMPK in brown and beige adipose tissue, focusing on its role in adipogenesis and non-shivering thermogenesis. RECENT FINDINGS Ablation of AMPK in mouse adipocytes results in cold intolerance, a reduction in non-shivering thermogenesis in brown adipose tissue (BAT), and the development of non-alcoholic fatty liver disease (NAFLD) and insulin resistance; effects associated with a defect in mitochondrial specific autophagy (mitophagy) within BAT. The effects of a β3-adrenergic agonist on the induction of BAT thermogenesis and the browning of white adipose tissue (WAT) are also blunted in mice lacking adipose tissue AMPK. A specific AMPK activator, A-769662, also results in the activation of BAT and the browning of WAT, effects which may involve demethylation of the PR domain containing 16 (Prdm16) promoter region, which is important for BAT development. AMPK plays an important role in the development and maintenance of brown and beige adipose tissue. Adipose tissue AMPK is reduced in people with insulin resistance, consistent with findings that mice lacking adipocyte AMPK develop greater NAFLD and insulin resistance. These data suggest that pharmacologically targeting adipose tissue AMPK may represent a promising strategy to enhance energy expenditure and reduce circulating glucose and lipids, which may be effective for the treatment of NAFLD and T2D.
Collapse
Affiliation(s)
- Eric M Desjardins
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8N 3Z5, Canada.
| |
Collapse
|
29
|
Billeter AT, de la Garza Herrera JR, Scheurlen KM, Nickel F, Billmann F, Müller-Stich BP. MANAGEMENT OF ENDOCRINE DISEASE: Which metabolic procedure? Comparing outcomes in sleeve gastrectomy and Roux-en Y gastric bypass. Eur J Endocrinol 2018; 179:R77-R93. [PMID: 29764908 DOI: 10.1530/eje-18-0009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/14/2018] [Indexed: 12/15/2022]
Abstract
Obesity and its associated comorbidities have become one of the largest challenges for health care in the near future. Conservative therapy for obesity and related comorbidities has a very high failure rate and poor long-term results. Similarly, the conservative and medical management of the majority of metabolic diseases such as type 2 diabetes mellitus are only able to slow down disease progression but have no causal effect on the disease process. Obesity surgery has evolved as a highly effective therapy for severe obesity achieving long-lasting weight loss. Furthermore, several studies have demonstrated the beneficial effects of obesity surgery on reduction of overall mortality, reduction of cardiovascular events and superior control of obesity-related diseases such as type 2 diabetes mellitus, dyslipidemia and also the non-alcoholic steatohepatitis compared to medical therapy. Based on these findings, the term 'metabolic surgery' with the focus on treating metabolic diseases independent of body weight has been coined. Of great interest are recent studies that show that even existing complications of metabolic diseases such as diabetic nephropathy or the non-alcoholic steatohepatitis can be reversed by metabolic surgery. Although metabolic surgery has proven to be a safe and effective treatment for obesity, resolution of comorbidities and enhancing quality of life, it is still uncertain and unclear, which surgical procedure is the most effective to achieve these metabolic effects. The aim of this review is to compare the effects of the two currently most widely used metabolic operations, the Roux-en-Y gastric bypass and the sleeve gastrectomy in the treatment of obesity and its related comorbidities.
Collapse
Affiliation(s)
- Adrian T Billeter
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | - Katharina M Scheurlen
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Felix Nickel
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Franck Billmann
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Beat P Müller-Stich
- Department of General, Visceral, and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
30
|
The relation between pro-oxidant antioxidant balance and glycolipid profile, 6 months after gastric bypass surgery. Surg Obes Relat Dis 2018; 14:361-367. [DOI: 10.1016/j.soard.2017.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/16/2017] [Accepted: 12/03/2017] [Indexed: 12/28/2022]
|
31
|
Abstract
Currently, there are no effective preventive strategies for pancreatic cancer. Obesity has been increasingly recognized as a strong but modifiable risk factor of pancreatic cancer. In this article, we aim to review the literature regarding weight loss on prevention of pancreatic cancer. Epidemiological and laboratory studies have shown that obesity is associated with increased incidence of pancreatic cancer and potentially worse cancer outcome. Whereas the underlying pathomechanisms remain unclear, chronic inflammation, insulin resistance, and altered intestinal microbiota are all implicated in the carcinogenic effect of obesity. Weight loss, especially the durable and significant weight loss after bariatric surgery, has been shown to reduce the risks of multiple cancers and may become a good intervention for pancreatic cancer prevention.
Collapse
|
32
|
Arab Sadeghabadi Z, Nourbakhsh M, Pasalar P, Emamgholipour S, Golestani A, Larijani B, Razzaghy-Azar M. Reduced gene expression of sirtuins and active AMPK levels in children and adolescents with obesity and insulin resistance. Obes Res Clin Pract 2017; 12:167-173. [PMID: 29150224 DOI: 10.1016/j.orcp.2017.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/18/2017] [Accepted: 10/10/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Sirtuins, including SIRT1 and SIRT2, are longevity-associated deacetylase enzymes that modulate metabolic homeostasis in response to the cellular energy state. Adenosine monophosphate activated protein kinase (AMPK) and SIRT1 are interrelated and share several common target pathways. This study aimed to evaluate the SIRT1 and SIRT2 gene expression in peripheral blood mononuclear cells (PBMCs) as well as plasma levels of AMPK, in obese children and adolescents. MATERIALS AND METHODS Participants included 60 children and adolescents (30 obese and 30 age- and gender-matched control subjects). Real-time polymerase chain reaction (PCR) was used to assess the SIRT1 and SIRT2 gene expression in PBMCs. Serum phospho-AMPK and insulin were measured using enzyme-linked immunosorbent assay (ELISA), and insulin resistance (IR) was calculated by the Homeostasis Model of Assessment of Insulin Resistance (HOMA-IR). Glucose and lipid profile were also measured. RESULTS SIRT1 gene expression and phospho-AMPK plasma levels were significantly diminished in obese subjects compared to the control group, and both SIRT1 and SIRT2 were significantly lower in obese children with IR compared to those without IR. SIRT1 expression revealed significant negative correlations with body mass index and waist circumference as well as insulin and HOMA-IR and a positive correlation with AMPK. SIRT2 negatively correlated with SIRT1 and positively correlated with high density lipoprotein-cholesterol (HDL-C). CONCLUSION SIRT1 and SIRT2 expression and AMPK levels decrease in children with obesity and IR. Targeting SIRT1 can be valuable in preventing obesity-associated IR in childhood and adolescence.
Collapse
Affiliation(s)
- Zahra Arab Sadeghabadi
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, 1449614535 Tehran, Iran.
| | - Parvin Pasalar
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solaleh Emamgholipour
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Golestani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Razzaghy-Azar
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran; H. Aliasghar Children's Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Abstract
The AMP-activated protein kinase (AMPK) is a central regulator of multiple metabolic pathways and may have therapeutic importance for treating obesity, insulin resistance, type 2 diabetes (T2D), non-alcoholic fatty liver disease (NAFLD), and cardiovascular disease (CVD). Given the ubiquitous expression of AMPK, it has been a challenge to evaluate which tissue types may be most beneficially poised for mediating the positive metabolic effects of AMPK-centered treatments. In this review we evaluate the metabolic phenotypes of transgenic mouse models in which AMPK expression and function have been manipulated, and the impact this has on controlling lipid metabolism, glucose homeostasis, and inflammation. This information may be useful for guiding the development of AMPK-targeted therapeutics to treat chronic metabolic diseases.
Collapse
Affiliation(s)
- Emily A Day
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada
| | - Rebecca J Ford
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada
| | - Gregory R Steinberg
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON, Canada.
| |
Collapse
|
34
|
Billeter AT, Vittas S, Israel B, Scheurlen KM, Hidmark A, Fleming TH, Kopf S, Büchler MW, Müller-Stich BP. Gastric bypass simultaneously improves adipose tissue function and insulin-dependent type 2 diabetes mellitus. Langenbecks Arch Surg 2017; 402:901-910. [DOI: 10.1007/s00423-017-1601-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/29/2017] [Indexed: 01/06/2023]
|
35
|
Does Bariatric Surgery Improve Obesity Associated Comorbid Conditions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:545-570. [PMID: 28585216 DOI: 10.1007/978-3-319-48382-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Obesity is a constantly growing health problem which reduces quality of life and life expectancy. Bariatric surgery for obesity is taken into account when all other conservative treatment modalities have failed. Comparison of the multidisciplinary programs with bariatric surgery regarding to weight loss showed that substantial and durable weight reduction have been achieved only with bariatric surgical treatments. However, the benefits of weight loss following bariatric procedures are still debated regarding the pro-inflammatory and metabolic profile of obesity.
Collapse
|
36
|
Hagman DK, Larson I, Kuzma JN, Cromer G, Makar K, Rubinow KB, Foster-Schubert KE, van Yserloo B, Billing PS, Landerholm RW, Crouthamel M, Flum DR, Cummings DE, Kratz M. The short-term and long-term effects of bariatric/metabolic surgery on subcutaneous adipose tissue inflammation in humans. Metabolism 2017; 70:12-22. [PMID: 28403936 PMCID: PMC5407411 DOI: 10.1016/j.metabol.2017.01.030] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/25/2017] [Accepted: 01/28/2017] [Indexed: 12/22/2022]
Abstract
CONTEXT The mechanisms mediating the short- and long-term improvements in glucose homeostasis following bariatric/metabolic surgery remain incompletely understood. OBJECTIVE To investigate whether a reduction in adipose tissue inflammation plays a role in the metabolic improvements seen after bariatric/metabolic surgery, both in the short-term and longer-term. DESIGN Fasting blood and subcutaneous abdominal adipose tissue were obtained before (n=14), at one month (n=9), and 6-12months (n=14) after bariatric/metabolic surgery from individuals with obesity who were not on insulin or anti-diabetes medication. Adipose tissue inflammation was assessed by a combination of whole-tissue gene expression and flow cytometry-based quantification of tissue leukocytes. RESULTS One month after surgery, body weight was reduced by 13.5±4.4kg (p<0.001), with improvements in glucose tolerance reflected by a decrease in area-under-the-curve (AUC) glucose in 3-h oral glucose tolerance tests (-105±98mmol/L * min; p=0.009) and enhanced pancreatic β-cell function (insulinogenic index: +0.8±0.9pmol/mmol; p=0.032), but no change in estimated insulin sensitivity (Matsuda insulin sensitivity index [ISI]; p=0.720). Furthermore, although biomarkers of systemic inflammation and pro-inflammatory gene expression in adipose tissue remained unchanged, the number of neutrophils increased in adipose tissue 15-20 fold (p<0.001), with less substantial increases in other leukocyte populations. By the 6-12month follow-up visit, body weight was reduced by 34.8±10.8kg (p<0.001) relative to baseline, and glucose tolerance was further improved (AUC glucose -276±229; p<0.001) along with estimated insulin sensitivity (Matsuda ISI: +4.6±3.2; p<0.001). In addition, improvements in systemic inflammation were reflected by reductions in circulating C-reactive protein (CRP; -2.0±5.3mg/dL; p=0.002), and increased serum adiponectin (+1358±1406pg/mL; p=0.003). However, leukocyte infiltration of adipose tissue remained elevated relative to baseline, with pro-inflammatory cytokine mRNA expression unchanged, while adiponectin mRNA expression trended downward (p=0.069). CONCLUSION Both the short- and longer-term metabolic improvements following bariatric/metabolic surgery occur without significant reductions in measures of adipose tissue inflammation, as assessed by measuring the expression of genes encoding key mediators of inflammation and by flow cytometric immunophenotyping and quantification of adipose tissue leukocytes.
Collapse
Affiliation(s)
- Derek K Hagman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Diabetes Research Center, University of Washington, Seattle, WA 98195, USA
| | - Ilona Larson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jessica N Kuzma
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Gail Cromer
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Karen Makar
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Katya B Rubinow
- Department of Medicine, Division of Metabolism, Endocrinology & Nutrition, University of Washington, Seattle, WA 98195, USA
| | - Karen E Foster-Schubert
- Department of Medicine, Division of Metabolism, Endocrinology & Nutrition, University of Washington, Seattle, WA 98195, USA
| | - Brian van Yserloo
- Department of Surgery, University of Washington, Seattle, WA 98195, USA
| | | | | | | | - David R Flum
- Department of Surgery, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| | - David E Cummings
- Department of Medicine, Division of Metabolism, Endocrinology & Nutrition, University of Washington, Seattle, WA 98195, USA
| | - Mario Kratz
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Medicine, Division of Metabolism, Endocrinology & Nutrition, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA; Department of Epidemiology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
37
|
Prior SL, Barry JD, Caplin S, Min T, Grant DA, Stephens JW. Temporal changes in plasma markers of oxidative stress following laparoscopic sleeve gastrectomy in subjects with impaired glucose regulation. Surg Obes Relat Dis 2017; 13:162-168. [DOI: 10.1016/j.soard.2016.08.501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/17/2016] [Accepted: 08/28/2016] [Indexed: 01/11/2023]
|
38
|
Mancini SJ, White AD, Bijland S, Rutherford C, Graham D, Richter EA, Viollet B, Touyz RM, Palmer TM, Salt IP. Activation of AMP-activated protein kinase rapidly suppresses multiple pro-inflammatory pathways in adipocytes including IL-1 receptor-associated kinase-4 phosphorylation. Mol Cell Endocrinol 2017; 440:44-56. [PMID: 27840174 PMCID: PMC5228585 DOI: 10.1016/j.mce.2016.11.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/29/2022]
Abstract
Inflammation of adipose tissue in obesity is associated with increased IL-1β, IL-6 and TNF-α secretion and proposed to contribute to insulin resistance. AMP-activated protein kinase (AMPK) regulates nutrient metabolism and is reported to have anti-inflammatory actions in adipose tissue, yet the mechanisms underlying this remain poorly characterised. The effect of AMPK activation on cytokine-stimulated proinflammatory signalling was therefore assessed in cultured adipocytes. AMPK activation inhibited IL-1β-stimulated CXCL10 secretion, associated with reduced interleukin-1 receptor associated kinase-4 (IRAK4) phosphorylation and downregulated MKK4/JNK and IKK/IκB/NFκB signalling. AMPK activation inhibited TNF-α-stimulated IKK/IκB/NFκB signalling but had no effect on JNK phosphorylation. The JAK/STAT3 pathway was also suppressed by AMPK after IL-6 stimulation and during adipogenesis. Adipose tissue from AMPKα1-/- mice exhibited increased JNK and STAT3 phosphorylation, supporting suppression of these distinct proinflammatory pathways by AMPK in vivo. The inhibition of multiple pro-inflammatory signalling pathways by AMPK may underlie the reported beneficial effects of AMPK activation in adipose tissue.
Collapse
Affiliation(s)
- Sarah J Mancini
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Anna D White
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Silvia Bijland
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Claire Rutherford
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, France
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Timothy M Palmer
- School of Pharmacy, University of Bradford, Bradford, West Yorkshire, BD7 1DP, United Kingdom
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, United Kingdom.
| |
Collapse
|
39
|
Bettaieb A, Cremonini E, Kang H, Kang J, Haj FG, Oteiza PI. Anti-inflammatory actions of (-)-epicatechin in the adipose tissue of obese mice. Int J Biochem Cell Biol 2016; 81:383-392. [PMID: 27592457 PMCID: PMC5514545 DOI: 10.1016/j.biocel.2016.08.044] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 12/19/2022]
Abstract
Obesity and type 2 diabetes (T2D) are major public health concerns. Visceral adipose tissue inflammation is considered a significant contributor to obesity-associated T2D development. We previously showed that the flavan-3-ol (-)-epicatechin (EC) can mitigate insulin resistance in mice fed a high fat diet (HFD). This study investigated the capacity of EC to inhibit visceral adipose tissue inflammation occurring as a consequence of HFD consumption in C57BL/6J mice, and characterized the underlying mechanisms. In association with the development of obesity and insulin resistance, HFD consumption caused inflammation in the visceral adipose tissue as evidenced by activation of the pro-inflammatory transcription factor NF-κB and increased tissue levels of the macrophage marker F4/80, tumor necrosis factor alpha (TNFα), and the chemokine MCP-1. EC supplementation mitigated all these events. In addition, we observed activation of the three branches of the unfolded protein response (UPR), and upregulation of NADPH oxidases NOX4 and NOX2 in visceral fat of mice fed HFD. These can account, at least in part, for the associated oxidative stress and activation of the redox sensitive NF-κB. Notably, EC supplementation mitigated this and the release of pro-inflammatory proteins from metabolically stressed adipocytes. Attenuation of adipocyte endoplasmic reticulum (ER) and oxidative stress by EC could contribute to decreased inflammation and improved visceral adipose tissue insulin sensitivity. Our results support the concept that consumption of EC-rich foods could mitigate obesity-associated insulin resistance through attenuation of adipose tissue inflammation.
Collapse
Affiliation(s)
- Ahmed Bettaieb
- Department of Nutrition, University of Tennessee-Knoxville, Knoxville, TN, USA
| | - Eleonora Cremonini
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Heeteak Kang
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Jiye Kang
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA
| | - Fawaz G Haj
- Department of Nutrition, University of California, Davis, USA; Department of Internal Medicine, University of California, Davis, USA
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, USA; Department of Environmental Toxicology, University of California, Davis, USA.
| |
Collapse
|
40
|
Backes CF, Lopes E, Tetelbom A, Heineck I. Medication and nutritional supplement use before and after bariatric surgery. SAO PAULO MED J 2016; 134:0. [PMID: 27812597 PMCID: PMC11448729 DOI: 10.1590/1516-3180.2015.0241030516] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 11/23/2015] [Accepted: 05/03/2016] [Indexed: 01/01/2023] Open
Abstract
CONTEXT AND OBJECTIVE: Bariatric surgery has been an effective alternative treatment for morbid obesity and has resulted in decreased mortality, better control over comorbidities and reduced use of drugs. The objective of this study was to analyze the impact of bariatric surgery on medication drug and nutritional supplement use. DESIGN AND SETTING: Longitudinal study of before-and-after type, on 69 morbidly obese patients in a public hospital in Porto Alegre. METHODS: Through interviews, the presence of comorbidities and use of drugs with and without prescription were evaluated. RESULTS: Among the 69 patients interviewed, 85.5% had comorbidities in the preoperative period, with an average of 2.3 (± 1.5) per patient. The main comorbidities reported were hypertension, diabetes and dyslipidemia. 84.1% of the patients were using prescribed drugs in the preoperative period. The mean drug use per patient was 4.8, which decreased to 4.4 after the procedure. The surgery enabled significant reduction in use of most antidiabetic (84%), antilipemic (77%) and antihypertensive drugs (49.5%). On the other hand, there was a significant increase in use of multivitamins and drugs for disorders of the gastrointestinal tract. The dosages of most of the drugs that continued to be prescribed after surgery were decreased, but not significantly. CONCLUSION: After bariatric surgery, there were increases in the use of vitamins, gastric antisecretory drugs and antianemic drugs. Nevertheless, there was an overall reduction in drug use during this period, caused by suspension of drugs or dose reduction.
Collapse
Affiliation(s)
- Charline Fernanda Backes
- Master’s Student in the Postgraduate Pharmaceutical Sciences Program, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Edyane Lopes
- PhD. Pharmacist, School of Public Health, Health Department of the State of Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Airton Tetelbom
- MD. Coordinator of the Health Technology Assessment Center, Grupo Hospitalar Conceição; Head Professor of Public Health, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA); Associate Professor of Public Health, Universidade Luterana do Brasil (ULBRA); and Contributing Professor in the Postgraduate Epidemiology Program, Department of Social Medicine, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Isabela Heineck
- PhD. Associate Professor, Postgraduate Pharmaceutical Sciences Program and Postgraduate Pharmaceutical Services, School of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
41
|
Short-term caloric restriction in db/db mice improves myocardial function and increases high molecular weight (HMW) adiponectin. ACTA ACUST UNITED AC 2016; 13:28-34. [PMID: 27942464 DOI: 10.1016/j.ijcme.2016.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Obesity and metabolic syndrome lead to the development of metabolic heart disease (MHD) that is characterized by left ventricular hypertrophy (LVH), diastolic dysfunction, and increased mitochondrial ROS. Caloric restriction (CR) is a nutritional intervention that protects against obesity, diabetes, and cardiovascular disease. Healthy adipose tissue is cardioprotective via releasing adipokines such as adiponectin. We tested the hypothesis that CR can ameliorate MHD and it is associated with improved adipose tissue function as reflected by increased circulating levels of high molecular weight (HMW) adiponectin and AMP-activated protein kinase (AMPK) in db/db mice. METHODS Genetically obese db/db and lean db/+ male mice were fed either ad libitum or subjected to 30% CR for 5 weeks. At the end of the study period, echocardiography was carried out to assess diastolic function. Blood, heart, and epididymal fat pads were harvested for mitochondrial study, ELISA, and Western blot analyses. RESULTS CR reversed the development of LVH, prevented diastolic dysfunction, and decreased cardiac mitochondrial H2O2 in db/db (vs. ad lib) mice. These beneficial effects on the heart were associated with increased circulating level of HMW adiponectin. Furthermore, CR increased AMPK and eNOS activation in white adipose tissue of db/db mice, but not in the heart. CONCLUSIONS These findings indicate that even short-term CR protects the heart from MHD. Whether the beneficial effects of CR on the heart could be related to the improved adipose tissue function warrants future investigation.
Collapse
|
42
|
Mach L, Bedanova H, Soucek M, Karpisek M, Nemec P, Orban M. Tobacco smoking and cytokine levels in human epicardial adipose tissue: Impact of smoking cessation. Atherosclerosis 2016; 255:37-42. [PMID: 27816807 DOI: 10.1016/j.atherosclerosis.2016.10.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/28/2016] [Accepted: 10/12/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND & AIMS Epicardial adipose tissue (EAT) is a source of a number of cytokines which could act in the pathogenesis of coronary artery disease (CAD). The potential relationship between known cardiovascular risk factors, such as smoking, dyslipidaemia or diabetes mellitus and EAT humoral signalling, has not been fully elucidated. Therefore, we designed and conducted a cross-sectional study to determine whether selected cardiovascular risk factors are linked to levels of cytokines in epicardial and subcutaneous adipose tissue (SAT). METHODS Samples of SAT and EAT were collected from consecutive patients undergoing scheduled cardiac surgery. Tissue concentrations of tumour necrosis factor-ɑ (TNF-α), interleukin-6 (IL-6), adipocyte fatty acid-binding protein, leptin, and adiponectin were determined by ELISA. RESULTS We enrolled 140 patients. TNF-α and IL-6 concentrations in EAT and SAT were significantly higher in current smokers (CS) than in never smokers (NS) and former smokers (FS). There were no differences between FS and NS. No other clinical variables were associated with cytokine concentrations in a regression analysis. CONCLUSIONS Smoking was independently associated with higher TNF-α and IL-6 concentrations in EAT and SAT. A novel observation that pro-inflammatory cytokines are elevated in EAT in smokers could contribute to identify potential mechanisms involved in the pathogenesis of adverse effects of tobacco smoking. There were no differences between EAT cytokine production in NS and FS, which support the importance of smoking cessation for cardiovascular risk reduction.
Collapse
Affiliation(s)
- Lukas Mach
- International Clinical Research Centre, St. Anne's University Hospital Brno, Pekarska 53, Brno, 602 00, Czech Republic; University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, SO15 6YD, United Kingdom
| | - Helena Bedanova
- International Clinical Research Centre, St. Anne's University Hospital Brno, Pekarska 53, Brno, 602 00, Czech Republic; Centre of Cardiovascular Surgery and Organ Transplantation Brno, Pekarska 53, Brno, 602 00, Czech Republic
| | - Miroslav Soucek
- International Clinical Research Centre, St. Anne's University Hospital Brno, Pekarska 53, Brno, 602 00, Czech Republic
| | - Michal Karpisek
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho tr. 1, Brno, 612 42, Czech Republic
| | - Petr Nemec
- International Clinical Research Centre, St. Anne's University Hospital Brno, Pekarska 53, Brno, 602 00, Czech Republic; Centre of Cardiovascular Surgery and Organ Transplantation Brno, Pekarska 53, Brno, 602 00, Czech Republic
| | - Marek Orban
- International Clinical Research Centre, St. Anne's University Hospital Brno, Pekarska 53, Brno, 602 00, Czech Republic; Centre of Cardiovascular Surgery and Organ Transplantation Brno, Pekarska 53, Brno, 602 00, Czech Republic; Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Palackeho tr. 1, Brno, 612 42, Czech Republic.
| |
Collapse
|
43
|
Zhang X, Yu B, Yang D, Qiao Z, Cao T, Zhang P. Gastric volume reduction is essential for the remission of type 2 diabetes mellitus after bariatric surgery in nonobese rats. Surg Obes Relat Dis 2016; 12:1569-1576. [PMID: 27425832 DOI: 10.1016/j.soard.2016.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 04/18/2016] [Indexed: 01/19/2023]
|
44
|
Frikke-Schmidt H, O'Rourke RW, Lumeng CN, Sandoval DA, Seeley RJ. Does bariatric surgery improve adipose tissue function? Obes Rev 2016; 17:795-809. [PMID: 27272117 PMCID: PMC5328428 DOI: 10.1111/obr.12429] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/25/2016] [Accepted: 04/20/2016] [Indexed: 12/19/2022]
Abstract
Bariatric surgery is currently the most effective treatment for obesity. Not only do these types of surgeries produce significant weight loss but also they improve insulin sensitivity and whole body metabolic function. The aim of this review is to explore how altered physiology of adipose tissue may contribute to the potent metabolic effects of some of these procedures. This includes specific effects on various fat depots, the function of individual adipocytes and the interaction between adipose tissue and other key metabolic tissues. Besides a dramatic loss of fat mass, bariatric surgery shifts the distribution of fat from visceral to the subcutaneous compartment favoring metabolic improvement. The sensitivity towards lipolysis controlled by insulin and catecholamines is improved, adipokine secretion is altered and local adipose inflammation as well as systemic inflammatory markers decreases. Some of these changes have been shown to be weight loss independent, and novel hypothesis for these effects includes include changes in bile acid metabolism, gut microbiota and central regulation of metabolism. In conclusion bariatric surgery is capable of improving aspects of adipose tissue function and do so in some cases in ways that are not entirely explained by the potent effect of surgery. © 2016 World Obesity.
Collapse
Affiliation(s)
| | - R W O'Rourke
- Department of Surgery, University of Michigan, Ann Arbor, USA
| | - C N Lumeng
- Department of Pediatrics, University of Michigan, Ann Arbor, USA
| | - D A Sandoval
- Department of Surgery, University of Michigan, Ann Arbor, USA
| | - R J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, USA
| |
Collapse
|
45
|
Mottillo EP, Desjardins EM, Crane JD, Smith BK, Green AE, Ducommun S, Henriksen TI, Rebalka IA, Razi A, Sakamoto K, Scheele C, Kemp BE, Hawke TJ, Ortega J, Granneman JG, Steinberg GR. Lack of Adipocyte AMPK Exacerbates Insulin Resistance and Hepatic Steatosis through Brown and Beige Adipose Tissue Function. Cell Metab 2016; 24:118-29. [PMID: 27411013 PMCID: PMC5239668 DOI: 10.1016/j.cmet.2016.06.006] [Citation(s) in RCA: 247] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/20/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
Abstract
Brown (BAT) and white (WAT) adipose tissues play distinct roles in maintaining whole-body energy homeostasis, and their dysfunction can contribute to non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes. The AMP-activated protein kinase (AMPK) is a cellular energy sensor, but its role in regulating BAT and WAT metabolism is unclear. We generated an inducible model for deletion of the two AMPK β subunits in adipocytes (iβ1β2AKO) and found that iβ1β2AKO mice were cold intolerant and resistant to β-adrenergic activation of BAT and beiging of WAT. BAT from iβ1β2AKO mice had impairments in mitochondrial structure, function, and markers of mitophagy. In response to a high-fat diet, iβ1β2AKO mice more rapidly developed liver steatosis as well as glucose and insulin intolerance. Thus, AMPK in adipocytes is vital for maintaining mitochondrial integrity, responding to pharmacological agents and thermal stress, and protecting against nutrient-overload-induced NAFLD and insulin resistance.
Collapse
Affiliation(s)
- Emilio P Mottillo
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Eric M Desjardins
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Justin D Crane
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Brennan K Smith
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Alex E Green
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Serge Ducommun
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Tora I Henriksen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Aida Razi
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Kei Sakamoto
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Camilla Scheele
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Bruce E Kemp
- St Vincent's Institute and Department of Medicine, University of Melbourne, Fitzroy, Victoria 3065, Australia; Mary MacKillop Institute for Health Research Australian Catholic University, Victoria Parade, Fitzroy, Victoria 3065, Australia
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - Joaquin Ortega
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit 48201, MI, USA
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St. W., Hamilton, Ontario L8N 3Z5, Canada.
| |
Collapse
|
46
|
Weikel KA, Ruderman NB, Cacicedo JM. Unraveling the actions of AMP-activated protein kinase in metabolic diseases: Systemic to molecular insights. Metabolism 2016; 65:634-645. [PMID: 27085772 PMCID: PMC4834453 DOI: 10.1016/j.metabol.2016.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 01/08/2016] [Accepted: 01/09/2016] [Indexed: 12/13/2022]
Abstract
AMP-activated protein kinase (AMPK) plays a critical role both in sensing and regulating cellular energy state. In experimental animals, its activation has been shown to reduce the risk of obesity and diabetes-related co-morbidities such as insulin resistance, the metabolic syndrome and atherosclerotic cardiovascular disease. However, in humans, AMPK activation alone often does not completely resolve these conditions. Thus, an improved understanding of AMPK action and regulation in metabolic and other diseases is needed. Herein, we provide a brief description of the enzymatic regulation of AMPK and review its role in maintaining energy homeostasis. We then discuss tissue-specific actions of AMPK that become distorted during such conditions as obesity, type 2 diabetes and certain cancers. Finally, we explore recent findings regarding the interactions of AMPK with mammalian target of rapamycin complex 1 and the lysosome and discuss how changes in these relationships during overnutrition may lead to AMPK dysfunction. A more thorough understanding of AMPK's molecular interactions during diseases of overnutrition may provide key insights for the development of AMPK-based combinatorial treatments for metabolic disease.
Collapse
Affiliation(s)
- Karen A Weikel
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, MA, 02118, USA.
| | - Neil B Ruderman
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, MA, 02118, USA
| | - José M Cacicedo
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, MA, 02118, USA
| |
Collapse
|
47
|
Premenopausal Obesity and Breast Cancer Growth Rates in a Rodent Model. Nutrients 2016; 8:214. [PMID: 27077880 PMCID: PMC4848683 DOI: 10.3390/nu8040214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/20/2022] Open
Abstract
Obese premenopausal women with breast cancer have poorer prognosis for long term survival, in part because their tumors are larger at the time of diagnosis than are found in normal weight women. Whether larger tumor mass is due to obesity-related barriers to detection or to effects on tumor biology is not known. This study used polygenic models for obesity and breast cancer to deconstruct this question with the objective of determining whether cell autonomous mechanisms contribute to the link between obesity and breast cancer burden. Assessment of the growth rates of 259 chemically induced mammary carcinomas from rats sensitive to dietary induced obesity (DS) and of 143 carcinomas from rats resistant (DR) to dietary induced obesity revealed that tumors in DS rats grew 1.8 times faster than in DR rats. This difference may be attributed to alterations in cell cycle machinery that permit more rapid tumor cell accumulation. DS tumors displayed protein expression patterns consistent with reduced G1/S checkpoint inhibition and a higher threshold of factors required for execution of the apoptotic cell death pathway. These mechanistic insights identify regulatory targets for life style modifications or pharmacological interventions designed to disrupt the linkage between obesity and tumor burden.
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Sensing of nutrients and microbes in the gut are fundamental processes necessary for life. This review aims to provide an overview of the basic background and new data on cellular nutrient, energy, and microbe sensors. RECENT FINDINGS The nutrient sensors 5' adenosine monophosphate-activated protein kinase, activating transcription factor 4 and mechanistic target of rapamycin (mTOR) are critical in control of the cell cycle. Recent data demonstrate their role in metabolic syndrome, in cell growth, and as therapeutic targets. Regulation of mTOR by the amino acids is the subject of intense investigation. Recent studies have further elucidated the exact mechanism of amino acid-dependent mTOR regulation. Pathogen recognition receptors (PRRs) are receptors that recognize conserved microbial molecules. New data demonstrate how lymphocyte-specific PRRs are necessary to maintain homeostasis. Moreover, new studies explore the role of PRRs in controlling the gut bacterial and fungal microbiome. SUMMARY Nutrient sensing molecules are central to cell growth and metabolism and are implicated in cancer and the metabolic syndrome. Regulation of nutrient sensors is complex, and may be amenable to therapeutic targeting. Microbial sensors play critical roles in homeostasis and maintenance of the gut fungal and bacterial microbiome.
Collapse
|