101
|
Sun J, He Y, Bai L, Wang Z, Cao Z, Shao Y, Zhao J. An Analysis of the Risk Factors for New-Onset Diabetes Mellitus After Liver Transplantation. Int J Gen Med 2021; 14:4783-4792. [PMID: 34466023 PMCID: PMC8402980 DOI: 10.2147/ijgm.s324462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/11/2021] [Indexed: 11/23/2022] Open
Abstract
Objective To investigate the risk factors related to new-onset diabetes mellitus (NODM) and the significance of IL-6. Methods A retrospective analysis was conducted on clinical data from 64 patients who received either a living donor liver transplantation or a donation after circulatory death from September 2013 to October 2020 and attended regular follow-up visits for six or more months. During follow-up, patients were randomized into groups and followed up until the completion of the study or the death of the patient. Results The incidence of NODM was 31.25% (n = 20). The median age in the NODM group was 52.15 years (p < 0.01). Age (OR = 1.089; 95% CI: 0.0211-0.1495, p = 0.003) and elevated preoperative IL-6 (OR = 1.122; 95% CI: 0.0619-0.1677, p = 0.029) were found to be independent risk factors for NODM. HBV-induced liver cirrhosis, warm ischemia time (WIT), body mass index (BMI), and high preoperative fasting blood glucose (FBG) were also found to be risk factors for NODM. The recipient had a higher risk of NODM if the donor had a high BMI and poor hepatic function. The concentrations of IL-6, procalcitonin (PCT), FBG, and tacrolimus (TAC) in the first month postoperatively were significantly higher in the NODM group than in the NO-NODM group. The survival rate of the patients was not affected by NODM. Conclusion HBV-induced liver cirrhosis, WIT, BMI, and high preoperative FBG levels are risk factors for NODM, and age and preoperative IL-6 levels are independent risk factors. To a certain extent, higher BMI and poor hepatic function had reference significance for the incidence of NODM. Patients with a high concentration of FBG, IL-6, and TAC in the first month postoperatively had an increased risk of suffering from NODM.
Collapse
Affiliation(s)
- Jushan Sun
- Department of Liver and Laparoscopic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, People's Republic of China
| | - Yibiao He
- Department of Liver and Laparoscopic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, People's Republic of China
| | - Lei Bai
- Department of Liver and Laparoscopic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, People's Republic of China
| | - Zhipeng Wang
- Department of Liver and Laparoscopic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, People's Republic of China
| | - Zhu Cao
- Department of Liver and Laparoscopic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, People's Republic of China
| | - Yingmei Shao
- Xinjiang Uyghur Autonomous Region Clinical Research Center for Echinococcosis and Hepatobiliary Diseases, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, People's Republic of China
| | - Jinming Zhao
- Department of Liver and Laparoscopic Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, People's Republic of China
| |
Collapse
|
102
|
Geronikolou SA, Pavlopoulou A, Cokkinos DV, Bacopoulou F, Chrousos GP. Polycystic οvary syndrome revisited: An interactions network approach. Eur J Clin Invest 2021; 51:e13578. [PMID: 33955010 DOI: 10.1111/eci.13578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/26/2021] [Accepted: 04/19/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND The polycystic ovary syndrome (PCOS) has genetic, epigenetic, metabolic and reproductive aspects, while its complex pathophysiology has not been conclusively deciphered. AIM The goal of this research was to screen the gene/gene products associated with PCOS and to predict any possible interactions with the highest possible fidelity. MATERIALS AND METHODS STRING v10.5 database and a confidence level of 0.7 were used. RESULTS A highly interconnected network of 48 nodes was created, where insulin (INS) appears to be the major hub. INS upstream and downstream defects were analysed and revealed that only the kisspeptin- and glucagon-coding genes were upstream of INS. CONCLUSION A metabolic dominance was inferred and discussed herein with its implications in puberty, obesity, infertility and cardiovascular function. This study, thus, may contribute to the resolution of a scientific conflict between the USA and EU definitions of the syndrome and/or provide a new P4 medicine approach.
Collapse
Affiliation(s)
- Styliani A Geronikolou
- Clinical, Translational & Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,University Research Institute of Maternal & Child Health & Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center (IBG), Izmir, Turkey.,Izmir International Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | - Dennis V Cokkinos
- Clinical, Translational & Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Flora Bacopoulou
- University Research Institute of Maternal & Child Health & Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Chrousos
- Clinical, Translational & Experimental Surgery Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,University Research Institute of Maternal & Child Health & Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
103
|
Kim JS, Galvão DA, Newton RU, Gray E, Taaffe DR. Exercise-induced myokines and their effect on prostate cancer. Nat Rev Urol 2021; 18:519-542. [PMID: 34158658 DOI: 10.1038/s41585-021-00476-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Exercise is recognized by clinicians in the field of clinical oncology for its potential role in reducing the risk of certain cancers and in reducing the risk of disease recurrence and progression; yet, the underlying mechanisms behind this reduction in risk are not fully understood. Studies applying post-exercise blood serum directly to various types of cancer cell lines provide insight that exercise might have a role in inhibiting cancer growth via altered soluble and cell-free blood contents. Myokines, which are cytokines produced by muscle and secreted into the bloodstream, might offer multiple benefits to cellular metabolism (such as a reduction in insulin resistance, improved glucose uptake and reduced adiposity), and blood myokine levels can be altered with exercise. Alterations in the levels of myokines such as IL-6, IL-15, IL-10, irisin, secreted protein acidic risk in cysteine (SPARC), myostatin, oncostatin M and decorin might exert a direct inhibitory effect on cancer growth via inhibiting proliferation, promoting apoptosis, inducing cell-cycle arrest and inhibiting the epithermal transition to mesenchymal cells. The association of insulin resistance, hyperinsulinaemia and hyperlipidaemia with obesity can create a tumour-favourable environment; exercise-induced myokines can manipulate this environment by regulating adipose tissue and adipocytes. Exercise-induced myokines also have a critical role in increasing cytotoxicity and the infiltration of immune cells into the tumour.
Collapse
Affiliation(s)
- Jin-Soo Kim
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Daniel A Galvão
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia. .,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.
| | - Robert U Newton
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Elin Gray
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Dennis R Taaffe
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
104
|
Žiberna L, Jenko-Pražnikar Z, Petelin A. Serum Bilirubin Levels in Overweight and Obese Individuals: The Importance of Anti-Inflammatory and Antioxidant Responses. Antioxidants (Basel) 2021; 10:antiox10091352. [PMID: 34572984 PMCID: PMC8472302 DOI: 10.3390/antiox10091352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
Obesity is a chronic condition involving low-grade inflammation and increased oxidative stress; thus, obese and overweight people have lower values of serum bilirubin. Essentially, bilirubin is a potent endogenous antioxidant molecule with anti-inflammatory, immunomodulatory, antithrombotic, and endocrine properties. This review paper presents the interplay between obesity-related pathological processes and bilirubin, with a focus on adipose tissue and adipokines. We discuss potential strategies to mildly increase serum bilirubin levels in obese patients as an adjunctive therapeutic approach.
Collapse
Affiliation(s)
- Lovro Žiberna
- Institute of Pharmacology and Experimental Toxicology, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia;
| | | | - Ana Petelin
- Faculty of Health Sciences, University of Primorska, SI-6310 Izola, Slovenia;
- Correspondence: ; Tel.: +386-5-66-2469
| |
Collapse
|
105
|
Kotikalapudi N, Sampath SJP, Sukesh Narayan S, R B, Nemani H, Mungamuri SK, Venkatesan V. The promise(s) of mesenchymal stem cell therapy in averting preclinical diabetes: lessons from in vivo and in vitro model systems. Sci Rep 2021; 11:16983. [PMID: 34417511 PMCID: PMC8379204 DOI: 10.1038/s41598-021-96121-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity (Ob) poses a significant risk factor for the onset of metabolic syndrome with associated complications, wherein the Mesenchymal Stem Cell (MSC) therapy shows pre-clinical success. Here, we explore the therapeutic applications of human Placental MSCs (P-MSCs) to address Ob-associated Insulin Resistance (IR) and its complications. In the present study, we show that intramuscular injection of P-MSCs homed more towards the visceral site, restored HOMA-IR and glucose homeostasis in the WNIN/GR-Ob (Ob-T2D) rats. P-MSC therapy was effective in re-establishing the dysregulated cytokines. We report that the P-MSCs activates PI3K-Akt signaling and regulates the Glut4-dependant glucose uptake and its utilization in WNIN/GR-Ob (Ob-T2D) rats compared to its control. Our data reinstates P-MSC treatment's potent application to alleviate IR and restores peripheral blood glucose clearance evidenced in stromal vascular fraction (SVF) derived from white adipose tissue (WAT) of the WNIN/GR-Ob rats. Gaining insights, we show the activation of the PI3K-Akt pathway by P-MSCs both in vivo and in vitro (palmitate primed 3T3-L1 cells) to restore the insulin sensitivity dysregulated adipocytes. Our findings suggest a potent application of P-MSCs in pre-clinical/Ob-T2D management.
Collapse
Affiliation(s)
- Nagasuryaprasad Kotikalapudi
- Division of Cell and Molecular Biology, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Samuel Joshua Pragasam Sampath
- Division of Cell and Molecular Biology, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Sinha Sukesh Narayan
- Division of Food Safety, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Bhonde R
- Department of Regenerative Medicine, Manipal Institute of Regenerative Medicine, GKVK Post, Bellary Road, Allalasandra, Yelahanka, Bangalore, 560065, India
- Dr. D. Y. Patil Vidyapeeth, Pune, 411018, India
| | - Harishankar Nemani
- Division of Animal Facility, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Sathish Kumar Mungamuri
- Division of Food Safety, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Vijayalakshmi Venkatesan
- Division of Cell and Molecular Biology, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India.
| |
Collapse
|
106
|
Vasyukova OV, Kasyanova YV, Okorokov PL, Bezlepkina OB. [Myokines and adipomyokines: inflammatory mediators or unique molecules of targeted therapy for obesity?]. ACTA ACUST UNITED AC 2021; 67:36-45. [PMID: 34533012 DOI: 10.14341/probl12779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/02/2021] [Accepted: 08/10/2021] [Indexed: 11/06/2022]
Abstract
Skeletal muscles make up about 25% of the total mass in children and more than 40% in adults. Studies of the last twenty years have shown that along with the main functions, muscle tissue has hormonal activity. It was found that myocytes are able to release signaling molecules-myokines. They act auto-and paracrine within the muscle, and at a high level-through the systemic circulation, carrying out interactions between skeletal muscles and various organs and tissues, such as the liver, bone and adipose tissue, the brain. It is proved that the key factor in the expression of myokines is physical activity, and their level largely depends on physical fitness, the amount of skeletal muscle mass and its composition (the ratio of fast and slow fibers), on the intensity and duration of physical activity. Myokines have a wide range of physiological effects: myostatin suppresses the growth and differentiation of muscle tissue, and decorin, acting as its antagonist, promotes muscle hypertrophy. Interleukin 6 provides an energy substrate for contracting muscle fibers, fibroblast growth factor 21 activates the mechanisms of energy production during fasting and improves tissue sensitivity to insulin; irisin stimulates thermogenesis, glucose uptake by myocytes, and also contributes to an increase in bone mineral density. The study of myokines is one of the key links in understanding the mechanisms underlying obesity and metabolic complications, the consequences of a sedentary lifestyle, as well as the implementation of the action of physical activity. Taking into account the physiological effects of myokines in the body, in the future they can become therapeutic targets for the treatment of these conditions.
Collapse
|
107
|
Cunningham AL, Stephens JW, Harris DA. Gut microbiota influence in type 2 diabetes mellitus (T2DM). Gut Pathog 2021; 13:50. [PMID: 34362432 PMCID: PMC8343927 DOI: 10.1186/s13099-021-00446-0] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/25/2021] [Indexed: 12/12/2022] Open
Abstract
A strong and expanding evidence base supports the influence of gut microbiota in human metabolism. Altered glucose homeostasis is associated with altered gut microbiota, and is clearly associated with the development of type 2 diabetes mellitus (T2DM) and associated complications. Understanding the causal association between gut microbiota and metabolic risk has the potential role of identifying susceptible individuals to allow early targeted intervention.
Collapse
Affiliation(s)
- A L Cunningham
- Department of Surgery, Swansea Bay University Health Board, Singleton Hospital, Swansea, SA2 8QA, Wales. .,School of Medicine, Swansea University Medical School, Institute of Life Science 2, Swansea, SA2 8QA, Wales.
| | - J W Stephens
- Department of Surgery, Swansea Bay University Health Board, Singleton Hospital, Swansea, SA2 8QA, Wales.,School of Medicine, Swansea University Medical School, Institute of Life Science 2, Swansea, SA2 8QA, Wales
| | - D A Harris
- Department of Surgery, Swansea Bay University Health Board, Singleton Hospital, Swansea, SA2 8QA, Wales.,School of Medicine, Swansea University Medical School, Institute of Life Science 2, Swansea, SA2 8QA, Wales
| |
Collapse
|
108
|
Effect of Sake Lees on the Inhibition of Lipid Accumulation in Adipocytes. FERMENTATION 2021. [DOI: 10.3390/fermentation7030145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Recent lifestyle changes, such as the Westernization of diets and the rise in the prevalence of obesity, with an associated increase in the number of patients with lifestyle-related diseases, have become a social issue. Fermented food has attracted attention as a solution to problems caused by obesity. Sake lees, a byproduct of sake brewing, represent one such food that is expected to have health benefits. In this study, we investigated the effects of sake lees components on preadipocytes (3T3-L1). We cultured preadipocytes in a medium with indigestible sake lees components (ISLCs) to investigate lipid accumulation, analyzed the glycerol 3-phosphate dehydrogenase (GPDH) and LPL activities of those cells, and performed a real-time PCR analysis of the IL6 expression in the cells. The results show that lipid accumulation and GPDH activity were significantly decreased in adipocytes treated with 1.0 mg/mL ISLCs compared to untreated cells. Furthermore, the expression of IL6 in adipocytes treated with 1.0 mg/mL ISLCs was significantly decreased and the lipase activity was significantly increased in adipocytes treated with ISLCs after differentiation. IL6 is known to have multiple functions in adipose tissue. In conclusion, ISLCs were associated with reduced lipid accumulation in adipocytes, with effects on IL6 expression and LPL activity observed throughout the differentiation period.
Collapse
|
109
|
de Godoi RS, Almerão MP, da Silva FR. In silico evaluation of the antidiabetic activity of natural compounds from Hovenia dulcis Thunberg. J Herb Med 2021. [DOI: 10.1016/j.hermed.2020.100349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
110
|
Arumugam MK, Chava S, Rasineni K, Paal MC, Donohue TM, Osna NA, Kharbanda KK. Elevated S-adenosylhomocysteine induces adipocyte dysfunction to promote alcohol-associated liver steatosis. Sci Rep 2021; 11:14693. [PMID: 34282217 PMCID: PMC8289835 DOI: 10.1038/s41598-021-94180-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/28/2021] [Indexed: 02/08/2023] Open
Abstract
It has been previously shown that chronic ethanol administration-induced increase in adipose tissue lipolysis and reduction in the secretion of protective adipokines collectively contribute to alcohol-associated liver disease (ALD) pathogenesis. Further studies have revealed that increased adipose S-adenosylhomocysteine (SAH) levels generate methylation defects that promote lipolysis. Here, we hypothesized that increased intracellular SAH alone causes additional related pathological changes in adipose tissue as seen with alcohol administration. To test this, we used 3-deazaadenosine (DZA), which selectively elevates intracellular SAH levels by blocking its hydrolysis. Fully differentiated 3T3-L1 adipocytes were treated in vitro for 48 h with DZA and analysed for lipolysis, adipokine release and differentiation status. DZA treatment enhanced adipocyte lipolysis, as judged by lower levels of intracellular triglycerides, reduced lipid droplet sizes and higher levels of glycerol and free fatty acids released into the culture medium. These findings coincided with activation of both adipose triglyceride lipase and hormone sensitive lipase. DZA treatment also significantly reduced adipocyte differentiation factors, impaired adiponectin and leptin secretion but increased release of pro-inflammatory cytokines, IL-6, TNF and MCP-1. Together, our results demonstrate that elevation of intracellular SAH alone by DZA treatment of 3T3-L1 adipocytes induces lipolysis and dysregulates adipokine secretion. Selective elevation of intracellular SAH by DZA treatment mimics ethanol's effects and induces adipose dysfunction. We conclude that alcohol-induced elevations in adipose SAH levels contribute to the pathogenesis and progression of ALD.
Collapse
Affiliation(s)
- Madan Kumar Arumugam
- Research Service (151), Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE, 68105, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Srinivas Chava
- Research Service (151), Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE, 68105, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Karuna Rasineni
- Research Service (151), Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE, 68105, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Matthew C Paal
- Research Service (151), Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE, 68105, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Terrence M Donohue
- Research Service (151), Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE, 68105, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Natalia A Osna
- Research Service (151), Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE, 68105, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kusum K Kharbanda
- Research Service (151), Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Avenue, Omaha, NE, 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
111
|
Van de Velde F, Ouwens DM, Batens AH, Van Nieuwenhove Y, Lapauw B. Divergent dynamics in systemic and tissue-specific metabolic and inflammatory responses during weight loss in subjects with obesity. Cytokine 2021; 144:155587. [PMID: 34052657 DOI: 10.1016/j.cyto.2021.155587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022]
Abstract
AIM Dysfunction of adipose and muscle tissue associates with obesity-related co-morbidities such as insulin resistance (IR) and inflammation. This study investigates changes in systemic and tissue-specific markers of IR and inflammation after gastric bypass surgery (GBS) in subjects with obesity. METHODS Prospective study, twenty subjects with obesity (50 ± 10 years, 14 men). Prior to, and six months and one year after GBS, subcutaneous abdominal adipose tissue (SAT), skeletal muscle and fasting serum samples were collected. Serum levels of C-reactive protein (CRP), glucose and insulin were determined using standard laboratory assays and serum IL-6, IL-10 and TNF-α levels were determined using ELISA. Tissue mRNA expression of inflammation and insulin/glucose metabolism markers were analyzed using qPCR. RESULTS After GBS, HOMA-IR, CRP and IL-6 serum levels decreased. In SAT, expression of bone morphogenetic protein 4 (BMP4), IL-6, IL-10 and MCP1 decreased and GLUT4 increased (all p < 0.05). In muscle, expression of BMP4, GLUT4 and IL-6 decreased and of MCP1 and IRS-1 increased (all p < 0.05). CONCLUSION Systemic improvements in inflammation and IR after GBS are only partially mirrored by corresponding changes in adipokine and myokine expression patterns. As changes in expression of other markers of inflammation and insulin/glucose metabolism appear less consistent and even divergent between tissues, the inflammatory and IR status at systemic level cannot be extrapolated to the situation in metabolically active tissues.
Collapse
Affiliation(s)
| | - D Margriet Ouwens
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium; Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Duesseldorf, Germany; German Center for Diabetes Research (DZD), Muenchen-Neuherberg, Germany
| | | | - Yves Van Nieuwenhove
- Department of Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Bruno Lapauw
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
112
|
Gravina G, Ferrari F, Nebbiai G. The obesity paradox and diabetes. Eat Weight Disord 2021; 26:1057-1068. [PMID: 32954485 DOI: 10.1007/s40519-020-01015-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Obesity has been proven to be a risk factor for type 2 diabetes mellitus (T2DM) through numerous pathogenetic mechanisms. Unexpectedly, some studies suggest that subjects with overweight/obesity and T2DM have better clinical outcome than their normal weight peers. This finding is described as "obesity paradox" and calls into question the importance of weight loss in this specific population. OBJECTIVE This article is a narrative overview on the obesity and type 2 diabetes mellitus, particularly regarding the obesity paradox in T2DM patients. METHODS We used as sources MEDLINE/PubMed, CINAHL, EMBASE, and Cochrane Library, from inception to March 2020; we chose 30 relevant papers regarding the association of obesity with clinical outcome and mortality of patients affected by T2DM. RESULTS Many studies report that in patients with T2DM, overweight and obesity are associated with a better prognosis than underweight or normal weight, suggesting the presence of an obesity paradox. However, these studies have numerous limitations due to their mainly retrospective nature and to numerous confounding factors, such as associated pathologies, antidiabetic treatments, smoking habit, lack of data about distribution of body fat or weight history. CONCLUSION Literature data regarding the phenomenon of obesity paradox in T2DM patients are controversial due to the several limitations of the studies; therefore in the management of patients with overweight/obesity and T2DM is recommended referring to the established guidelines, which indicate diet and physical activity as the cornerstone of the treatment. LEVEL OF EVIDENCE Level V: narrative review.
Collapse
Affiliation(s)
- Giovanni Gravina
- Center for Eating Behaviour and Metabolism Disorders, Casa di Cura San Rossore, Pisa, Italy.
| | - Federica Ferrari
- Obesity and Lipodystrophy Center, Endocrinology Unit, University Hospital of Pisa, Pisa, Italy
| | - Grazia Nebbiai
- Center for Eating Behaviour and Metabolism Disorders, Casa di Cura San Rossore, Pisa, Italy
| |
Collapse
|
113
|
Bisphenols and the Development of Type 2 Diabetes: The Role of the Skeletal Muscle and Adipose Tissue. ENVIRONMENTS 2021. [DOI: 10.3390/environments8040035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bisphenol A (BPA) and bisphenol S (BPS) are environmental contaminants that have been associated with the development of insulin resistance and type 2 diabetes (T2D). Two organs that are often implicated in the development of insulin resistance are the skeletal muscle and the adipose tissue, however, seldom studies have investigated the effects of bisphenols on their metabolism. In this review we discuss metabolic perturbations that occur in both the skeletal muscle and adipose tissue affected with insulin resistance, and how exposure to BPA or BPS has been linked to these changes. Furthermore, we highlight the possible effects of BPA on the cross-talk between the skeletal muscle and adipose tissue.
Collapse
|
114
|
DI Renzo C, Vitale A, D'Amico F, Cillo U. NAFLD: a multi-faceted morbid spectrum with uncertain diagnosis and complicated management. Where do we stand? Review of the literature. Minerva Surg 2021; 76:450-466. [PMID: 33855376 DOI: 10.23736/s2724-5691.21.08729-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
NASH can be considered the "contemporary era pandemic", because of its global widespread in parallel with obesity, diabetes and metabolic dysfunction. It is a disease that often poses many difficulties, since making a early diagnosis is often impossible since specific diagnostic tests and criteria are missing: so, it needs a high degree of suspicion. Most of the times the evolution to its more severe and terminal step, NASH cirrhosis, is unavoidable and so are the social pressure on health sistem and economic consequences it brings back. In this work we aim to review the literature about both NAFLD and NASH, thus structuring a wide, comprehensive, 360 degree work with a focus on all major aspects of NAFLD, spanning from diagnosis, physiopathology and its repercussions on liver transplantation. Moreover we also focused on patients related issues both in pre- and post-transplant management (when these patients are listed for liver transplant). NAFLD and NASH are a contemporary plague, and an exaustive knowledge of the problem throughout all its aspects is necessary in order to lower economic weight that metabolic issues bring back and to have a open view to possible solutions to all management issues that NASH patients have and that are oten prohibitive to a definitive cure (for example cardiovascular risk in patients otherwise eligible to liver transplantation). We aim to offer a complete view on the actual knowledge about NAFLD and NASH, by an extensive review of the literature.
Collapse
Affiliation(s)
- Chiara DI Renzo
- Hepatobiliary Surgery and Liver Transplantation, Padova University Hospital, Padova, Italy -
| | - Alessandro Vitale
- Hepatobiliary Surgery and Liver Transplantation, Padova University Hospital, Padova, Italy
| | - Francesco D'Amico
- Hepatobiliary Surgery and Liver Transplantation, Padova University Hospital, Padova, Italy
| | - Umberto Cillo
- Hepatobiliary Surgery and Liver Transplantation, Padova University Hospital, Padova, Italy
| |
Collapse
|
115
|
Lee CB, Chae SU, Jo SJ, Jerng UM, Bae SK. The Relationship between the Gut Microbiome and Metformin as a Key for Treating Type 2 Diabetes Mellitus. Int J Mol Sci 2021; 22:ijms22073566. [PMID: 33808194 PMCID: PMC8037857 DOI: 10.3390/ijms22073566] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 02/08/2023] Open
Abstract
Metformin is the first-line pharmacotherapy for treating type 2 diabetes mellitus (T2DM); however, its mechanism of modulating glucose metabolism is elusive. Recent advances have identified the gut as a potential target of metformin. As patients with metabolic disorders exhibit dysbiosis, the gut microbiome has garnered interest as a potential target for metabolic disease. Henceforth, studies have focused on unraveling the relationship of metabolic disorders with the human gut microbiome. According to various metagenome studies, gut dysbiosis is evident in T2DM patients. Besides this, alterations in the gut microbiome were also observed in the metformin-treated T2DM patients compared to the non-treated T2DM patients. Thus, several studies on rodents have suggested potential mechanisms interacting with the gut microbiome, including regulation of glucose metabolism, an increase in short-chain fatty acids, strengthening intestinal permeability against lipopolysaccharides, modulating the immune response, and interaction with bile acids. Furthermore, human studies have demonstrated evidence substantiating the hypotheses based on rodent studies. This review discusses the current knowledge of how metformin modulates T2DM with respect to the gut microbiome and discusses the prospect of harnessing this mechanism in treating T2DM.
Collapse
Affiliation(s)
- Chae Bin Lee
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
| | - Soon Uk Chae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
| | - Seong Jun Jo
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
| | - Ui Min Jerng
- Department of Internal Medicine, College of Korean Medicine, Sangji University, Wonju 26339, Korea;
| | - Soo Kyung Bae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon 14662, Korea; (C.B.L.); (S.U.C.); (S.J.J.)
- Correspondence: ; Tel.: +82-2-2164-4054
| |
Collapse
|
116
|
Wang CR, Tsai HW. Anti- and non-tumor necrosis factor-α-targeted therapies effects on insulin resistance in rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis. World J Diabetes 2021; 12:238-260. [PMID: 33758645 PMCID: PMC7958474 DOI: 10.4239/wjd.v12.i3.238] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
In addition to β-cell failure with inadequate insulin secretion, the crucial mechanism leading to establishment of diabetes mellitus (DM) is the resistance of target cells to insulin, i.e. insulin resistance (IR), indicating a requirement of beyond-normal insulin concentrations to maintain euglycemic status and an ineffective strength of transduction signaling from the receptor, downstream to the substrates of insulin action. IR is a common feature of most metabolic disorders, particularly type II DM as well as some cases of type I DM. A variety of human inflammatory disorders with increased levels of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, have been reported to be associated with an increased risk of IR. Autoimmune-mediated arthritis conditions, including rheumatoid arthritis (RA), psoriatic arthritis (PsA) and ankylosing spondylitis (AS), with the involvement of proinflammatory cytokines as their central pathogenesis, have been demonstrated to be associated with IR, especially during the active disease state. There is an increasing trend towards using biologic agents and small molecule-targeted drugs to treat such disorders. In this review, we focus on the effects of anti-TNF-α- and non-TNF-α-targeted therapies on IR in patients with RA, PsA and AS. Anti-TNF-α therapy, IL-1 blockade, IL-6 antagonist, Janus kinase inhibitor and phospho-diesterase type 4 blocker can reduce IR and improve diabetic hyper-glycemia in autoimmune-mediated arthritis.
Collapse
Affiliation(s)
- Chrong-Reen Wang
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| | - Hung-Wen Tsai
- Department of Pathology, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| |
Collapse
|
117
|
He Y, Wu N. Research Progress on Gestational Diabetes Mellitus and Endothelial Dysfunction Markers. Diabetes Metab Syndr Obes 2021; 14:983-990. [PMID: 33688231 PMCID: PMC7937366 DOI: 10.2147/dmso.s295737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/15/2021] [Indexed: 01/24/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is the onset or first recognition of diabetes during pregnancy in women with normal glucose metabolism or potentially impaired glucose tolerance before pregnancy. Studies have shown that vascular endothelial cells (VECs) are an important target organ of insulin, which is injured by multiple factors in the case of GDM, thereby leading to worsened insulin resistance (IR) and the further development of GDM. When VECs are abnormal, there will be changes in the content of a variety of cell markers, which may be helpful for the clinical prediction and diagnosis of GDM. This study attempted to investigate the mechanism and markers of VECs injury in GDM patients.
Collapse
Affiliation(s)
- Yujing He
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, People’ s Republic of China
| | - Na Wu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, People’ s Republic of China
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
118
|
Werida RH, El-Gharbawy NM, Mostafa TM. Circulating IL-6, clusterin and irisin in obese subjects with different grades of obesity: association with insulin resistance and sexual dimorphism. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2021; 65:126-136. [PMID: 33905632 PMCID: PMC10065324 DOI: 10.20945/2359-3997000000336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Objective There are discrepancies about the relationship of IL-6, clusterin and irisin with obesity and obesity associated insulin resistance and also about their sexual dimorphism. This study aimed at evaluating the circulating levels of IL-6, clusterin and irisin in obese subjects of both sexes who had different grades of obesity and examining their sexual dimorphism and their association with insulin resistance. Methods This study included 176 non-diabetic subjects of both sexes who were classified according to their sex into two groups; the male and the female groups. The male group (88 men) was classified according to BMI into; group 1 (22 lean men), group 2 (22 class I obese men), group 3 (22 class II obese men) and group 4 (22 class III obese men). The female group (88 women) was classified according to BMI exactly as the male group. Metabolic parameters, IL-6, clusterin, and irisin levels were measured. Data were analyzed by ANOVA test, post hoc Tukey's test and independent t-test. Pearson correlation was used to assess the association between variables. Results In obese subjects of both sexes, circulating IL-6, clusterin and irisin levels were significantly elevated and positively correlated with HOMA-IR. Obese males showed significantly higher HOMA-IR, IL-6, clusterin and irisin levels than obese females. Conclusion Obesity in both sexes, especially in males was associated with high levels of IL-6, clusterin and irisin and worsened the metabolic pattern. Circulating IL-6, clusterin and irisin may represent possible therapeutic targets for insulin resistance in obese subjects.
Collapse
Affiliation(s)
- Rehab H Werida
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Damanhur University, Damanhur, Egypt
| | - Nashwa M El-Gharbawy
- Department of Internal Medicine, Diabetes & Endocrinology Unit, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Tarek M Mostafa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt,
| |
Collapse
|
119
|
Rajesh Y, Sarkar D. Association of Adipose Tissue and Adipokines with Development of Obesity-Induced Liver Cancer. Int J Mol Sci 2021; 22:ijms22042163. [PMID: 33671547 PMCID: PMC7926723 DOI: 10.3390/ijms22042163] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/20/2022] Open
Abstract
Obesity is rapidly dispersing all around the world and is closely associated with a high risk of metabolic diseases such as insulin resistance, dyslipidemia, and nonalcoholic fatty liver disease (NAFLD), leading to carcinogenesis, especially hepatocellular carcinoma (HCC). It results from an imbalance between food intake and energy expenditure, leading to an excessive accumulation of adipose tissue (AT). Adipocytes play a substantial role in the tumor microenvironment through the secretion of several adipokines, affecting cancer progression, metastasis, and chemoresistance via diverse signaling pathways. AT is considered an endocrine organ owing to its ability to secrete adipokines, such as leptin, adiponectin, resistin, and a plethora of inflammatory cytokines, which modulate insulin sensitivity and trigger chronic low-grade inflammation in different organs. Even though the precise mechanisms are still unfolding, it is now established that the dysregulated secretion of adipokines by AT contributes to the development of obesity-related metabolic disorders. This review focuses on several obesity-associated adipokines and their impact on obesity-related metabolic diseases, subsequent metabolic complications, and progression to HCC, as well as their role as potential therapeutic targets. The field is rapidly developing, and further research is still required to fully understand the underlying mechanisms for the metabolic actions of adipokines and their role in obesity-associated HCC.
Collapse
Affiliation(s)
- Yetirajam Rajesh
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Devanand Sarkar
- Massey Cancer Center, Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine (VIMM), Virginia Commonwealth University, Richmond, VA 23298, USA
- Correspondence: ; Tel.: +1-804-827-2339
| |
Collapse
|
120
|
Adipose Tissue T Regulatory Cells: Implications for Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:125-139. [PMID: 33523447 DOI: 10.1007/978-981-15-6407-9_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity dramatically increases the risk of numerous conditions, including type 2 diabetes mellitus and other components of the metabolic syndrome. Pro-inflammatory changes that occur in adipose tissue are critical to the pathogenesis of these obesity-induced complications. Adipose tissue is one of the body's largest endocrine organs, and the cells that comprise the adipose tissue immunoenvironment secrete multiple factors (including adipokines and cytokines) that impact systemic metabolism. In particular, immunosuppressive regulatory T cells (Tregs) decline in obesity, partly in response to its complex interaction with adipocytes, and this decline contributes to disruption of the typical homeostasis observed in lean adipose tissue. Although the regulation of Treg differentiation, function, and enrichment is incompletely understood, factors including various cell-surface co-stimulatory molecules, certain lipid species, and cytokines such as PPARγ, adiponectin, and leptin are important mediators. It is also clear that there may be depot-specific differences in Tregs, rendering adipose tissue Tregs distinct from lymphoid or circulating Tregs, with implications on maintenance and functionality. While most of these findings are derived from studies in murine models, comparatively little is known about the human adipose tissue Treg signature, which requires further investigation.
Collapse
|
121
|
Sinisgalli C, Vezza T, Diez-Echave P, Ostuni A, Faraone I, Hidalgo-Garcia L, Russo D, Armentano MF, Garrido-Mesa J, Rodriguez-Cabezas ME, Rodríguez-Nogales A, Milella L, Galvez J. The Beneficial Effects of Red Sun-Dried Capsicum annuum L. Cv Senise Extract with Antioxidant Properties in Experimental Obesity are Associated with Modulation of the Intestinal Microbiota. Mol Nutr Food Res 2021; 65:e2000812. [PMID: 33300660 DOI: 10.1002/mnfr.202000812] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/12/2020] [Indexed: 12/13/2022]
Abstract
SCOPE Capsicum annuum L. cv Senise is a sweet pepper containing health promoting compounds that can be modified by ripening and drying. This study focuses on finding the peppers with the best antioxidant properties, which are evaluated on an experimental model of obesity. METHODS AND RESULTS Phytochemical profile and antioxidant activity are evaluated on several peppers obtained from the same cultivar at different ripening stages. Red sweet peppers show the highest content in polyphenols, β-carotene, lycopene, and capsinoids, and demonstrate the best antioxidant activity in vitro. Mice fed a high fat diet are orally treated with an extract from these peppers (Capsicum annuum extract [CAE]) (1, 10, and 25 mg/kg/day). It promotes weight loss and improves plasma markers related to glucose and lipid metabolisms. CAE also ameliorates obesity-associated systemic inflammation reducing the expression of pro-inflammatory cytokines in adipose and hepatic tissues and improving the expression of different markers involved in the gut epithelial barrier function. These effects are associated with a modulation of the intestinal microbiome, which appears altered. CONCLUSIONS The extract can be considered a new potential approach for the treatment of obesity, complementary to dietary restrictions.
Collapse
Affiliation(s)
- Chiara Sinisgalli
- Department of Science, University of Basilicata, via dell'Ateneo Lucano 10, Potenza, 85100, Italy
- Spinoff BioActiPlant S.R.L, University of Basilicata, Potenza, 85100, Italy
| | - Teresa Vezza
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, 18071, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18014, Spain
| | - Patricia Diez-Echave
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, 18071, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18014, Spain
| | - Angela Ostuni
- Department of Science, University of Basilicata, via dell'Ateneo Lucano 10, Potenza, 85100, Italy
- Spinoff BioActiPlant S.R.L, University of Basilicata, Potenza, 85100, Italy
| | - Immacolata Faraone
- Department of Science, University of Basilicata, via dell'Ateneo Lucano 10, Potenza, 85100, Italy
- Spinoff BioActiPlant S.R.L, University of Basilicata, Potenza, 85100, Italy
| | - Laura Hidalgo-Garcia
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, 18071, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18014, Spain
| | - Daniela Russo
- Department of Science, University of Basilicata, via dell'Ateneo Lucano 10, Potenza, 85100, Italy
- Spinoff BioActiPlant S.R.L, University of Basilicata, Potenza, 85100, Italy
| | - Maria Francesca Armentano
- Department of Science, University of Basilicata, via dell'Ateneo Lucano 10, Potenza, 85100, Italy
- Spinoff BioActiPlant S.R.L, University of Basilicata, Potenza, 85100, Italy
| | - José Garrido-Mesa
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, 18071, Spain
| | - Maria Elena Rodriguez-Cabezas
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, 18071, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18014, Spain
| | - Alba Rodríguez-Nogales
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, 18071, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18014, Spain
- Digestive Unit, Hospital Universitario Virgen de las Nieves, Granada, 18012, Spain
| | - Luigi Milella
- Department of Science, University of Basilicata, via dell'Ateneo Lucano 10, Potenza, 85100, Italy
- Spinoff BioActiPlant S.R.L, University of Basilicata, Potenza, 85100, Italy
| | - Julio Galvez
- CIBER-EHD, Department of Pharmacology, Center for Biomedical Research (CIBM), University of Granada, Granada, 18071, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, 18014, Spain
| |
Collapse
|
122
|
Wu HQ, Wei X, Yao JY, Qi JY, Xie HM, Sang AM, Jiang K. Association between retinopathy, nephropathy, and periodontitis in type 2 diabetic patients: a Meta-analysis. Int J Ophthalmol 2021; 14:141-147. [PMID: 33469496 DOI: 10.18240/ijo.2021.01.20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/22/2020] [Indexed: 12/28/2022] Open
Abstract
AIM To verify the association between retinopathy, nephropathy, and periodontitis in type 2 diabetic (T2D) patients. METHODS Several electronic databases were available for our comprehensive search including China National Knowledge Infrastructure (CNKI), Chinese VIP Information (VIP), Wanfang, Web of Science, ScienceDirect and PubMed and were queried for relevant citations (updated to Mar. 2019). RevMan was utilized to perform Meta analysis and publication bias detection. After evaluation of the methodological quality of included studies, a fixed or random effect model was utilized to analyze data from included studies. RESULTS A total of eight articles were finally included in this Meta analysis. In all 3987 subjects, there were 1207 T2D patients accompanying with microvascular complications and 1734 patients with periodontitis as well. The Meta forest plot presented little heterogeneity of the eight studies (P<0.00001, I 2=89%). The total effect demonstrated periodontitis was associated with overall microvascular complications (OR: 1.96, 95%CI: 1.67-2.30, Z=8.25, P<0.00001). Subgroup investigations among the studies in Asian (OR: 2.33, 95%CI: 1.91-2.85) and North American (OR: 1.42, 95%CI: 1.08-1.86) populations confirmed the existed association between retinopathy, nephropathy, and periodontitis. While the strength of such associations between periodontitis and diabetic microvascular complications were more obvious in the Asians than North Americans. All the results indicated that periodontitis was associated with diabetic retinopathy (OR: 3.77, 95%CI: 2.71-5.24), diabetic nephropathy (OR: 1.55, 95%CI: 1.24-1.94) in T2D patients. CONCLUSION The periodontitis is associated with diabetic retinopathy, diabetic nephropathy among T2D patients and further large sample size clinical trials are in need to confirm the findings.
Collapse
Affiliation(s)
- Hui-Qun Wu
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiao Wei
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jin-Yi Yao
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jian-Yan Qi
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Hui-Min Xie
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Ai-Min Sang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Kui Jiang
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
123
|
Pang MD, Goossens GH, Blaak EE. The Impact of Artificial Sweeteners on Body Weight Control and Glucose Homeostasis. Front Nutr 2021; 7:598340. [PMID: 33490098 PMCID: PMC7817779 DOI: 10.3389/fnut.2020.598340] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
A poor diet is one of the leading causes for non-communicable diseases. Due to the increasing prevalence of overweight and obesity, there is a strong focus on dietary overconsumption and energy restriction. Many strategies focus on improving energy balance to achieve successful weight loss. One of the strategies to lower energy intake is refraining from sugars and replacing them with artificial sweeteners, which maintain the palatability without ingesting calories. Nevertheless, the safety and health benefits of artificial sweeteners consumption remain a topic of debate within the scientific community and society at large. Notably, artificial sweeteners are metabolized differently from each other due to their different properties. Therefore, the difference in metabolic fate of artificial sweeteners may underlie conflicting findings that have been reported related to their effects on body weight control, glucose homeostasis, and underlying biological mechanisms. Thus, extrapolation of the metabolic effects of a single artificial sweetener to all artificial sweeteners is not appropriate. Although many rodent studies have assessed the metabolic effects of artificial sweeteners, long-term studies in humans are scarce. The majority of clinical studies performed thus far report no significant effects or beneficial effects of artificial sweeteners on body weight and glycemic control, but it should be emphasized that the study duration of most studies was limited. Clearly, further well-controlled, long-term human studies investigating the effects of different artificial sweeteners and their impact on gut microbiota, body weight regulation and glucose homeostasis, as well as the underlying mechanisms, are warranted.
Collapse
Affiliation(s)
- Michelle D. Pang
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | | | | |
Collapse
|
124
|
Al-Awaida WJ, Hameed WS, Al Hassany HJ, Al-Dabet MM, Al-Bawareed O, Hadi NR. Evaluation of the Genetic Association and Expressions of Notch-2 /Jagged-1 in Patients with Type 2 Diabetes Mellitus. Med Arch 2021; 75:101-108. [PMID: 34219868 PMCID: PMC8228649 DOI: 10.5455/medarh.2021.75.101-108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Diabetes mellitus (DM) is the world's most common cause of chronic kidney diseases (CKD), with approximately 1 in 4 adults with DM having CKD and 1 out of 10 to 20% of DM patients die from CKD. Objective The current study aims to investigate the correlation between Notch-2 and Jag-1expressions and specific inflammation biomarkers IL-1β and IL-6 with different stages of diabetic nephropathy. Methods From August 2018 to January 2019, three hundred subjects were recruited for this study. One hundred and fifty subjects were healthy and age-matched to the diabetic group and selected as a control group. Another 150 patients with an established diagnosis of type 2 diabetes (T2DM) according to the criteria of the American Diabetes Association (ADA) were also recruited. Blood specimens were eventually used to identify the expressions Notch-2 and Jagged-1 and the levels of inflammatory biomarkers IL-1β and IL-6. Result The current study shows a significant increase in gene expression and inflammatory biomarkers in patients with moderate and severe diabetic nephropathy compared to the control group. However, there was no significant difference between healthy control and mild diabetic nephropathy patients. This study shows a close association between the increase in the levels of inflammatory biomarkers IL-1β and IL-6 as well as the gene expressions levels of both Notch-2 and Jag-1 with human diabetic nephropathy. Conclusion According to our findings, we emphasize the use of Notch-2 and Jag-1 expressions and IL-1β and IL-6 levels as potential biomarkers for different stages of diabetic nephropathy.
Collapse
Affiliation(s)
- Wajdy J Al-Awaida
- Department of Biology and Biotechnology, American University of Madaba, Madaba, Jordan
| | - Wasan S Hameed
- Department of Microbiology, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | - Haider J Al Hassany
- Department of Microbiology, Faculty of Medicine, University of Kufa, Kufa, Iraq
| | | | - Omar Al-Bawareed
- Department of normal physiology, RUDN University, Moscow, Russia
| | - Najah R Hadi
- Department of Pharmacology and Therapeutics,Faculty of Medicine, University of Kufa, Kufa, Iraq
| |
Collapse
|
125
|
Eskandarani RM, Sawan S. Diabetic Ketoacidosis on Hospitalization with COVID-19 in a Previously Nondiabetic Patient: A Review of Pathophysiology. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2020; 13:1179551420984125. [PMID: 33488135 PMCID: PMC7768872 DOI: 10.1177/1179551420984125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/04/2020] [Indexed: 01/08/2023]
Abstract
Hyperglycaemia during inpatient admission is indicative of higher morbidity and mortality risks in critically ill patients. The severe acute respiratory distress coronavirus 2 (SARS-CoV-2) has been reported to induce ketoacidosis and diabetic ketoacidosis (DKA) even in nondiabetic patients. The pathophysiology of the SARS-CoV-2 infection that can contribute to hyperglycaemia, and the exacerbated inflammatory cytokine storm can overlap with the metabolic chronic inflammatory state attributable to the metabolic syndrome, which underlies diabetes mellitus. In this report, we explore the possible pathophysiology and metabolic mechanisms that lead to metabolic acidosis in nondiabetic patients.
Collapse
|
126
|
Meng F, Sun Q, Zhou D, Li Q, Han J, Liu D, Yang J. Inhibition of Aurora-A improves insulin resistance by ameliorating islet inflammation and controlling interleukin-6 in a diabetic mouse model. Adipocyte 2020; 9:609-619. [PMID: 33043822 PMCID: PMC7553512 DOI: 10.1080/21623945.2020.1829851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Aurora-A kinase, a serine/threonine mitotic kinase, is reportedly upregulated in skin tissues of individuals with type 2 diabetes mellitus , although its function in diabetes is unclear. C57BL/6 J mice were utilized to establish a type 2 diabetic model and explore the functions of Aurora-A in diabetes. Aurora-A was highly expressed in the pancreas of the diabetic mice as confirmed by western blot. Inhibition of Aurora-A did not affect fasting blood glucose and body weight, but did improve insulin resistance, as indicated by improved oral glucose tolerance, insulin tolerance, and the Homoeostasis Model Assessment-Insulin Resistance index. Blockade of Aurora-A dramatically decreased the number of infiltrating macrophages in the pancreas in parallel with decreases in the levels of serum insulin and interleukin-6 (IL-6) mRNA. The levels of phosphorylated forms of protein kinase B, which are the key mediators of in insulin resistance, were not induced in liver, adipocyte tissues, and skeletal muscle by alisertib treatment. Our findings indicate that suppression of Aurora-A could at least partially enhance insulin sensitivity by decreasing the number of infiltrating macrophages and IL-6 level in a type 2 diabetic mouse model.
Collapse
Affiliation(s)
- Fandong Meng
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University
| | - Qiangwei Sun
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University
- Department of Endocrinology, Wuhan Third Hospital, Wuhan, China
| | - Dongmei Zhou
- Department of Endocrinology, Affiliated Hospital of Xuzhou Medical University
| | - Qiang Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University
| | - Jing Han
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Jing Yang
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University
| |
Collapse
|
127
|
Rodriguez-Ayala E, Gallegos-Cabrales EC, Gonzalez-Lopez L, Laviada-Molina HA, Salinas-Osornio RA, Nava-Gonzalez EJ, Leal-Berumen I, Escudero-Lourdes C, Escalante-Araiza F, Buenfil-Rello FA, Peschard VG, Laviada-Nagel A, Silva E, Veloz-Garza RA, Martinez-Hernandez A, Barajas-Olmos FM, Molina-Segui F, Gonzalez-Ramirez L, Espadas-Olivera R, Lopez-Muñoz R, Arjona-Villicaña RD, Hernandez-Escalante VM, Rodriguez-Arellano ME, Gaytan-Saucedo JF, Vaquera Z, Acebo-Martinez M, Cornejo-Barrera J, Jancy Andrea HQ, Castillo-Pineda JC, Murillo-Ramirez A, Diaz-Tena SP, Figueroa-Nuñez B, Valencia-Rendon ME, Garzon-Zamora R, Viveros-Paredes JM, Ángeles-Chimal J, Santa-Olalla Tapia J, Remes-Troche JM, Valdovinos-Chavez SB, Huerta-Avila EE, Lopez-Alvarenga JC, Comuzzie AG, Haack K, Han X, Orozco L, Weintraub S, Kent JW, Cole SA, Bastarrachea RA. Towards precision medicine: defining and characterizing adipose tissue dysfunction to identify early immunometabolic risk in symptom-free adults from the GEMM family study. Adipocyte 2020; 9:153-169. [PMID: 32272872 PMCID: PMC7153654 DOI: 10.1080/21623945.2020.1743116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
Interactions between macrophages and adipocytes are early molecular factors influencing adipose tissue (AT) dysfunction, resulting in high leptin, low adiponectin circulating levels and low-grade metaflammation, leading to insulin resistance (IR) with increased cardiovascular risk. We report the characterization of AT dysfunction through measurements of the adiponectin/leptin ratio (ALR), the adipo-insulin resistance index (Adipo-IRi), fasting/postprandial (F/P) immunometabolic phenotyping and direct F/P differential gene expression in AT biopsies obtained from symptom-free adults from the GEMM family study. AT dysfunction was evaluated through associations of the ALR with F/P insulin-glucose axis, lipid-lipoprotein metabolism, and inflammatory markers. A relevant pattern of negative associations between decreased ALR and markers of systemic low-grade metaflammation, HOMA, and postprandial cardiovascular risk hyperinsulinemic, triglyceride and GLP-1 curves was found. We also analysed their plasma non-coding microRNAs and shotgun lipidomics profiles finding trends that may reflect a pattern of adipose tissue dysfunction in the fed and fasted state. Direct gene differential expression data showed initial patterns of AT molecular signatures of key immunometabolic genes involved in AT expansion, angiogenic remodelling and immune cell migration. These data reinforce the central, early role of AT dysfunction at the molecular and systemic level in the pathogenesis of IR and immunometabolic disorders.
Collapse
Affiliation(s)
- Ernesto Rodriguez-Ayala
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | | | - Laura Gonzalez-Lopez
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - Rocio A. Salinas-Osornio
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - Irene Leal-Berumen
- Facultad de Medicina y Ciencias Biomédicas, Universidad Autónoma de Chihuahua, México
| | | | - Fabiola Escalante-Araiza
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Fatima A. Buenfil-Rello
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Vanessa-Giselle Peschard
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Antonio Laviada-Nagel
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Eliud Silva
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México City, México
| | - Rosa A. Veloz-Garza
- Facultad de Enfermería, Universidad Autónoma de Nuevo León (UANL), Monterrey, México
| | - Angelica Martinez-Hernandez
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Francisco M. Barajas-Olmos
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | | | | | | | - Ricardo Lopez-Muñoz
- Escuela de Ciencias de la Salud, Universidad Marista de Mérida, Yucatán, Mexico
| | | | - Victor M. Hernandez-Escalante
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | - Janeth F. Gaytan-Saucedo
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Zoila Vaquera
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | - Judith Cornejo-Barrera
- Departamento de Enseñanza, Postgrado e Investigación, Hospital Infantil de Tamaulipas, Ciudad, México
| | - Huertas-Quintero Jancy Andrea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | | | | | - Sara P. Diaz-Tena
- Departamento de Nutrición Humana, Universidad Latina de América, Morelia, México
| | | | | | - Rafael Garzon-Zamora
- Dirección de Postgrado e Investigación, Universidad del Valle de Atemajac (UNIVA), Zapopan, México
| | | | - José Ángeles-Chimal
- Facultad de Medicina, Universidad Autónoma Estado de Morelos, Cuernavaca, México
| | | | - José M. Remes-Troche
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, México
| | | | - Eira E. Huerta-Avila
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Juan Carlos Lopez-Alvarenga
- School of Medicine & South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Xianlin Han
- Department of Medicine, Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Lorena Orozco
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica, México City, México
| | - Susan Weintraub
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jack W. Kent
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Shelley A. Cole
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| | - Raul A. Bastarrachea
- Population Health Program, Texas Biomedical Research Institute and Southwest National Primate Research Center (SNPRC), San Antonio, TX, USA
| |
Collapse
|
128
|
Jiang YY, Shui JC, Zhang BX, Chin JW, Yue RS. The Potential Roles of Artemisinin and Its Derivatives in the Treatment of Type 2 Diabetes Mellitus. Front Pharmacol 2020; 11:585487. [PMID: 33381036 PMCID: PMC7768903 DOI: 10.3389/fphar.2020.585487] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic disease that has become a global public health problem. Studies on T2DM prevention and treatment mostly focus on discovering therapeutic drugs. Artemisinin and its derivatives were originally used as antimalarial treatments. In recent years, the roles of artemisinins in T2DM have attracted much attention. Artemisinin treatments not only attenuate insulin resistance and restore islet ß-cell function in T2DM but also have potential therapeutic effects on diabetic complications, including diabetic kidney disease, cognitive impairment, diabetic retinopathy, and diabetic cardiovascular disease. Many in vitro and in vivo experiments have confirmed the therapeutic utility of artemisinin and its derivatives on T2DM, but no article has systematically demonstrated the specific role artemisinin plays in the treatment of T2DM. This review summarizes the potential therapeutic effects and mechanism of artemisinin and its derivatives in T2DM and associated complications, providing a reference for subsequent related research.
Collapse
Affiliation(s)
- Ya-Yi Jiang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia-Cheng Shui
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo-Xun Zhang
- Department of Endocrinology, Guang'anmen Hospital of China, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia-Wei Chin
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ren-Song Yue
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
129
|
Sanap A, Bhonde R, Joshi K. Mesenchymal stem cell conditioned medium ameliorates diabetic serum-induced insulin resistance in 3T3-L1 cells. Chronic Dis Transl Med 2020; 7:47-56. [PMID: 34013180 PMCID: PMC8110877 DOI: 10.1016/j.cdtm.2020.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 11/14/2022] Open
Abstract
Background Pharmacological factors used to induce insulin resistance (IR) in in vitro models may not mimic the full in vivo features of type 2 diabetes mellitus (T2DM). This study aimed to examine the ability of diabetic serum (DS) to induce IR and investigate whether adipose-derived mesenchymal stem cell conditioned medium (ADMSC-CM) reverses DS-induced IR. Methods DS was obtained from newly diagnosed T2DM patients. IR was induced in differentiated 3T3-L1 cells by employing dexamethasone, tumor necrosis factor alpha (TNF-α), palmitate and DS. Glucose uptake (2-[N-[7-nitrobenz-2-oxa-1,3-diazol-4-yl] amino]-2-deoxyglucose(2-NBDG) uptake assay), intracellular levels of reactive oxygen species (ROS), and superoxide radicals (O2−) (fluorescence microscopy and fluorometry) were analyzed in control and experimental samples. mRNA expression of key genes involved in glucose transport and inflammation were analyzed by using reverse transcription polymerase chain reaction (RT-PCR). Pro-inflammatory cytokines and phospho-insulin receptor substrate (IRS) (Ser-307) protein expression were analyzed by fluorescence activated cell sorter analysis. Statistical significance was determined by using one-way ANOVA followed by Tukey's multiple comparison tests. Results ADMSC-CM significantly increased the DS-mediated decrease in 2-NBDG uptake (11.01 ± 0.50 vs. 7.20 ± 0.30, P < 0.01) and reduced DS-driven ROS (fluorescence count, 6.35 ± 0.46 vs. 9.80 ± 0.10, P < 0.01) and O2− (fluorescence count, 3.00 ± 0.10 vs. 4.60 ± 0.09, P < 0.01) production. Further, the ADMSC-CM restored DS-induced down regulation GLUT4 (1.52-fold, P < 0.05) as well as the up-regulation of PPARγ (0.35-fold, P < 0.01), and IKKβ (0.37-fold, P < 0.01) mRNA, and phospho-IRS (Ser-307) protein expression compared to the baseline (median fluorescence intensity, 88,192 ± 2720 vs. 65,450 ± 3111, P < 0.01). DS induced IR, similar to the traditionally used pharmacological factors, namely dexamethasone, TNF-α, and palmitate, which can be attributed to the significantly higher pro-inflammatory cytokines levels (TNF-α (2.28 ± 0.03 pg/mL vs. 2.38 ± 0.03 pg/mL, P < 0.01), interleukin 6 (IL)-6 (1.94 ± 0.02 pg/mL vs. 2.17 ± 0.04 pg/mL, P < 0.01), IL-17 (2.16 ± 0.02 pg/mL vs. 2.22 ± 0.002 pg/mL, P < 0.05), and interferon gamma (IFN-γ) (2.07 ± 0.02 pg/mL vs. 2.15 ± 0.04 pg/mL, P < 0.05)) in DS. Conclusions DS can be explored as a novel inducer of IR in in vitro studies with further standardization, substituting the conventionally used pharmacological factors. Our findings also affirm the validity of ADMSC-CM as a prospective insulin sensitizer for T2DM therapy.
Collapse
Affiliation(s)
- Avinash Sanap
- Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune 411007, India.,Regenerative Medicine Laboratory, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | - Ramesh Bhonde
- Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | - Kalpana Joshi
- Department of Biotechnology, Sinhgad College of Engineering affiliated to Savitribai Phule Pune University, Pune 411041, India
| |
Collapse
|
130
|
Kobayashi H, Seki R, Ujita M, Hirayama K, Yamada S, Ohashi R, Otsuki Y, Watanabe T, Yoshino T. An Autopsy Case of an Elderly Patient with Classic Hodgkin Lymphoma Presenting with a Plethora of Clinical Symptoms and Signs. AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e926177. [PMID: 33087692 PMCID: PMC7588351 DOI: 10.12659/ajcr.926177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/14/2020] [Accepted: 07/25/2020] [Indexed: 11/14/2022]
Abstract
BACKGROUND Hodgkin lymphoma (HL) is a potentially curable disease with favorable outcomes. However, elderly patients with HL usually have more adverse prognostic factors and hence a much worse prognosis than younger patients. CASE REPORT The patient was a woman in her 80s. She reported high fever, anorexia, and a weight loss of 8 kg within 5 months. She had been on treatment for diabetes mellitus and hypertension. She had undergone percutaneous coronary intervention and pacemaker implantation to treat acute coronary syndrome and sinus arrhythmia, respectively. Blood tests showed elevation of alkaline phosphatase, C-reactive protein, leukocyte count, CA 19-9, and carcinoembryonic antigen. Computed tomography did not show tumors in the liver, and cholangitis and sepsis were suspected. Aspartate transaminase, alanine aminotransferase, and total bilirubin gradually increased through the course of the patient's hospital stay. Despite treatment, her condition deteriorated and she died 22 days after hospital admission. At autopsy, we found stage IV HL with lymph node swelling on both sides of the diaphragm, as well as diffusely disseminated nodules in the liver and spleen. CONCLUSIONS Our patient had several poor prognostic factors including B symptoms, comorbidity, advanced stage, Epstein-Barr virus infection, and expression of programmed death-ligand 1 and interleukin-6, all of which were closely connected with her advanced age. Her age and comorbidities may have been the most adverse prognostic factors for her illness. An effective HL screening method for elderly individuals should be developed to ameliorate poor prognosis and adverse outcomes.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Pathology, Tachikawa General Hospital, Nagaoka, Niigata, Japan
| | - Ryouya Seki
- Department of of Gastroenterology, Tachikawa General Hopspital, Nagaoka, Niigata, Japan
| | - Masuo Ujita
- Department of Radiology, Tachikawa General Hospital, Nagaoka, Niigata, Japan
| | - Kana Hirayama
- Department of Dermatology, Niigata University Medical and Dental Hospital, Niigata City, Niigata, Japan
| | - Satoshi Yamada
- Health Examination Center, Nagaoka Central General Hospital, Nagaoka, Niigata, Japan
| | - Riuko Ohashi
- Histopathology Core Facility, Niigata University School of Medicine, Niigata City, Niigata, Japan
| | - Yoshiro Otsuki
- Department of Pathology, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan
| | - Takuya Watanabe
- Department of General Medicine, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan
| | - Tadashi Yoshino
- Department of Pathology, Okayama University, Okayama City, Okayama, Japan
| |
Collapse
|
131
|
Escalona-Garrido C, Vázquez P, Mera P, Zagmutt S, García-Casarrubios E, Montero-Pedrazuela A, Rey-Stolle F, Guadaño-Ferraz A, Rupérez FJ, Serra D, Herrero L, Obregon MJ, Valverde ÁM. Moderate SIRT1 overexpression protects against brown adipose tissue inflammation. Mol Metab 2020; 42:101097. [PMID: 33049408 PMCID: PMC7600394 DOI: 10.1016/j.molmet.2020.101097] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Metainflammation is a chronic low-grade inflammatory state induced by obesity and associated comorbidities, including peripheral insulin resistance. Brown adipose tissue (BAT), a therapeutic target against obesity, is an insulin target tissue sensitive to inflammation. Therefore, it is necessary to find strategies to protect BAT against the effects of inflammation in energy balance. In this study, we explored the impact of moderate sirtuin 1 (SIRT1) overexpression on insulin sensitivity and β-adrenergic responses in BAT and brown adipocytes (BA) under pro-inflammatory conditions. METHODS The effect of inflammation on BAT functionality was studied in obese db/db mice and lean wild-type (WT) mice or mice with moderate overexpression of SIRT1 (SIRT1Tg+) injected with a low dose of bacterial lipopolysaccharide (LPS) to mimic endotoxemia. We also conducted studies on differentiated BA (BA-WT and BA-SIRT1Tg+) exposed to a macrophage-derived pro-inflammatory conditioned medium (CM) to evaluate the protection of SIRT1 overexpression in insulin signaling and glucose uptake, mitochondrial respiration, fatty acid oxidation (FAO), and norepinephrine (NE)-mediated-modulation of uncoupling protein-1 (UCP-1) expression. RESULTS BAT from the db/db mice was susceptible to metabolic inflammation manifested by the activation of pro-inflammatory signaling cascades, increased pro-inflammatory gene expression, tissue-specific insulin resistance, and reduced UCP-1 expression. Impairment of insulin and noradrenergic responses were also found in the lean WT mice upon LPS injection. In contrast, BAT from the mice with moderate overexpression of SIRT1 (SIRT1Tg+) was protected against LPS-induced activation of pro-inflammatory signaling, insulin resistance, and defective thermogenic-related responses upon cold exposure. Importantly, the decline in triiodothyronine (T3) levels in the circulation and intra-BAT after exposure of the WT mice to LPS and cold was markedly attenuated in the SIRT1Tg+ mice. In vitro BA experiments in the two genotypes revealed that upon differentiation with a T3-enriched medium and subsequent exposure to a macrophage-derived pro-inflammatory CM, only BA-SIRT1Tg+ fully recovered insulin and noradrenergic responses. CONCLUSIONS This study has ascertained the benefit of the moderate overexpression of SIRT1 to confer protection against defective insulin and β-adrenergic responses caused by BAT inflammation. Our results have potential therapeutic value in combinatorial therapies for BAT-specific thyromimetics and SIRT1 activators to combat metainflammation in this tissue.
Collapse
Affiliation(s)
- Carmen Escalona-Garrido
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), 28029 Madrid, Spain
| | - Patricia Vázquez
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), 28029 Madrid, Spain.
| | - Paula Mera
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Sebastián Zagmutt
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain
| | - Ester García-Casarrubios
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain
| | - Ana Montero-Pedrazuela
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Fernanda Rey-Stolle
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universitiy, Urbanización Montepríncipe, Boadilla del Monte, 28660, Madrid, Spain
| | - Ana Guadaño-Ferraz
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Francisco J Rupérez
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universitiy, Urbanización Montepríncipe, Boadilla del Monte, 28660, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, E-08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Maria Jesus Obregon
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols (Centro Mixto CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), 28029 Madrid, Spain.
| |
Collapse
|
132
|
Mitchell BI, Laws EI, Chow DC, Sah Bandar IN, Gangcuangco LMA, Shikuma CM, Ndhlovu LC. Increased Monocyte Inflammatory Responses to Oxidized LDL Are Associated with Insulin Resistance in HIV-Infected Individuals on Suppressive Antiretroviral Therapy. Viruses 2020; 12:v12101129. [PMID: 33028018 PMCID: PMC7601436 DOI: 10.3390/v12101129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/26/2020] [Accepted: 10/02/2020] [Indexed: 11/25/2022] Open
Abstract
Despite long term antiretroviral therapy (ART), insulin resistance (IR) is common among people living with HIV/AIDS (PLWHA) exposing this population to a greater risk of cardiometabolic complications when compared to their uninfected counterparts. We previously identified an expansion in monocyte subpopulations in blood that were linked to the degree of IR in persons with HIV on stable ART. In this study, we directly assessed monocyte inflammatory functional properties from PLWHA on ART (n = 33) and HIV-uninfected controls (n = 14) of similar age, gender, and cardiovascular disease risk and determined the relationship with IR (homeostatic model assessment-insulin resistance (HOMA-IR)), calculated from fasting blood glucose and insulin measurements. Peripheral blood mononuclear cells were stimulated with oxidized low-density lipoproteins (oxLDL) and polyfunctional monocyte cytokine responses (IL-1β, IL-6, IL-8, or TNF-α) were determined by flow cytometry. Higher monocyte IL-1β and IL-8 responses to oxLDL were associated with higher IR in PLWHA but not in the control group. We observed that higher basal monocyte cytokine responses were associated with both duration since HIV diagnosis and ART initiation. In the management of IR in chronic HIV, strategies lowering monocyte IL-1β and IL-8 responses should be considered in addition to ART in order to limit adverse cardio-metabolic outcomes.
Collapse
|
133
|
Chow BC, Li S, Zhu X, Jiao J, Quach B, Baker JS, Zhang H. Effects of descending or ascending stair exercise on body composition, insulin sensitivity, and inflammatory markers in young Chinese women with obesity: A randomized controlled trial. J Sports Sci 2020; 39:496-502. [PMID: 33012244 DOI: 10.1080/02640414.2020.1829362] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We examined the effects of descending (DSE) or ascending (ASE) stair exercise on body composition, insulin sensitivity, and inflammatory markers in young Chinese women with obesity. Thirty-six participants were randomly assigned into three groups DSE, ASE and a control group. The DSE and ASE groups performed three sessions of stair walking per week for 12 weeks with a gradual increase in repetitions. Following the exercise interventions, body composition related variables obtained by Dual-energy X-ray absorptiometry scans significantly decreased. Abdominal fat decreased in the DSE group only. Moreover, Insulin sensitivity improved significantly 3.5-fold in the DSE group compared with ASE group (insulin: -33.2% vs. -9.8%, homoeostasis model assessment for insulin resistance: -35.6% vs. -10.8%). Pro-inflammatory factors showed significant decreases in tumour necrosis factor-α (TNF-α) (-39.9% vs. -23.2%) for both intervention groups. The reduction in TNF-α concentrations in the DSE group was significantly different compared to the other two groups. Interleukin-6 significantly decreased in both exercise protocols. Our results show that 12-weeks induced stair walking improved body composition parameters in Chinese females with obesity. The results also demonstrate the superiority of the DSE protocol for improving insulin sensitivity. These findings may be attributable to the decreases observed in TNF- α levels.
Collapse
Affiliation(s)
- Bik Chu Chow
- Dr. Stephen Hui Research Centre for Physical Recreation and Wellness, Hong Kong Baptist University, Hong Kong, China.,Department of Sport and Physical Education, Hong Kong Baptist University, Hong Kong, China
| | - Shuoqi Li
- Physical Education College, Hebei Normal University, Shijiazhuang, China.,School of Health Sciences, University Sains Malaysia, Kota Bahru, Malaysia
| | - Xiangui Zhu
- Dr. Stephen Hui Research Centre for Physical Recreation and Wellness, Hong Kong Baptist University, Hong Kong, China.,Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Jiao Jiao
- Dr. Stephen Hui Research Centre for Physical Recreation and Wellness, Hong Kong Baptist University, Hong Kong, China
| | - Binh Quach
- Department of Sport and Physical Education, Hong Kong Baptist University, Hong Kong, China
| | - Julien S Baker
- Department of Sport and Physical Education, Hong Kong Baptist University, Hong Kong, China
| | - Haifeng Zhang
- Physical Education College, Hebei Normal University, Shijiazhuang, China.,Hebei Provincial Key Lab of Measurement and Evaluation in Human Movement and Bio-Information, Hebei Normal University, Shijiazhuang, China
| |
Collapse
|
134
|
High fat diet-triggered non-alcoholic fatty liver disease: A review of proposed mechanisms. Chem Biol Interact 2020; 330:109199. [DOI: 10.1016/j.cbi.2020.109199] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
|
135
|
Acierno C, Caturano A, Pafundi PC, Nevola R, Adinolfi LE, Sasso FC. Nonalcoholic fatty liver disease and type 2 diabetes: pathophysiological mechanisms shared between the two faces of the same coin. EXPLORATION OF MEDICINE 2020. [DOI: 10.37349/emed.2020.00019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathophysiological mechanisms underlying the close relationship between nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) are multiple, complex and only partially known. The purpose of this paper was to review the current knowledge of these mechanisms in a unified manner. Subjects with NAFLD and T2DM have established insulin resistance (IR), which exacerbates the two comorbidities. IR worsens NAFLD by increasing the accumulation of free fatty acids (FFAs) in the liver. This occurs due to an increase in the influx of FFAs from peripheral adipose tissue by the activation of hormone-sensitive lipase. In addition, there is de novo increased lipogenesis, a transcription factor, the sterols regulatory element-binding transcription factor 1c (SREBP-1c), which activates the expression of several genes strongly promotes lipogenesis by the liver and facilitate storage of triglycerides. Lipids accumulation in the liver induces a chronic stress in the endoplasmic reticulum of the hepatocytes. Genome-wide association studies have identified genetic variants associated with NAFLD severity, but unrelated to IR. In particular, the alteration of patatin-like phospholipase domain-containing protein 3 contributes to the susceptibility to NAFLD. Furthermore, the lipotoxicity of ceramides and diacylglycerol, well known in T2DM, triggers a chronic inflammatory process favoring the progression from hepatic steatosis to steatohepatitis. Reactive oxygen species produced by mitochondrial dysfunction trigger both liver inflammation and beta-cells damage, promoting the progression of both NAFLD and T2DM. The close association between NAFLD and T2DM is bidirectional, as T2DM may trigger both NAFLD onset and its progression, but NAFLD itself may contribute to the development of IR and T2DM. Future studies on the mechanisms will have to deepen the knowledge of the interaction between the two pathologies and should allow the identification of new therapeutic targets for the treatment of NAFLD, currently substantially absent.
Collapse
Affiliation(s)
- Carlo Acierno
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Riccardo Nevola
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Luigi Elio Adinolfi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Italy
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, I-80138 Naples, Ital
| |
Collapse
|
136
|
Abstract
PURPOSE OF REVIEW To provide an overview of mediators involved in the pathogenesis of postacute pancreatitis diabetes mellitus. RECENT FINDINGS The 'holistic prevention of pancreatitis' framework has brought to the fore the sequelae of not only end-stage chronic pancreatitis and extensive pancreatic necrosis but also mild acute pancreatitis. Insights from the DORADO project have provided a wealth of information on the signalling molecules that do and do not affect glucose metabolism in individuals after mild acute pancreatitis and have challenged conventional views of the pathogenesis of postpancreatitis diabetes mellitus. SUMMARY Growing evidence compels a reconsideration of the dogma that mechanical β-cell destruction (and the resulting insulin deficiency) is the only underlying mechanism of postpancreatitis diabetes mellitus. Chronic low-grade inflammation, β-cell compensation, lipolysis, altered secretion of gut hormones, and changes in iron metabolism characterize postacute pancreatitis diabetes mellitus. Some of these are druggable targets that offer novel opportunities to reduce the burden of pancreatitis through tertiary prevention.
Collapse
|
137
|
Endometrial Cancer as a Metabolic Disease with Dysregulated PI3K Signaling: Shedding Light on Novel Therapeutic Strategies. Int J Mol Sci 2020; 21:ijms21176073. [PMID: 32842547 PMCID: PMC7504460 DOI: 10.3390/ijms21176073] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Endometrial cancer (EC) is one of the most common malignancies of the female reproductive organs. The most characteristic feature of EC is the frequent association with metabolic disorders. However, the components of these disorders that are involved in carcinogenesis remain unclear. Accumulating epidemiological studies have clearly revealed that hyperinsulinemia, which accompanies these disorders, plays central roles in the development of EC via the insulin-phosphoinositide 3 kinase (PI3K) signaling pathway as a metabolic driver. Recent comprehensive genomic analyses showed that over 90% of ECs have genomic alterations in this pathway, resulting in enhanced insulin signaling and production of optimal tumor microenvironments (TMEs). Targeting PI3K signaling is therefore an attractive treatment strategy. Several clinical trials for recurrent or advanced ECs have been attempted using PI3K-serine/threonine kinase (AKT) inhibitors. However, these agents exhibited far lower efficacy than expected, possibly due to activation of alternative pathways that compensate for the PIK3-AKT pathway and allow tumor growth, or due to adaptive mechanisms including the insulin feedback pathway that limits the efficacy of agents. Overcoming these responses with careful management of insulin levels is key to successful treatment. Further interest in specific TMEs via the insulin PI3K-pathway in obese women will provide insight into not only novel therapeutic strategies but also preventive strategies against EC.
Collapse
|
138
|
Gerges SH, Wahdan SA, Elsherbiny DA, El-Demerdash E. Diosmin ameliorates inflammation, insulin resistance, and fibrosis in an experimental model of non-alcoholic steatohepatitis in rats. Toxicol Appl Pharmacol 2020; 401:115101. [PMID: 32512072 DOI: 10.1016/j.taap.2020.115101] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/30/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is becoming of increasing significance due to its growing global prevalence and risk of progression to end-stage liver disease. This study was carried out to investigate the potential anti-inflammatory, insulin sensitizing, and antifibrotic effects of diosmin in an experimental model of NASH induced in rats using high-fat diet (HFD) and 30 mg/kg streptozotocin (STZ). Diosmin was administered orally at dose of 100 mg/kg for 8 weeks. Stained tissue sections were examined for histopathological signs of NASH, collagen deposition, and alpha smooth muscle actin (α-SMA) expression. In addition, insulin resistance, dyslipidemia, inflammation, and fibrosis markers were assessed. HFD/STZ successfully induced different NASH features such as insulin resistance seen by elevated fasting blood glucose levels and homeostasis model assessment for insulin resistance. Moreover, induced rats demonstrated dyslipidemia, a significant elevation in tumor necrosis factor alpha (TNF-α) and interleukin-6 levels, and an imbalance in the oxidative status of the liver. Those events altogether precipitated initiation of liver fibrosis as confirmed by elevated transforming growth factor beta (TGF-β) levels. Treatment with diosmin demonstrated multiple beneficial effects as it significantly ameliorated histopathological NASH findings, lowered TNF-α, interleukin-6, and malondialdehyde levels, improved lipid and glucose metabolism, and lowered hepatic TGF-β, α-SMA, and collagen content compared to untreated rats. The present study represents a drug repositioning scenario as diosmin is widely used for management of blood vessel disorders and is known to be well tolerated. This encourages the extension of our study to the clinical setting to explore diosmin effects in NASH patients.
Collapse
Affiliation(s)
- Samar H Gerges
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Abbasia, Cairo 11566, Egypt
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Abbasia, Cairo 11566, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Abbasia, Cairo 11566, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Abbasia, Cairo 11566, Egypt.
| |
Collapse
|
139
|
Bastin M, Andreelli F. [Corticosteroid-induced diabetes: Novelties in pathophysiology and management]. Rev Med Interne 2020; 41:607-616. [PMID: 32782164 DOI: 10.1016/j.revmed.2020.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 01/08/2023]
Abstract
Diabetes frequently occurs during corticosteroid treatment, sometimes necessitating urgent therapeutic management, with insulin for example. Corticosteroids induce insulin resistance in the liver, adipocytes and skeletal muscle, and have direct deleterious effects on insulin secretion. The development of insulin resistance during corticosteroid treatment, and the insufficient adaptation of insulin secretion, are key elements in the pathophysiology of corticosteroid-induced diabetes. The capacity of pancreatic β-cells to increase insulin secretion in response to insulin resistance is partly genetically determined. A familial history of type 2 diabetes is, therefore, a major risk factor for diabetes development on corticosteroid treatment. Corticosteroid treatments are usually initiated at a fairly high dose, which is subsequently decreased to the lowest level sufficient to achieve disease control. Pharmacological management of diabetes is needed in patients with blood glucose levels exceeding 2.16 g/l (12 mmol/l) and insulin therapy can be started when blood glucose levels are higher than 3.6 g/l (20 mmol/l) with clinical symptoms of diabetes. Insulin can then be replaced with oral hypoglycemic compounds when both blood glucose levels and corticosteroid dose have decreased. Patient education is essential, particularly for the management of hypoglycemia when corticosteroids are withdrawn or their dose tapered.
Collapse
Affiliation(s)
- M Bastin
- CHU Pitié-Salpêtrière, Service de diabétologie-métabolismes, Assistance Publique Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - F Andreelli
- CHU Pitié-Salpêtrière, Service de diabétologie-métabolismes, Assistance Publique Hôpitaux de Paris, Sorbonne Université, Paris, France.
| |
Collapse
|
140
|
Diversity of Gut Microbiota and Bifidobacterial Community of Chinese Subjects of Different Ages and from Different Regions. Microorganisms 2020; 8:microorganisms8081108. [PMID: 32722057 PMCID: PMC7464982 DOI: 10.3390/microorganisms8081108] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/13/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota composition and functionality are closely linked to host health. In this study, the fecal microbiota and bifidobacterial communities of 111 healthy volunteers from four regions of China of varying age profiles (Child, 1–5 years; Young, 18–50 years; Elder, 60–80 years; Longevity, ≥90 years) were investigated via high-throughput sequencing. Canonical analysis revealed that the gut microbiota, as well as bifidobacteria profiles of the subjects, clustered according to their regions and age. Eight genera were shared among all subjects, however, certain genera distributed differently in subjects grouped by region and age. Faecalibacterium was enriched in samples from Zhongxiang, unclassified Ruminococcaceae and Christensenellaceae were enriched in the Longevity group, and Bifidobacterium was enriched in Child. Within Bifidobacterium, B. longum was the most abundant species in almost all samples except for Child, in which B. pseudocatenulatum was the most abundant. Additionally, the abundances of B. adolescentis and B. dentium were lower in Child. In conclusion, our results suggest that geography and age affect the structure of the gut microbiota, as well as Bifidobacterium composition, and this variation may greatly associate with the metabolic and immune changes that occur during the process of aging.
Collapse
|
141
|
Azevedo Foinquinos G, Azevedo Acioli ME, Santana Cavalcanti AH, Barbosa Junior WL, Lima RE, Juca NT, de Azevedo Foinquinos RC, Rocha da Cruz C, Fernandez Pereira FM, de Carvalho SR, de Mendonça Belmont TF, Vasconcelos LRS, Beltrão Pereira LMM. Influence of LGALS3 and PNPLA3 genes in non-alcoholic steatohepatitis (NASH) in patients undergone bariatric surgery. Obes Res Clin Pract 2020; 14:326-332. [PMID: 32690320 DOI: 10.1016/j.orcp.2020.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/10/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
AIM This study evaluated the genesPNPLA3 and LGALS3 in patients who have undergone bariatric surgery. METHODS Individuals with NAFLD and NASH were evaluated, the DNA was extracted from total blood for genotyping of rs4644, rs4652 from LGALS3 and rs738409 from PNPLA3 genes, the total RNA was obtained from liver biopsy. For the detection of the molecular targets, real-time PCR through Taqman probes was used. RESULTS From a total of 46 collected patients, of those 21 (456%) were included as NASH and 25 (544%) as steatosis group. This groups showed significant difference to aspartate aminotransferase (AST), alanine aminotransferase (ALT) and Glutamyl transpeptidase (GGT) (p = 0.0108, p = 0.0090 and p = 0.0044). Regarding to gene expression in studied groups, hepatic steatosis vs NASH, we observed a higher expression of the LGALS3 gene in NASH (p = 0.0273). In addition, patients with C allele in homozygous for rs4644 and rs4652 of LGALS3 gene had higher expression, in NASH group (p = 0.0500 and p = 0.0242, respectively), furthermore for rs4644 both alleles in homozygous showed higher expression (AA/CC vs AC) (p = 0.0500), when analyzed PNPLA3 rs738409, NASH patients with G allele in homozygous had higher expression (p = 0.0494). CONCLUSIONS Therefore, an increased expression of the LGALS3 gene in patients with NASH may be important in the etiopathogenesis of the disease, as well as the presence of rs4652 and rs4644 SNPs in the regulation of transcriptional levels of the gene in patients with NAFLD and NASH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Luydson Richardson Silva Vasconcelos
- School of Medical Sciences, University of Pernambuco, Brazil; Aggeu Magalhães Institute - IAM- FIOCRUZ-PE, Brazil; Institute of Liver and Transplant of Pernambuco, Brazil.
| | | |
Collapse
|
142
|
Pyrina I, Chung KJ, Michailidou Z, Koutsilieris M, Chavakis T, Chatzigeorgiou A. Fate of Adipose Progenitor Cells in Obesity-Related Chronic Inflammation. Front Cell Dev Biol 2020; 8:644. [PMID: 32760729 PMCID: PMC7372115 DOI: 10.3389/fcell.2020.00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Adipose progenitor cells, or preadipocytes, constitute a small population of immature cells within the adipose tissue. They are a heterogeneous group of cells, in which different subtypes have a varying degree of commitment toward diverse cell fates, contributing to white and beige adipogenesis, fibrosis or maintenance of an immature cell phenotype with proliferation capacity. Mature adipocytes as well as cells of the immune system residing in the adipose tissue can modulate the function and differentiation potential of preadipocytes in a contact- and/or paracrine-dependent manner. In the course of obesity, the accumulation of immune cells within the adipose tissue contributes to the development of a pro-inflammatory microenvironment in the tissue. Under such circumstances, the crosstalk between preadipocytes and immune or parenchymal cells of the adipose tissue may critically regulate the differentiation of preadipocytes into white adipocytes, beige adipocytes, or myofibroblasts, thereby influencing adipose tissue expansion and adipose tissue dysfunction, including downregulation of beige adipogenesis and development of fibrosis. The present review will outline the current knowledge about factors shaping cell fate decisions of adipose progenitor cells in the context of obesity-related inflammation.
Collapse
Affiliation(s)
- Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Zoi Michailidou
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Antonios Chatzigeorgiou
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
143
|
Tanataweethum N, Zhong F, Trang A, Lee C, Cohen RN, Bhushan A. Towards an Insulin Resistant Adipose Model on a Chip. Cell Mol Bioeng 2020; 14:89-99. [PMID: 33643468 DOI: 10.1007/s12195-020-00636-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 07/07/2020] [Indexed: 12/25/2022] Open
Abstract
Introduction Adipose tissue and adipocytes are primary regulators of insulin sensitivity and energy homeostasis. Defects in insulin sensitivity of the adipocytes predispose the body to insulin resistance (IR) that could lead to diabetes. However, the mechanisms mediating adipocyte IR remain elusive, which emphasizes the need to develop experimental models that can validate the insulin signaling pathways and discover new mechanisms in the search for novel therapeutics. Currently in vitro adipose organ-chip devices show superior cell function over conventional cell culture. However, none of these models represent disease states. Only when these in vitro models can represent both healthy and disease states, they can be useful for developing therapeutics. Here, we establish an organ-on-chip model of insulin-resistant adipocytes, as well as characterization in terms of insulin signaling pathway and lipid metabolism. Methods We differentiated, maintained, and induced insulin resistance into primary adipocytes in a microfluidic organ-on-chip. We then characterized IR by looking at the insulin signaling pathway and lipid metabolism, and validated by studying a diabetic drug, rosiglitazone. Results We confirmed the presence of insulin resistance through reduction of Akt phosphorylation, Glut4 expression, Glut4 translocation and glucose uptake. We also confirmed defects of disrupted insulin signaling through reduction of lipid accumulation from fatty acid uptake and elevation of glycerol secretion. Testing with rosiglitazone showed a significant improvement in insulin sensitivity and fatty acid metabolism as suggested by previous reports. Conclusions The adipose-chip exhibited key characteristics of IR and can serve as model to study diabetes and facilitate discovery of novel therapeutics.
Collapse
Affiliation(s)
- Nida Tanataweethum
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
| | - Franklin Zhong
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
| | - Allyson Trang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
| | - Chaeeun Lee
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
| | - Ronald N Cohen
- Section of Endocrinology, Department of Medicine, The University of Chicago, Chicago, IL 60637 USA
| | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
| |
Collapse
|
144
|
Abstract
The skeletal muscle is the largest organ in the body, by mass. It is also the regulator of glucose homeostasis, responsible for 80% of postprandial glucose uptake from the circulation. Skeletal muscle is essential for metabolism, both for its role in glucose uptake and its importance in exercise and metabolic disease. In this article, we give an overview of the importance of skeletal muscle in metabolism, describing its role in glucose uptake and the diseases that are associated with skeletal muscle metabolic dysregulation. We focus on the role of skeletal muscle in peripheral insulin resistance and the potential for skeletal muscle-targeted therapeutics to combat insulin resistance and diabetes, as well as other metabolic diseases like aging and obesity. In particular, we outline the possibilities and pitfalls of the quest for exercise mimetics, which are intended to target the molecular mechanisms underlying the beneficial effects of exercise on metabolic disease. We also provide a description of the molecular mechanisms that regulate skeletal muscle glucose uptake, including a focus on the SNARE proteins, which are essential regulators of glucose transport into the skeletal muscle. © 2020 American Physiological Society. Compr Physiol 10:785-809, 2020.
Collapse
Affiliation(s)
- Karla E. Merz
- Department of Molecular and Cellular Endocrinology, City of Hope Beckman Research Institute, Duarte, California, USA
- The Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, California, USA
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, City of Hope Beckman Research Institute, Duarte, California, USA
| |
Collapse
|
145
|
Rostamtabar M, Esmaeilzadeh S, Tourani M, Rahmani A, Baee M, Shirafkan F, Saleki K, Mirzababayi SS, Ebrahimpour S, Nouri HR. Pathophysiological roles of chronic low-grade inflammation mediators in polycystic ovary syndrome. J Cell Physiol 2020; 236:824-838. [PMID: 32617971 DOI: 10.1002/jcp.29912] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/29/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common hormonal imbalance disease in reproductive-aged women. Its basic characteristics are ovulatory dysfunction and ovarian overproduction of androgens that lead to severe symptoms such as insulin resistance, hirsutism, infertility, and acne. Notwithstanding the disease burden, its underlying mechanisms remain unknown, and no causal therapeutic exists. In recent years, further studies showed that inflammation processes are involved in ovulation and play a key role in ovarian follicular dynamics. Visceral adipose tissue can cause inflammatory response and maintenance of the inflammation state in adipocytes by augmented production of inflammatory cytokines, monocyte chemoattractant proteins, and recruitment of the immune cell. Therefore, the PCOS can be related to a low-grade inflammation state and inflammatory markers. Investigating the inflammatory processes and mediators that contribute to the commencement and development of PCOS can be a critical step for better understanding the pathophysiology of the disease and its treatment through inhibition or control of related pathways. In the present review, we discuss the pathophysiological roles of chronic low-grade inflammation mediators including inflammasome-related cytokines, interleukin-1β (IL-1β), and IL-18 in PCOS development.
Collapse
Affiliation(s)
- Maryam Rostamtabar
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Sedigheh Esmaeilzadeh
- Infertility and Reproductive Health Research Center, Health Research Institute, Babol University of Medical Science, Babol, Iran
| | - Mehdi Tourani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abolfazl Rahmani
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Masoud Baee
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Shirafkan
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Reza Nouri
- Infertility and Reproductive Health Research Center, Health Research Institute, Babol University of Medical Science, Babol, Iran.,Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
146
|
Reilly SM, Hung CW, Ahmadian M, Zhao P, Keinan O, Gomez AV, DeLuca JH, Dadpey B, Lu D, Zaid J, Poirier B, Peng X, Yu RT, Downes M, Liddle C, Evans RM, Murphy AN, Saltiel AR. Catecholamines suppress fatty acid re-esterification and increase oxidation in white adipocytes via STAT3. Nat Metab 2020; 2:620-634. [PMID: 32694788 PMCID: PMC7384260 DOI: 10.1038/s42255-020-0217-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Catecholamines stimulate the mobilization of stored triglycerides in adipocytes to provide fatty acids (FAs) for other tissues. However, a large proportion is taken back up and either oxidized or re-esterified. What controls the disposition of these FAs in adipocytes remains unknown. Here, we report that catecholamines redirect FAs for oxidation through the phosphorylation of signal transducer and activator of transcription 3 (STAT3). Adipocyte STAT3 is phosphorylated upon activation of β-adrenergic receptors, and in turn suppresses FA re-esterification to promote FA oxidation. Adipocyte-specific Stat3 KO mice exhibit normal rates of lipolysis, but exhibit defective lipolysis-driven oxidative metabolism, resulting in reduced energy expenditure and increased adiposity when they are on a high-fat diet. This previously unappreciated, non-genomic role of STAT3 explains how sympathetic activation can increase both lipolysis and FA oxidation in adipocytes, revealing a new regulatory axis in metabolism.
Collapse
Affiliation(s)
- Shannon M Reilly
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Chao-Wei Hung
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Maryam Ahmadian
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Peng Zhao
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Omer Keinan
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Andrew V Gomez
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Julia H DeLuca
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Benyamin Dadpey
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Donald Lu
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jessica Zaid
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - BreAnne Poirier
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoling Peng
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Christopher Liddle
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Anne N Murphy
- Department of Pharmacology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Cytokinetics, South San Francisco, CA, USA
| | - Alan R Saltiel
- Division of Metabolism and Endocrinology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Pharmacology, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
147
|
Jakkawanpitak C, Hutadilok-Towatana N, Sermwittayawong D. Fungal-like particles and macrophage-conditioned medium are inflammatory elicitors for 3T3-L1 adipocytes. Sci Rep 2020; 10:9437. [PMID: 32523023 PMCID: PMC7287055 DOI: 10.1038/s41598-020-66283-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/18/2020] [Indexed: 11/09/2022] Open
Abstract
Adipocytes from white-adipose tissue are known to produce inflammatory cytokines, which play a major role in energy balance and metabolism. While they can respond to pathogen-associated molecular pattern (PAMPs) such as lipopolysaccharide (LPS) from bacteria, it is not known whether adipocytes can be stimulated by fungal cells. Previously, adipocytes were shown to produce toll-like receptor 2 (TLR2), a β-glucan receptor, suggesting that they could respond to β-glucan on the fungal cell wall. In this study, we show that heat-killed yeast induce an inflammatory response in adipocytes. Using fungal-like particles, namely laminarin-coated beads (LCB), we find that these particles trigger the expression of many key inflammatory genes in dose- and time-dependent fashions in adipocytes. These results suggest that β-glucan on the fungal cell wall is sufficient to elicit an inflammatory response in adipocytes. In addition, we show that both LCB and LCB-treated conditioned medium from RAW 264.7 murine macrophages (LCB-RM) induce the expression of those inflammatory genes through IKKβ-IκBα proteins. Together, we conclude that the fungal-like particles and the conditioned medium elicit an inflammatory response in adipocytes through the canonical or classical NF-κB pathway.
Collapse
Affiliation(s)
- Chanawee Jakkawanpitak
- Department of Biochemistry, Faculty of Science, Prince of Songkla University, Hat Yai, 90110, Thailand
| | | | - Decha Sermwittayawong
- Department of Biochemistry, Faculty of Science, Prince of Songkla University, Hat Yai, 90110, Thailand.
| |
Collapse
|
148
|
Development of novel human in vitro vascularized adipose tissue model with functional macrophages. Cytotechnology 2020; 72:665-683. [PMID: 32524217 PMCID: PMC7547925 DOI: 10.1007/s10616-020-00407-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 06/04/2020] [Indexed: 01/16/2023] Open
Abstract
Inflammation has been proven significant factor in development of type 2 diabetes. So far, most of the adipose tissue related research has been performed in animals, mainly rodent models. The relevance of translation of animal results to humans is questionable. However, in vitro model with relevant human cell source, such as human adipose tissue stromal cells (hASC), can be developed and should be utilized for human adipose tissue research. We developed in vitro models of human adipose tissue utilizing hASC, endothelial cells and monocytes/macrophages. By isolating endothelial cells and macrophages from same adipose tissue as hASC, we were able to provide method for constructing personalized models of adipose tissue. With these models, we studied the effect of macrophages on adipogenesis and protein secretion, with and without vasculature. The models were analyzed for immunocytochemical markers, cell number, triglyceride accumulation and protein secretion. We found that lipid accumulation was greater in adipocytes in the presence of macrophages. Interferon gamma increased this difference between adipocyte culture and Adipocyte-Macrophage co-culture. Protein secretion was affected more by macrophages when vasculature was not present compared to the mild effect when vasculature was present. The vascularized adipose model with macrophages is valuable tool for human adipose tissue research, especially for the personalized medicine approaches; for choosing the right treatments and for studying rare medical conditions.
Collapse
|
149
|
Mu J, Li Q. Anomalous expression of miR-103 in polycystic ovary syndrome influenced by hormonal, and metabolic variables. Exp Mol Pathol 2020; 116:104482. [PMID: 32504622 DOI: 10.1016/j.yexmp.2020.104482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/21/2020] [Accepted: 06/01/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS), a common endocrine disorder in reproductive-aged women, is correlated with obesity and insulin resistance (IR), androgens excess, chronic anovulation, and infertility. MicroRNAs (miRNAs) are small, single-stranded, noncoding RNA molecules that participate in inflammation, reproduction and metabolism, may contribute to PCOS. Current study aiming to manifest the correlation of body mass index (BMI) and testosterone (T) with miR-103 expression before and after fat loss. METHODS 46 controls (N = 23 with BMI < 24 kg/m2, N = 23 with BMI ≥ 28 kg/m2) and 46 patients with PCOS (N = 23 with BMI < 24 kg/m2, N = 23 with BMI ≥ 28 kg/m2) aged between 20 and 30 were recruited. Waist-to-hip (WHR) and Body fat% (BF%) was measured and calculated. Serum hormones, serum lipid, metabolism parameters, and serum miR-103 were measured. All the assessments were measured before and after fat loss in a three-month intervention period. RESULTS miR-103 was correlated with BMI rather than testosterone (T), and there was a significant difference between the non-obese and obese groups in miR-103 expression. Compared to before fat loss, miR-103 expression showed a slight downward trend. CONCLUSIONS Serum miR-103 differentially expressed between controls and PCOS subjects, miR-103 was positively correlated with BMI. There was significant difference between the non-obese and obese groups in miR-103 expression.
Collapse
Affiliation(s)
- Jiawei Mu
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Qiang Li
- Department of Endocrinology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
150
|
Recent advances in the role of interleukin-6 in health and disease. Curr Opin Pharmacol 2020; 52:47-51. [DOI: 10.1016/j.coph.2020.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/21/2022]
|