101
|
Exploring and Identifying Candidate Genes and Genomic Regions Related to Economically Important Traits in Hanwoo Cattle. Curr Issues Mol Biol 2022; 44:6075-6092. [PMID: 36547075 PMCID: PMC9777506 DOI: 10.3390/cimb44120414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
The purpose of the current review was to explore and summarize different studies concerning the detection and characterization of candidate genes and genomic regions associated with economically important traits in Hanwoo beef cattle. Hanwoo cattle, the indigenous premium beef cattle of Korea, were introduced for their marbled fat, tenderness, characteristic flavor, and juiciness. To date, there has been a strong emphasis on the genetic improvement of meat quality and yields, such as backfat thickness (BFT), marbling score (MS), carcass weight (CW), eye muscle area (EMA), and yearling weight (YW), as major selection criteria in Hanwoo breeding programs. Hence, an understanding of the genetics controlling these traits along with precise knowledge of the biological mechanisms underlying the traits would increase the ability of the industry to improve cattle to better meet consumer demands. With the development of high-throughput genotyping, genomewide association studies (GWAS) have allowed the detection of chromosomal regions and candidate genes linked to phenotypes of interest. This is an effective and useful tool for accelerating the efficiency of animal breeding and selection. The GWAS results obtained from the literature review showed that most positional genes associated with carcass and growth traits in Hanwoo are located on chromosomes 6 and 14, among which LCORL, NCAPG, PPARGC1A, ABCG2, FAM110B, FABP4, DGAT1, PLAG1, and TOX are well known. In conclusion, this review study attempted to provide comprehensive information on the identified candidate genes associated with the studied traits and genes enriched in the functional terms and pathways that could serve as a valuable resource for future research in Hanwoo breeding programs.
Collapse
|
102
|
Singh A, Mayengbam SS, Yaduvanshi H, Wani MR, Bhat MK. Obesity Programs Macrophages to Support Cancer Progression. Cancer Res 2022; 82:4303-4312. [PMID: 36191083 DOI: 10.1158/0008-5472.can-22-1257] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/14/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023]
Abstract
Obesity induces multifactorial effects such as dyslipidemia, insulin resistance, and arterial hypertension that influence the progression of many diseases. Obesity is associated with an increased incidence of cancers, and multiple mechanisms link obesity with cancer initiation and progression. Macrophages participate in the homeostasis of adipose tissue and play an important role in cancer. Adipose tissue expansion in obesity alters the balance between pro- and anti-inflammatory macrophages, which is a primary cause of inflammation. Chronic low-grade inflammation driven by macrophages is also an important characteristic of cancer. Adipocytes secrete various adipokines, including adiponectin, leptin, IL6, and TNFα, that influence macrophage behavior and tumor progression. Furthermore, other metabolic effects of obesity, such as hyperlipidemia, hyperglycemia, and hypercholesterolemia, can also regulate macrophage functionality in cancer. This review summarizes how obesity influences macrophage-tumor cell interactions and the role of macrophages in the response to anticancer therapies under obese conditions.
Collapse
Affiliation(s)
- Abhijeet Singh
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Shyamananda Singh Mayengbam
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Himanshi Yaduvanshi
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Mohan R Wani
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| | - Manoj Kumar Bhat
- Department of Biotechnology, National Centre for Cell Science, Government of India, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, Maharashtra, India
| |
Collapse
|
103
|
Choi W, Kim JW, Kang HJ, Kim HK, Kang HC, Lee JY, Kim SW, Stewart R, Kim JM. Interactive Effects of Serum Leptin Levels and Physical Comorbidity on the Pharmacotherapeutic Response of Depressive Disorders. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2022; 20:662-674. [PMID: 36263641 PMCID: PMC9606432 DOI: 10.9758/cpn.2022.20.4.662] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To investigate individual and interactive associations of baseline serum leptin levels and physical comorbidity with short- and long-term treatment outcomes in outpatients with depressive disorders who received stepwise antidepressant treatment in a naturalistic prospective study design. METHODS Baseline serum leptin levels were measured, and the number of concurrent physical disorders ascertained from 1,094 patients. These patients received initial antidepressant monotherapy; then, for patients with an insufficient response or who experienced uncomfortable side effects, treatment was administered using alternative strategies every 3 weeks in the acute treatment phase (at 3, 6, 9, and 12 weeks) and every 3 months in the continuation treatment phase (at 6, 9, and 12 months). Then, 12-week and 12-month remission, defined as a Hamilton Depression Rating Scale score of ≤7, was estimated. RESULTS In multivariable logistic regression analyses, individual effects were found only between higher baseline serum leptin levels and 12-week non-remission. Significant interactive effects between higher leptin levels and fewer physical disorders (< 2 physical disorders) on 12-week non-remission were observed. However, neither individual nor interactive effects between leptin levels and physical comorbidity were associated with 12-month remission. CONCLUSION The combination of serum leptin level and number of physical disorders may be a useful predictor of short-term treatment responses in patients with depressive disorders receiving pharmacotherapy.
Collapse
Affiliation(s)
- Wonsuk Choi
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ju-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Hee-Ju Kang
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ho-Cheol Kang
- Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea
| | - Ju-Yeon Lee
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Robert Stewart
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK,South London and Maudsley NHS Foundation Trust, London, UK
| | - Jae-Min Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea,Address for correspondence: Jae-Min Kim Department of Psychiatry, Chonnam National University Medical School, 160 Baekseo-ro, Dong-gu, Gwangju 61469, Korea, E-mail: , ORCID: https://orcid.org/0000-0001-7409-6306
| |
Collapse
|
104
|
Martinez-Sanchez N, Sweeney O, Sidarta-Oliveira D, Caron A, Stanley SA, Domingos AI. The sympathetic nervous system in the 21st century: Neuroimmune interactions in metabolic homeostasis and obesity. Neuron 2022; 110:3597-3626. [PMID: 36327900 PMCID: PMC9986959 DOI: 10.1016/j.neuron.2022.10.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The sympathetic nervous system maintains metabolic homeostasis by orchestrating the activity of organs such as the pancreas, liver, and white and brown adipose tissues. From the first renderings by Thomas Willis to contemporary techniques for visualization, tracing, and functional probing of axonal arborizations within organs, our understanding of the sympathetic nervous system has started to grow beyond classical models. In the present review, we outline the evolution of these findings and provide updated neuroanatomical maps of sympathetic innervation. We offer an autonomic framework for the neuroendocrine loop of leptin action, and we discuss the role of immune cells in regulating sympathetic terminals and metabolism. We highlight potential anti-obesity therapeutic approaches that emerge from the modern appreciation of SNS as a neural network vis a vis the historical fear of sympathomimetic pharmacology, while shifting focus from post- to pre-synaptic targeting. Finally, we critically appraise the field and where it needs to go.
Collapse
Affiliation(s)
| | - Owen Sweeney
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Davi Sidarta-Oliveira
- Physician-Scientist Graduate Program, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.
| |
Collapse
|
105
|
Normand E, Franco A, Alos N, Parent S, Moreau A, Marcil V. Circulatory Adipokines and Incretins in Adolescent Idiopathic Scoliosis: A Pilot Study. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1619. [PMID: 36360347 PMCID: PMC9688531 DOI: 10.3390/children9111619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 08/26/2023]
Abstract
Adolescent idiopathic scoliosis (AIS) is a three-dimensional malformation of the spine of unknown cause that develops between 10 and 18 years old and affects 2-3% of adolescents, mostly girls. It has been reported that girls with AIS have a taller stature, lower body mass index (BMI), and bone mineral density (BMD) than their peers, but the causes remain unexplained. Energy metabolism discrepancies, including alterations in adipokine and incretin circulatory levels, could influence these parameters and contribute to disease pathophysiology. This pilot study aims to compare the anthropometry, BMD, and metabolic profile of 19 AIS girls to 19 age-matched healthy controls. Collected data include participants' fasting metabolic profile, anthropometry (measurements and DXA scan), nutritional intake, and physical activity level. AIS girls (14.8 ± 1.7 years, Cobb angle 27 ± 10°), compared to controls (14.8 ± 2.1 years), were leaner (BMI-for-age z-score ± SD: -0.59 ± 0.81 vs. 0.09 ± 1.11, p = 0.016; fat percentage: 24.4 ± 5.9 vs. 29.2 ± 7.2%, p = 0.036), had lower BMD (total body without head z-score ± SD: -0.6 ± 0.83 vs. 0.23 ± 0.98, p = 0.038; femoral neck z-score: -0.54 ± 1.20 vs. 0.59 ± 1.59, p = 0.043), but their height was similar. AIS girls had higher adiponectin levels [56 (9-287) vs. 32 (7-74) μg/mL, p = 0.005] and lower leptin/adiponectin ratio [0.042 (0.005-0.320) vs. 0.258 (0.024-1.053), p = 0.005]. AIS participants with a Cobb angle superior to 25° had higher resistin levels compared to controls [98.2 (12.8-287.2) vs. 32.1 (6.6-73.8), p = 0.0013]. This pilot study suggests that adipokines are implicated in AIS development and/or progression, but more work is needed to confirm their role in the disease.
Collapse
Affiliation(s)
- Emilie Normand
- Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Anita Franco
- Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Viscogliosi Laboratory in Molecular Genetics and Musculoskeletal Diseases, Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
| | - Nathalie Alos
- Endocrine Service, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC H3T 1J4, Canada
| | - Stefan Parent
- Department of Surgery, CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
| | - Alain Moreau
- Viscogliosi Laboratory in Molecular Genetics and Musculoskeletal Diseases, Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montreal, QC H3A 1J4, Canada
| | - Valérie Marcil
- Research Center of the CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
106
|
Su H, Meng C, Xu J, Su Z, Xiao C, Yang D. Histone methyltransferase Smyd2 drives adipogenesis via regulating STAT3 phosphorylation. Cell Death Dis 2022; 13:890. [PMID: 36270984 PMCID: PMC9586978 DOI: 10.1038/s41419-022-05321-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 01/23/2023]
Abstract
Adipogenesis is a complex cascade involved with the preadipocytes differentiation towards mature adipocytes, accelerating the onset of obesity. Histone methyltransferase SET and MYND domain-containing protein 2 (Smyd2), is involved in a variety of cellular biological functions but the epigenetic regulation of Smyd2 in adipogenesis and adipocyte differentiation remains unclear. Both Smyd2 siRNA and LLY-507, an inhibitor of Smyd2, were used to examine the effect of Smyd2 on adipogenesis and adipocyte differentiation in vitro. Smyd2 heterozygous knockout (Smyd2+/-) mice were also constructed to validate the relationship between Smyd2 and adipogenesis in vivo. We found that Smyd2 is abundant in white adipose tissue and closely correlated with adipocyte differentiation. Knockdown or inhibition of Smyd2 restrained adipocyte differentiation in vitro, which requires the phosphorylation of STAT3. In vivo functional validation, Smyd2+/- mice exert significant fat loss but not susceptible to HFD-induced obesity. Taken together, our findings revealed that Smyd2 is a novel regulator of adipocyte differentiation by regulating the phosphorylation of STAT3, which provides insights into the effects of epigenetic regulation in adipogenesis. Inhibition of Smyd2 might represent a viable strategy for anti-adipogenesis and maybe further alleviate obesity-related diseases in humans.
Collapse
Affiliation(s)
- Haibi Su
- grid.8547.e0000 0001 0125 2443Pharmacophenomics Laboratory, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203 P. R. China
| | - Chen Meng
- grid.8547.e0000 0001 0125 2443Pharmacophenomics Laboratory, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203 P. R. China
| | - Jie Xu
- grid.8547.e0000 0001 0125 2443Pharmacophenomics Laboratory, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203 P. R. China
| | - Zhenghua Su
- grid.8547.e0000 0001 0125 2443Pharmacophenomics Laboratory, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203 P. R. China
| | - Chenxi Xiao
- grid.8547.e0000 0001 0125 2443Pharmacophenomics Laboratory, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203 P. R. China
| | - Di Yang
- grid.8547.e0000 0001 0125 2443Pharmacophenomics Laboratory, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 201203 P. R. China
| |
Collapse
|
107
|
Haidari F, Elahikhah M, Shariful Islam SM, Mohammadshahi M, Shahbazian H, Aghamohammadi V. Effects of milk protein concentrate supplementation on metabolic parameters, adipocytokines and body composition in obese women under weight-loss diet: study protocol for a randomised controlled trial. BMJ Open 2022; 12:e064727. [PMID: 36202586 PMCID: PMC9540842 DOI: 10.1136/bmjopen-2022-064727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Obesity impairs metabolic function and increases the risk of cardiovascular disease and type 2 diabetes mellitus. Evidence suggests that high-protein diets help to increase weight loss and protect against weight gain. Milk protein concentrate (MPC) is a dairy product with a high protein content with a ratio of casein and whey protein similar to skim milk. This trial aims to evaluate the effect of MPC supplementation in obese women under a weight-loss diet. METHODS AND ANALYSIS We will conduct a 2-month open-label, parallel-group, randomised controlled trial to determine the effect of MPC supplementation on levels of glycaemic and lipid profile, leptin, adiponectin, appetite, waist circumference, body mass index and body composition in 44 premenopausal obese women on a weight-loss diet. ETHICS AND DISSEMINATION This protocol, approved by the Medical Ethics Committee of Ahvaz University of Medical Sciences, is in accordance with the Declaration of Helsinki (approval number: IR.AJUMS.REC.1399.795). The trial results will be published in peer-reviewed journals. TRIAL REGISTRATION NUMBER Iranian Registry of Clinical Trials (IRCT20201223049804N1).
Collapse
Affiliation(s)
- Fatemeh Haidari
- Department of Nutrition, Nutrition and Metabolic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahsa Elahikhah
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Majid Mohammadshahi
- Department of Nutrition, Nutrition and Metabolic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hajieh Shahbazian
- Diabetes Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
108
|
Atawia RT, Faulkner JL, Mehta V, Austin A, Jordan CR, Kennard S, Belin de Chantemèle EJ. Endothelial leptin receptor is dispensable for leptin-induced sympatho-activation and hypertension in male mice. Vascul Pharmacol 2022; 146:107093. [PMID: 35914636 PMCID: PMC9561021 DOI: 10.1016/j.vph.2022.107093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 10/16/2022]
Abstract
Leptin plays a crucial role in blood pressure (BP) regulation, notably in the context of obesity through central sympatho-mediated pressor effects. Leptin also relaxes arteries via endothelial (EC) leptin receptor (LepREC)-mediated increases in nitric oxide (NO) bioavailability. Herein, we investigated whether leptin-mediated increases in NO bioavailability represent a buffering mechanism against leptin-induced sympatho-activation. We tested the direct contribution of LepREC to BP regulation in physiological conditions and in response to chronic leptin infusion using mice deficient in LepREC. LepREC deficiency did not alter baseline metabolic profile nor leptin-induced reduction in adiposity and increases in energy expenditure. LepREC-/- mice demonstrated no increase in baseline BP and heart rate (HR) (MAP: LepREC+/+:94.7 ± 1.6, LepREC-/-:95.1 ± 1.8 mmHg; HR:LepREC+/+:492.4 ± 11.7, LepREC-/-:509.5 ± 13.4 bpm) nor in response to leptin (MAP, LepREC+/+:101.1 ± 1.7, LepREC-/-:101.7 ± 1.8 mmHg; HR, LepREC+/+:535.6 ± 11.1, LepREC-/-:539.3 ± 14.2 bpm). Moreover, baseline neurogenic control of BP and HR was preserved in LepREC-/- mice as well as leptin-mediated increases in sympathetic control of BP and HR and decreases in vagal tone. Remarkably, LepREC deficiency did not alter endothelium-dependent relaxation in resistance vessels, nor NO contribution to vasodilatation. Lastly, leptin induced similar increases in adrenergic contractility in mesenteric arteries from both LepREC+/+ and LepREC-/- mice. Collectively, these results demonstrate that the NO buffering effects of leptin are absent in resistance arteries and do not contribute to BP regulation. We provide further evidence that leptin-mediated hypertension involves increased vascular sympatho-activation and extend these findings by demonstrating for the first time that increased cardiac sympatho-activation and reduced vagal tone also contribute to leptin-mediated hypertension.
Collapse
Affiliation(s)
- Reem T Atawia
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Jessica L Faulkner
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Vinay Mehta
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Andrew Austin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Coleton R Jordan
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Simone Kennard
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, Georgia
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, Georgia; Departments of Medicine (Cardiology), Medical College of Georgia, Augusta University, Augusta, GA, Georgia.
| |
Collapse
|
109
|
Jiang T, Li Y, Li L, Liang T, Du M, Yang L, Yang J, Yang R, Zhao H, Chen M, Ding Y, Zhang J, Wang J, Xie X, Wu Q. Bifidobacterium longum 070103 Fermented Milk Improve Glucose and Lipid Metabolism Disorders by Regulating Gut Microbiota in Mice. Nutrients 2022; 14:nu14194050. [PMID: 36235706 PMCID: PMC9573661 DOI: 10.3390/nu14194050] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 12/08/2022] Open
Abstract
Background: Fermented milk is beneficial for metabolic disorders, while the underlying mechanisms of action remain unclear. This study explored the benefits and underlying mechanisms of Bifidobacterium longum 070103 fermented milk (BLFM) in thirteen-week high-fat and high-sugar (HFHS) fed mice using omics techniques. Methods and results: BLFM with activated glucokinase (GK) was screened by a double-enzyme coupling method. After supplementing BLFM with 10 mL/kg BW per day, fasting blood glucose, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and leptin were significantly reduced compared with the HFHS group. Among them, the final body weight (BW), epididymal fat, perirenal fat, and brown fat in BLFM group had better change trends than Lacticaseibacillus rhamnosus GG fermented milk (LGGFM) group. The amplicon and metabolomic data analysis identified Bifibacterium as a key gut microbiota at regulating glycolipid metabolism. BLFM reverses HFHS-induced reduction in bifidobacteria abundance. Further studies showed that BLFM significantly reduces the content of 3-indoxyl sulofphate associated with intestinal barrier damage. In addition, mice treated with BLFM improved BW, glucose tolerance, insulin resistance, and hepatic steatosis. Conclusion: BLFM consumption attenuates obesity and related symptoms in HFHS-fed mice probably via the modulation of gut microbes and metabolites.
Collapse
Affiliation(s)
- Tong Jiang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Ying Li
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Longyan Li
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Tingting Liang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Mingzhu Du
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Lingshuang Yang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Juan Yang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Runshi Yang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Hui Zhao
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Moutong Chen
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Yu Ding
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Jumei Zhang
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| | - Xinqiang Xie
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| | - Qingping Wu
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Correspondence: (J.W.); (X.X.); (Q.W.)
| |
Collapse
|
110
|
Zhang LM, Liu Z, Wang JQ, Li RQ, Ren JY, Gao X, Lv SS, Liang LY, Zhang F, Yin BW, Sun Y, Tian H, Zhu HC, Zhou YT, Ma YX. Randomized controlled trial for time-restricted eating in overweight and obese young adults. iScience 2022; 25:104870. [PMID: 36034217 PMCID: PMC9400087 DOI: 10.1016/j.isci.2022.104870] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 07/05/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Time-restricted eating (TRE) is known to improve metabolic health, whereas very few studies have compared the effects of early and late TRE (eTRE and lTRE) on metabolic health. Overweight and obese young adults were randomized to 6-h eTRE (eating from 7 a.m. to 1 p.m.) (n = 21), 6-h lTRE (eating from 12 p.m. to 6 p.m.) (n = 20), or a control group (ad libitum intake in a day) (n = 19). After 8 weeks, 6-h eTRE and lTRE produced comparable body weight loss compared with controls. Compared with control, 6-h eTRE reduced systolic blood pressure, mean glucose, fasting insulin, insulin resistance, leptin, and thyroid axis activity, whereas lTRE only reduced leptin. These findings shed light on the promise of 6-h eTRE and lTRE for weight loss. Larger studies are needed to assess the promise of eTRE to yield better thyroid axis modulation and overall cardiometabolic health improvement.
Collapse
Affiliation(s)
- Li-min Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Zhan Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jia-qi Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Rui-qiang Li
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Jing-yi Ren
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Xian Gao
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Shuai-shuai Lv
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Lu-yao Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Fan Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Bo-wen Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Yan Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Hao Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Hui-chen Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Yu-tian Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| | - Yu-xia Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Hebei Key Laboratory of Environment and Human Health, Shijiazhuang 050017, China
| |
Collapse
|
111
|
Effect of Gender on Serum Leptin in Type 2 Diabetes Mellitus: A System Review and Meta-Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4875799. [PMID: 36124171 PMCID: PMC9482490 DOI: 10.1155/2022/4875799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022]
Abstract
Objective To assess the effect of gender factors on serum leptin levels in patients with diabetes mellitus. Methods To remove any studies that indicated a relationship between leptin-based inflammatory variables and the prevalence of type 2 diabetes in particular patient categories, a comprehensive search of all articles published between July 2019 and June 2021 was performed on PubMed/MEDLINE, Web of Science, Scopus, and EBSCO Host, including Academic Search Premier, Africa-Wide Information, and Cumulative Index to Nursing and Allied Health Literature. A summary description of the combined analysis across multiple centers, regions, and continents will help us better understand the effect of gender on serum leptin levels in patients with diabetes. The meta-analysis was performed using RevMan 5.2 software on the literature that satisfied the inclusion and exclusion criteria. Results Plasma CRP levels in women with type 2 diabetes were found to be no different from those in males with type 2 diabetes, with an OR of 0.12, 95 percent confidence interval (CI) of 0.12 to 0.12, P = 0.01. There was no statistically significant difference in the plasma level of interleukin-6 (IL-6) between women with type 2 diabetes and males with type 2 diabetes However, the “inverted funnel” diagram is asymmetrical, indicating a publication bias in the included studies, despite the fact that there was no statistically significant difference in abnormal leptin levels between men with type 2 diabetes and women patients (OR = −0.69, 95 percent CI (0.88, 1.00), P < 0.05). Conclusion Gender factors did not affect the level of inflammatory factors and leptin level in type 2 diabetes.
Collapse
|
112
|
Martinez-Fierro ML, Ayala-Haro AE, Pinedo-Hurtado ME, Solis-Galvan JA, Garza-Veloz I, Velazquez-Lopez ZY, Camacho-Martinez AG, Avila-Carrasco L, Vazquez-Reyes S, Velasco-Elizondo P, Mauricio-Gonzalez A, Ortiz-Castro Y. Usefulness of a Mobile Application (Mentali) for Anxiety and Depression Screening in Medical Students and Description of the Associated Triggering Factors. Brain Sci 2022; 12:1223. [PMID: 36138959 PMCID: PMC9496953 DOI: 10.3390/brainsci12091223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 12/26/2022] Open
Abstract
The impact of the COVID-19 health crisis on the mental health of the population requires the implementation of new primary screening strategies of mental health disorders to intervene in a timelier manner, and technology may provide solutions. We aimed to evaluate the usefulness of the mobile app Mentali (version 1.1.2; creators: Jorge Alfonso Solís Galván Sodel Vázquez Reyes, Margarita de la Luz Martínez Fierro, Perla Velasco Elizondo, Idalia Garza Veloz, Alejandro Mauricio González and Claudia Caldera Villalobos, Zacatecas, México) as a primary screening tool for anxiety and depression disorders in medical students and to assess the triggering risk factors. This was a descriptive and longitudinal study and included 155 Mexican medical students. Participants interacted with Mentali for 6 months. The mobile app integrated the Beck anxiety and depression inventories together with a mood module. At the end of the interaction, the students received psychological and psychiatric interventions to confirm their primary diagnoses. Symptoms of moderate/severe anxiety and depression were present in 62.6% and 54.6% of the studied population. When corroborating the diagnoses, Mentali obtained a sensitivity of 100%, 95%, and 43% to classify a mental health disorder, anxiety, and depression, respectively. The most important triggers found were as follows: belonging to a dysfunctional family, being introverted, and having suffered from bullying. The proportion of users with excellent/good mood decreased from 78.7% to 34.4% at the end of the semester, and the proportion of users who claimed to have bad/very bad mood increased from 7.4% to 34.4% at the end of the semester (p < 0.05). Mentali was useful for identifying users with anxiety and/or depression, and as an auxiliary tool to coordinate the provision of specialized interventions, allowing us to increase the proportion of patients who needed psychological care and received it by 30%. The efficacy of Mentali in identifying activities through time with an impact on the mood and mental health of the users was confirmed. Our results support the use of Mentali for the primary screening of mental health disorders in young adults, including medical students.
Collapse
Affiliation(s)
- Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Anayantzin E. Ayala-Haro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Martha E. Pinedo-Hurtado
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Jorge A. Solis-Galvan
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Zihomara Y. Velazquez-Lopez
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Antonio G. Camacho-Martinez
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Lorena Avila-Carrasco
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Sodel Vazquez-Reyes
- Academic Unit of Electrical Engineering, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Perla Velasco-Elizondo
- Academic Unit of Electrical Engineering, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Alejandro Mauricio-Gonzalez
- Academic Unit of Electrical Engineering, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| | - Yolanda Ortiz-Castro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico
| |
Collapse
|
113
|
Amorim MR, Aung O, Mokhlesi B, Polotsky VY. Leptin-mediated neural targets in obesity hypoventilation syndrome. Sleep 2022; 45:zsac153. [PMID: 35778900 PMCID: PMC9453616 DOI: 10.1093/sleep/zsac153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/20/2022] [Indexed: 07/30/2023] Open
Abstract
Obesity hypoventilation syndrome (OHS) is defined as daytime hypercapnia in obese individuals in the absence of other underlying causes. In the United States, OHS is present in 10%-20% of obese patients with obstructive sleep apnea and is linked to hypoventilation during sleep. OHS leads to high cardiorespiratory morbidity and mortality, and there is no effective pharmacotherapy. The depressed hypercapnic ventilatory response plays a key role in OHS. The pathogenesis of OHS has been linked to resistance to an adipocyte-produced hormone, leptin, a major regulator of metabolism and control of breathing. Mechanisms by which leptin modulates the control of breathing are potential targets for novel therapeutic strategies in OHS. Recent advances shed light on the molecular pathways related to the central chemoreceptor function in health and disease. Leptin signaling in the nucleus of the solitary tract, retrotrapezoid nucleus, hypoglossal nucleus, and dorsomedial hypothalamus, and anatomical projections from these nuclei to the respiratory control centers, may contribute to OHS. In this review, we describe current views on leptin-mediated mechanisms that regulate breathing and CO2 homeostasis with a focus on potential therapeutics for the treatment of OHS.
Collapse
Affiliation(s)
- Mateus R Amorim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - O Aung
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Babak Mokhlesi
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Vsevolod Y Polotsky
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
114
|
Gündüz B, Okimoto DK. Methyl donor supplementation alters serum leptin levels and increases appetite but not body weight in cross-fostered male Syrian hamster offspring (Mesocricetus auratus). J Anim Physiol Anim Nutr (Berl) 2022; 106:1130-1138. [PMID: 34865266 DOI: 10.1111/jpn.13665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/11/2021] [Indexed: 11/29/2022]
Abstract
A pregnant hamster's exposure to changes in environmental factors, such as light, temperature and nutrition, may influence behavioural and physiological changes in offspring. In this study, dietary methyl donor supplementation was employed to examine the role of maternal diet on appetite, body weight, serum leptin levels and locomotor activity in male Syrian hamster offspring. Dams were fed a standard control (SC) or methyl donor-supplemented (MDSD) diet through pregnancy and lactation. At birth, offspring were cross-fostered to dams fed an SC or MDSD diet (SC-MDSD and MDSD-SC) or remained with their birth mothers (SC-SC and MDSD-MDSD). At weaning, offspring were fed a SC or MDSD diet until 60 days of age. Food intake, serum leptin levels and locomotor activity were measured from 30-60 days of age. Offspring fed a MDSD diet post-weaning (MDSD-MDSD and SC-MDSD) consumed more than double the amount of food daily compared with offspring fed a SC diet post-weaning (SC-SC, MDSD-SC). Interestingly, there were no observed differences in body weight among all four groups. Serum leptin levels at 60 days of age were depressed in offspring fed a MDSD diet post-weaning (MDSD-MDSD and SC-MDSD). There were no observed differences in wheel running activity between the SC-SC and MDSC-SC groups. Wheel running activity was at least twice the amount in offspring fed a MDSD diet post-weaning (SC-MDSD and MDSD-MDSD). Taken together, these results indicate that the timing of methyl donor supplementation appears to be an important factor during the development of offspring.
Collapse
Affiliation(s)
- Bülent Gündüz
- Faculty of Arts and Sciences, Department of Biology, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Darren K Okimoto
- University of Hawaii Sea Grant College Program, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
115
|
Demirci Ş, Gün C. Zinc Supplementation Improved Neuropeptide Y, Nesfatin-1, Leptin, C-reactive protein, and HOMA-IR of Diet-Induced Obese Rats. Biol Trace Elem Res 2022; 200:3996-4006. [PMID: 34708332 DOI: 10.1007/s12011-021-02987-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 11/24/2022]
Abstract
Obesity is a mild chronic inflammation that causes many metabolic diseases. It was aimed to investigate some parameters affective on the energy metabolism by adding zinc (Zn, ZnSO4) to drinking water of diet-induced obese rats. Five-week aged, male Sprague Dawley rats divided into as control group, consuming standard rat diet, and high-fat diet (HFD) group. After obesity induced by feeding HFD for 8 weeks, the obese rats were divided into Zn-supplemented obese group (HFD + obese + Zn; 150 mg Zn/L (for 6 weeks), 235 mg Zn/L (7th week), 250 mg Zn/L (8th week) in drinking water) and obese group (HFD + obese). Mean body weight, serum concentrations of C-reactive protein, neuropeptide-Y, leptin, insulin fasting blood glucose, and HOMA-IR were statistically decreased by given Zn in HFD + obese + Zn group compared to HFD + obese rats. It was observed that the total cholesterol, LDL, and HDL cholesterol levels of HFD + obese + Zn group became closer to the control group level, and Zn supplementation caused a statistically significant decrease in cholesterol profile than HFD + obese rats. Also, increased mean serum nesfatin-1 level, an effective protein for the formation of satiety, was analyzed in HFD + obese + Zn group when compared to HFD + obese ones. Serum triglyceride concentration tended to decrease with the effect of Zn in obese rats. In conclusion, it can be said that oral use of Zn could improve energy balance and prevent the occurrence of metabolic diseases related to obesity depending on the anti-inflammatory effect of Zn.
Collapse
Affiliation(s)
- Şule Demirci
- Physiology Department, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Campus, Burdur, Turkey.
| | | |
Collapse
|
116
|
Ito Y, Sun R, Yagimuma H, Taki K, Mizoguchi A, Kobayashi T, Sugiyama M, Onoue T, Tsunekawa T, Takagi H, Hagiwara D, Iwama S, Suga H, Konishi H, Kiyama H, Arima H, Banno R. Protein Tyrosine Phosphatase 1B Deficiency Improves Glucose Homeostasis in Type 1 Diabetes Treated With Leptin. Diabetes 2022; 71:1902-1914. [PMID: 35748319 PMCID: PMC9862406 DOI: 10.2337/db21-0953] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 06/19/2022] [Indexed: 02/05/2023]
Abstract
Leptin, a hormone secreted by adipocytes, exhibits therapeutic potential for the treatment of type 1 diabetes (T1D). Protein tyrosine phosphatase 1B (PTP1B) is a key enzyme that negatively regulates leptin receptor signaling. Here, the role of PTP1B in the treatment of T1D was investigated using PTP1B-deficient (knockout [KO]) mice and a PTP1B inhibitor. T1D wild-type (WT) mice induced by streptozotocin showed marked hyperglycemia compared with non-T1D WT mice. KO mice displayed significantly improved glucose metabolism equivalent to non-T1D WT mice, whereas peripheral or central administration of leptin partially improved glucose metabolism in T1D WT mice. Peripheral combination therapy of leptin and a PTP1B inhibitor in T1D WT mice improved glucose metabolism to the same level as non-T1D WT mice. Leptin was shown to act on the arcuate nucleus in the hypothalamus to suppress gluconeogenesis in liver and enhance glucose uptake in both brown adipose tissue and soleus muscle through the sympathetic nervous system. These effects were enhanced by PTP1B deficiency. Thus, treatment of T1D with leptin, PTP1B deficiency, or a PTP1B inhibitor was shown to enhance leptin activity in the hypothalamus to improve glucose metabolism. These findings suggest a potential alternative therapy for T1D.
Collapse
Affiliation(s)
- Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Runan Sun
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Yagimuma
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keigo Taki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Mizoguchi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Konishi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Research Center of Health, Physical Fitness and Sports, Nagoya University, Nagoya, Japan
- Corresponding author: Ryoichi Banno,
| |
Collapse
|
117
|
Inflammatory and metabolic markers in relation to outcome of in vitro fertilization in a cohort of predominantly overweight and obese women. Sci Rep 2022; 12:13331. [PMID: 35922472 PMCID: PMC9349206 DOI: 10.1038/s41598-022-17612-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 07/28/2022] [Indexed: 11/09/2022] Open
Abstract
For overweight and obese women undergoing in vitro fertilization (IVF) the pregnancy and live birth rates are compromised while the underlying mechanisms and predictors are unclear. The aim was to explore the association between adipose tissue-related inflammatory and metabolic markers and the pregnancy and live birth outcome of IVF in a cohort of predominantly overweight and obese women. Serum samples, fulfilling standardizing criteria, were identified from 195 women having participated in either the control (n = 131) or intervention (n = 64) group of a randomized controlled trial (RCT), seeking to evaluate the effect of a weight reduction intervention on IVF outcome in obese women. Serum high-sensitivity C-reactive protein (hsCRP) and the adipokines leptin and adipocyte fatty acid-binding protein (AFABP) were analyzed for the whole cohort (n = 195) in samples collected shortly before IVF [at randomization (control group), after intervention (intervention group)]. Information on age, anthropometry [BMI, waist circumference, waist-to-height ratio (WHtR)], pregnancy and live birth rates after IVF, as well as the spontaneous pregnancy rate, was extracted or calculated from collected data. The women of the original intervention group were also characterized at randomization regarding all variables. Eight women [n = 3 original control group (2.3%), n = 5 original intervention group (7.8%)] conceived spontaneously before starting IVF. BMI category proportions in the cohort undergoing IVF (n = 187) were 1.6/20.1/78.3% (normal weight/overweight/obese). The pregnancy and live birth rates after IVF for the cohort were 35.8% (n = 67) and 24.6% (n = 46), respectively. Multivariable logistic regression revealed that none of the variables (age, hsCRP, leptin, AFABP, BMI, waist circumference, WHtR) were predictive factors of pregnancy or live birth after IVF. Women of the original intervention group displayed reductions in hsCRP, leptin, and anthropometric variables after intervention while AFABP was unchanged. In this cohort of predominantly overweight and obese women undergoing IVF, neither low-grade inflammation, in terms of hsCRP, other circulating inflammatory and metabolic markers released from adipose tissue (leptin, AFABP), nor anthropometric measures of adiposity or adipose tissue distribution (BMI, waist, WHtR) were identified as predictive factors of pregnancy or live birth rate.Trial registration: ClinicalTrials.gov number, NCT01566929. Trial registration date 30-03-2012, retrospectively registered.
Collapse
|
118
|
Froelich M, Lemes SAF, Elias MPS, Oliveira APSS, Lisboa PC, Souza JRDE, Moura EG, Almeida FJS, Pereira MP, Latorraca MQ, Kawashita NH. Hyperphagia and hyperleptinemia induced by low-protein, high-carbohydrate diet is reversed at a later stage of development in rats. AN ACAD BRAS CIENC 2022; 94:e20210902. [PMID: 35857934 DOI: 10.1590/0001-3765202220210902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/04/2021] [Indexed: 11/22/2022] Open
Abstract
This study investigated whether increased food intake after 15 days of low-protein, high-carbohydrate (LPHC) and its normalization in the later period of development change the content of key proteins related to leptin or adiponectin signaling in the hypothalamus. Male rats were divided into five groups: Control groups received a control diet (17% protein, 63% carbohydrate) for 15 (C15) or 45 (C45) days; LPHC groups received an LPHC diet (6% protein, 74% carbohydrate) for 15 (LPHC15) or 45 (LPHC45) days; and Reverse group (R): received LPHC diet for 15 days followed by control diet for another 30 days. The LPHC15 group showed increased adiposity index, leptin level, and adiponectin level, as well as decreased the leptin receptor (ObRb) and pro-opiomelanocortin (POMC) content in the hypothalamus compared with the C15 group. LPHC diet for 45 days or diet reversion (R group) rescued these alterations, except the adiponectin level in LPHC45 rats, which was higher. In summary, LPHC diet reduced hypothalamic leptin action by diminishing ObRb and POMC levels, leading to hyperphagia and adiposity body. Medium-term administration of LPHC diet or reverting to control diet restored the levels of these proteins, thereby improving body lipid mass rearrangement in adulthood.
Collapse
Affiliation(s)
- Mendalli Froelich
- Universidade Federal de Mato Grosso, Departamento de Química, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| | - Suelem A F Lemes
- Universidade Federal de Mato Grosso, Departamento de Química, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| | - Maísa P S Elias
- Universidade Federal de Mato Grosso, Departamento de Química, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| | - Ana Paula S S Oliveira
- Universidade do Estado do Rio de Janeiro, Centro Biomédico, Instituto de Biologia Roberto Alcântara Gomes, Boulevard vinte e oito de setembro 87, 20551-031 Rio de Janeiro, RJ, Brazil.,Universidade Federal do Rio de Janeiro, NUMPEX-Bio, Campus Duque de Caxias, BR-040, Km 105, Santa Cruz da Serra, 25245-390 Duque de Caxias, RJ, Brazil
| | - Patricia C Lisboa
- Universidade do Estado do Rio de Janeiro, Centro Biomédico, Instituto de Biologia Roberto Alcântara Gomes, Boulevard vinte e oito de setembro 87, 20551-031 Rio de Janeiro, RJ, Brazil
| | - José Ricardo DE Souza
- Universidade Federal de Mato Grosso, Faculdade de Medicina Veterinária, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| | - Egberto G Moura
- Universidade do Estado do Rio de Janeiro, Centro Biomédico, Instituto de Biologia Roberto Alcântara Gomes, Boulevard vinte e oito de setembro 87, 20551-031 Rio de Janeiro, RJ, Brazil
| | - Fhelipe J S Almeida
- Universidade Federal de Mato Grosso, Departamento de Química, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| | - Mayara P Pereira
- Universidade Federal de Mato Grosso, Departamento de Química, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| | - Marcia Q Latorraca
- Universidade Federal de Mato Grosso, Faculdade de Nutrição, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| | - Nair H Kawashita
- Universidade Federal de Mato Grosso, Departamento de Química, Av. Fernando Correa da Costa, 2367, 78060-900 Cuiabá, MT, Brazil
| |
Collapse
|
119
|
Mohanraj J, D’Souza UJA, Fong SY, Karkada IR, Jaiprakash H. Association between Leptin (G2548A) and Leptin Receptor (Q223R) Polymorphisms with Plasma Leptin, BMI, Stress, Sleep and Eating Patterns among the Multiethnic Young Malaysian Adult Population from a Healthcare University. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148862. [PMID: 35886710 PMCID: PMC9316401 DOI: 10.3390/ijerph19148862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023]
Abstract
Relative leptin resistance in childhood to absolute leptin resistance in maturity suggests sleep, eating behaviour, and the psychological state as probable causes. The current body of research provides inconclusive evidence linking G2548A and Q223R to obesity. Furthermore, we could find very little data that have observed the association between the environment and gene polymorphism, especially in the multiethnic population that exists in Malaysia. This study searched for a possible link between sleeping habits, eating behaviour, and stress indicators with plasma leptin and its genetic variation in young adult Malaysian healthcare students. The study involved 185 first- and second-year medical and dental students from a healthcare university. Polymerase Chain Reaction−Restriction Fragment Length Polymorphism(PCR-RFLP) determined the genotype, Enzyme Linked Immunoabsorbant Assay (ELISA) tested the serum leptin, and a self-administered questionnaire evaluated sleep, eating behaviour, and psychological condition. Gender and ethnicity are linked to fasting plasma leptin levels (p < 0.001). Plasma leptin also affects stress, anxiety, and sadness. Leptin (LEP) and Leptin Receptor (LEPR) polymorphisms were not associated with BMI, plasma leptin, sleep, eating behaviour, or psychological state. Young adult Malaysian Indians were obese and overweight, while Chinese were underweight. These findings imply overweight and obese participants were in stage I of leptin resistance and lifestyle change or leptin therapy could prevent them from becoming cripplingly obese as they age.
Collapse
Affiliation(s)
- Jaiprakash Mohanraj
- Department of Biochemistry, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia; or
- Faculty of Medicine & Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Urban J. A. D’Souza
- Department of Physiology, Father Muller College of Allied Health Sciences, Father Muller Medical College, Mangalore 575002, India;
| | - Siat Yee Fong
- Faculty of Medicine & Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Correspondence:
| | - Ivan Rolland Karkada
- Department of Physiology, Faculty of Medicine, MAHSA Universiti, Jenjarom 42610, Malaysia;
| | - Heethal Jaiprakash
- Department of Pharmacology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia;
| |
Collapse
|
120
|
Guo Z, Yang H, Zhang JR, Zeng W, Hu X. Leptin receptor signaling sustains metabolic fitness of alveolar macrophages to attenuate pulmonary inflammation. SCIENCE ADVANCES 2022; 8:eabo3064. [PMID: 35857512 PMCID: PMC9286500 DOI: 10.1126/sciadv.abo3064] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Alveolar macrophages (AMs) are critical mediators of pulmonary inflammation. Given the unique lung tissue environment, whether there exist AM-specific mechanisms that control inflammation is not known. Here, we found that among various tissue-resident macrophage populations, AMs specifically expressed Lepr, encoding receptor for a key metabolic hormone leptin. AM-intrinsic Lepr signaling attenuated pulmonary inflammation in vivo, manifested as subdued acute lung injury yet compromised host defense against Streptococcus pneumoniae infection. Lepr signaling protected AMs from necroptosis and thus constrained neutrophil recruitment and tissue damage secondary to release of proinflammatory cytokine interleukin-1α. Mechanistically, Lepr signaling sustained activation of adenosine monophosphate-activated protein kinase in a Ca2+ influx-dependent manner and rewired cellular metabolism, thus preventing excessive lipid droplet formation and overloaded metabolic stress in a lipid-rich alveolar microenvironment. In conclusion, our results defined AM-expressed Lepr as a metabolic checkpoint of pulmonary inflammation and exemplified a macrophage tissue adaptation strategy for maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Ziyi Guo
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Haoqi Yang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Jing-Ren Zhang
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
121
|
Palma G, Sorice GP, Genchi VA, Giordano F, Caccioppoli C, D’Oria R, Marrano N, Biondi G, Giorgino F, Perrini S. Adipose Tissue Inflammation and Pulmonary Dysfunction in Obesity. Int J Mol Sci 2022; 23:ijms23137349. [PMID: 35806353 PMCID: PMC9267094 DOI: 10.3390/ijms23137349] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity is a chronic disease caused by an excess of adipose tissue that may impair health by altering the functionality of various organs, including the lungs. Excessive deposition of fat in the abdominal area can lead to abnormal positioning of the diaphragm and consequent reduction in lung volume, leading to a heightened demand for ventilation and increased exposure to respiratory diseases, such as chronic obstructive pulmonary disease, asthma, and obstructive sleep apnoea. In addition to mechanical ventilatory constraints, excess fat and ectopic deposition in visceral depots can lead to adipose tissue dysfunction, which promotes metabolic disorders. An altered adipokine-secretion profile from dysfunctional adipose tissue in morbid obesity fosters systemic, low-grade inflammation, impairing pulmonary immune response and promoting airway hyperresponsiveness. A potential target of these adipokines could be the NLRP3 inflammasome, a critical component of the innate immune system, the harmful pro-inflammatory effect of which affects both adipose and lung tissue in obesity. In this review, we will investigate the crosstalk between adipose tissue and the lung in obesity, highlighting the main inflammatory mediators and novel therapeutic targets in preventing pulmonary dysfunction.
Collapse
|
122
|
Vakil RM, Marine JE, Kolandaivelu A, Dickfeld T, Weiss RG, Tomaselli GF, Chrispin J, Wu KC. The Association of Clustered Ventricular Arrhythmia and Cycle Length With Scar Burden in Cardiomyopathy. JACC Clin Electrophysiol 2022; 8:957-966. [PMID: 35981800 PMCID: PMC10111293 DOI: 10.1016/j.jacep.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND Patients with ≥2 ventricular arrhythmia (VA) events within 3 months (clustered VA) have increased risk for mortality. OBJECTIVES The aim of this study was to examine the association of risk factors including scar characteristics on cardiovascular magnetic resonance imaging with clustered VA and VA cycle length in nonischemic cardiomyopathy (NICM) and ischemic cardiomyopathy (ICM). METHODS Data from 329 primary prevention implantable cardioverter-defibrillator recipients (mean age 57 years, 26% women) were analyzed from the Left Ventricular Structural Predictors of Sudden Cardiac Death study. RESULTS Twenty-one patients developed clustered VA (median time 2.7 years after implantable cardioverter-defibrillator placement). Men had the greatest risk for recurrent VA. Patients with NICM and scar had the highest incidence rate of clustered VA. In patients with NICM, each 1-g increase in core scar correlated with greater clustered VA risk (HR: 1.19; 95% CI: 1.07-1.32). Gray scar was similar among subgroups. Patients with NICM with clustered VA had the longest mean VA cycle length (297 ± 40 milliseconds). Higher core scar burden correlated with longer VA cycle length in patients with NICM (P = 0.002), and higher body mass index correlated with shorter VA cycle length in those with ICM (P = 0.02). Type of VA was similar between cardiomyopathy subgroups, and no scar pattern predominated. CONCLUSIONS Clustered VA was most common in patients with NICM and scar, with greatest risk among those with larger core scar. Core scar correlated with slower VA in patients with NICM, and higher body mass index correlated with faster VA in those with ICM. Type of VA was similar by cardiomyopathy etiology, and no dominant scar pattern was associated with clustered VA.
Collapse
|
123
|
Yue F, Du L, Wang R, Han B, Zhang X, Yao Z, Zhang W, Cai C, Zhang Z, Xu K. In vivo Protein Interference: Oral Administration of Recombinant Yeast-Mediated Partial Leptin Reduction for Obesity Control. Front Microbiol 2022; 13:923656. [PMID: 35774455 PMCID: PMC9237534 DOI: 10.3389/fmicb.2022.923656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/17/2022] [Indexed: 12/17/2022] Open
Abstract
Obesity-related diseases are always the major health problems that concern the whole world. Serial studies have reported that obesity development is closely related to the out-of-control leptin encoded by the obesity gene (ob). The latest report declaimed “Less Is More,” a model explaining that partial leptin reduction triggers leptin sensitization and contributes to obesity control. Here, we came up with a novel concept, in vivo protein interference (iPRTi), an interesting protein knock-down strategy for in vivo partial leptin reduction. First, the specific immune response against leptin induced by the oral administration of ob recombinant yeast was confirmed. Subsequentally, leptin resistance was observed in diet-induced obese mice, and oral administration with ob recombinant yeast declined the circulating leptin and reduced significantly the body weight gain. To further investigate whether the iPRTi strategy is capable of obesity management, the diet-induced obese mice were administrated with ob recombinant yeast. All the indexes examined including the circulating leptin, triglyceride, and total cholesterol, as well as food intake and weight gain, demonstrated a positive effect of the iPRTi strategy on obesity control. In short, this study provides a novel strategy for the potential application of recombinant yeast for the therapy of obese individuals with leptin resistance.
Collapse
|
124
|
Levitsky DA, Barre L, Michael JJ, Zhong Y, He Y, Mizia A, Kaila S. The Rise and Fall of Physiological Theories of the Control of Human Eating Behavior. Front Nutr 2022; 9:826334. [PMID: 35662925 PMCID: PMC9159371 DOI: 10.3389/fnut.2022.826334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/17/2022] [Indexed: 12/14/2022] Open
Abstract
Kuhns was the first to suggest that theories in science do not develop in small increments but rather in major leaps to paradigms that examine the same question through very different perspectives. Theories on the mechanism responsible for control of human food intake fall into Kuhn’s description. This article describes how the two major theories of the control of food intake in humans, the Glucostatic Theory, and the Lipostatic Theory, showed initial promise as explanations, but later deteriorated with the slow accumulation experimental data. The locus of theories considered eating behavior as a part of physiological system that regulates the storage of energy on the body. We challenge this fundamental belief with data which suggests that we must be ready to accept a major change in the way we think about eating behavior if we are ever to decrease the prevalence of obesity.
Collapse
Affiliation(s)
- David A Levitsky
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States.,Department of Psychology, Cornell University, Ithaca, NY, United States
| | - Laura Barre
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | | | - Yingyi Zhong
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Yitong He
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Alyse Mizia
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Sahib Kaila
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
125
|
Wooten JS, Breden M, Hoeg T, Smith BK. Effects of weight-loss on adipokines, total and regional body composition and markers of metabolic syndrome in women who are overweight and obese. ENDOCRINE AND METABOLIC SCIENCE 2022. [DOI: 10.1016/j.endmts.2022.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
|
126
|
de Souza GO, Wasinski F, Donato J. Characterization of the metabolic differences between male and female C57BL/6 mice. Life Sci 2022; 301:120636. [PMID: 35568227 DOI: 10.1016/j.lfs.2022.120636] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 01/22/2023]
Abstract
AIMS The present study aims to compare the responses between male and female C57BL/6 mice to multiple metabolic challenges to understand the importance of sex in the control of energy homeostasis. MAIN METHODS Male and female C57BL/6 mice were subjected to nutritional and hormonal challenges, such as food restriction and refeeding, diet-induced obesity, feeding response to ghrelin and leptin, ghrelin-induced growth hormone secretion, and central responsiveness to ghrelin and leptin. The hypothalamic expression of transcripts that control energy homeostasis was also evaluated. KEY FINDINGS Male mice lost more weight and lean body mass in response to food restriction, compared to females. During refeeding, males accumulated more body fat and exhibited lower energy expenditure and glycemia, as compared to females. Additionally, female mice exhibited a higher protection against diet-induced obesity and related metabolic imbalances in comparison to males. Low dose ghrelin injection elicited higher food intake and growth hormone secretion in male mice, whereas the acute anorexigenic effect of leptin was more robust in females. However, the sex differences in the feeding responses to ghrelin and leptin were not explained by variations in the central responsiveness to these hormones nor by differences in the fiber density from arcuate nucleus neurons. Female, but not male, mice exhibited compensatory increases in hypothalamic Pomc mRNA levels in response to diet-induced obesity. SIGNIFICANCE Our findings revealed several sexually differentiated responses to metabolic challenges in C57BL/6 mice, highlighting the importance of taking into account sex differences in metabolic studies.
Collapse
Affiliation(s)
- Gabriel O de Souza
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil..
| |
Collapse
|
127
|
Picarda E, Galbo PM, Zong H, Rajan MR, Wallenius V, Zheng D, Börgeson E, Singh R, Pessin J, Zang X. The immune checkpoint B7-H3 (CD276) regulates adipocyte progenitor metabolism and obesity development. SCIENCE ADVANCES 2022; 8:eabm7012. [PMID: 35476450 PMCID: PMC9045715 DOI: 10.1126/sciadv.abm7012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/11/2022] [Indexed: 05/09/2023]
Abstract
The immune checkpoint B7-H3 (CD276) is a member of the B7 family that has been studied in the tumor microenvironment and immunotherapy, but its potential role in metabolism remains largely unknown. Here, we show that B7-H3 is highly expressed in mouse and human adipose tissue at steady state, with the highest levels in adipocyte progenitor cells. B7-H3 is rapidly down-regulated upon the initiation of adipocyte differentiation. Combined RNA sequencing and metabolic studies reveal that B7-H3 stimulates glycolytic and mitochondrial activity of adipocyte progenitors. Loss of B7-H3 in progenitors results in impaired oxidative metabolism program and increased lipid accumulation in derived adipocytes. Consistent with these observations, mice knocked out for B7-H3 develop spontaneous obesity, metabolic dysfunction, and adipose tissue inflammation. Our results reveal an unexpected metabolic role for B7-H3 in adipose tissue and open potential new avenues for the treatment of metabolic diseases by targeting the B7-H3 pathway.
Collapse
Affiliation(s)
- Elodie Picarda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Phillip M. Galbo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Haihong Zong
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Meenu Rohini Rajan
- Institute of Medicine, Department of Molecular and Clinical Medicine, The Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emma Börgeson
- Institute of Medicine, Department of Molecular and Clinical Medicine, The Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Vaestra Goetaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rajat Singh
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeffrey Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
128
|
Abdel-Aal NM, Mostafa MSEM, Saweres JW, Ghait RS. Cavitation and radiofrequency versus cryolipolysis on leptin regulation in central obese subjects: A randomized controlled study. Lasers Surg Med 2022; 54:955-963. [PMID: 35481595 DOI: 10.1002/lsm.23555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 03/18/2022] [Accepted: 04/11/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND AND OBJECTIVE To investigate the efficacy of adding ultrasound cavitation and radiofrequency versus cryolipolysis to weight reduction program on leptin, insulin, waist circumference, skinfold, body weight in central obese subjects. MATERIAL AND METHODS Sixty centrally obese participants were randomly allocated into three equal groups. Subjects in the study group (I) received cavitation and radiofrequency plus dietary regimen, subjects in the second study group (II) received cryolipolysis in conjunction with the same diet program, and subjects in the control group (III) received the same dietary regimen only. Leptin, insulin level, waist circumference, skinfold, body weight, and body mass index were measured shortly before intervention techniques and 3 months afterward. RESULTS There were no statistically significant differences between cavitation plus radiofrequency and cryolipolysis on leptin and insulin levels after 3 months of intervention. However, statistically significant differences were found in waist circumference, skinfold, weight reduction, and body mass index in favor of the cavitation group (p < 0.05). In addition, both cavitation-radiofrequency and cryolipolysis were statistically significantly different than the diet alone in favor of the study groups (p < 0.05) in all the outcome measures. Furthermore, there were statistically significant differences in all outcome measures (p < 0.05) when comparing the baseline and postintervention results in each group except for leptin level in the diet group (p = 0.14). CONCLUSION Subjects who underwent cavitation plus radiofrequency had better improvement on waist circumference, skinfold, and body mass index than subjects who received cryolipolysis. However, no differences were found between cavitation plus radiofrequency and cryolipolysis on leptin and insulin levels.
Collapse
Affiliation(s)
- Nabil M Abdel-Aal
- Basic Science Department, Faculty of Physical Therapy, Cairo University, Dokki, Giza, Egypt
| | - Mohamed S E M Mostafa
- Basic Science Department, Faculty of Physical Therapy, Cairo University, Dokki, Giza, Egypt.,Basic Science Department, Faculty of Physical Therapy, Heliopolis University, Cairo, Egypt
| | - Joseph W Saweres
- Basic Science Department, Faculty of Physical Therapy, Cairo University, Dokki, Giza, Egypt
| | - Ramy S Ghait
- Department of Internal Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
129
|
Socol CT, Chira A, Martinez-Sanchez MA, Nuñez-Sanchez MA, Maerescu CM, Mierlita D, Rusu AV, Ruiz-Alcaraz AJ, Trif M, Ramos-Molina B. Leptin Signaling in Obesity and Colorectal Cancer. Int J Mol Sci 2022; 23:4713. [PMID: 35563103 PMCID: PMC9102849 DOI: 10.3390/ijms23094713] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 12/05/2022] Open
Abstract
Obesity and colorectal cancer (CRC) are among the leading diseases causing deaths in the world, showing a complex multifactorial pathology. Obesity is considered a risk factor in CRC development through inflammation, metabolic, and signaling processes. Leptin is one of the most important adipokines related to obesity and an important proinflammatory marker, mainly expressed in adipose tissue, with many genetic variation profiles, many related influencing factors, and various functions that have been ascribed but not yet fully understood and elucidated, the most important ones being related to energy metabolism, as well as endocrine and immune systems. Aberrant signaling and genetic variations of leptin are correlated with obesity and CRC, with the genetic causality showing both inherited and acquired events, in addition to lifestyle and environmental risk factors; these might also be related to specific pathogenic pathways at different time points. Moreover, mutation gain is a crucial factor enabling the genetic process of CRC. Currently, the inconsistent and insufficient data related to leptin's relationship with obesity and CRC indicate the necessity of further related studies. This review summarizes the current knowledge on leptin genetics and its potential relationship with the main pathogenic pathways of obesity and CRC, in an attempt to understand the molecular mechanisms of these associations, in the context of inconsistent and contradictory data. The understanding of these mechanisms linking obesity and CRC could help to develop novel therapeutic targets and prevention strategies, resulting in a better prognosis and management of these diseases.
Collapse
Affiliation(s)
| | - Alexandra Chira
- 2nd Medical Clinic, Department of Internal Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Maria Antonia Martinez-Sanchez
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (M.A.M.-S.); (M.A.N.-S.)
| | - Maria Angeles Nuñez-Sanchez
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (M.A.M.-S.); (M.A.N.-S.)
| | | | - Daniel Mierlita
- Department of Nutrition, University of Oradea, 410048 Oradea, Romania;
| | - Alexandru Vasile Rusu
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania;
- Faculty of Animal Science and Biotechnology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania
| | - Antonio Jose Ruiz-Alcaraz
- Department of Biochemistry and Molecular B and Immunology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain;
| | - Monica Trif
- Department of Food Research, Centiv GmbH, 28857 Syke, Germany;
| | - Bruno Ramos-Molina
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (M.A.M.-S.); (M.A.N.-S.)
| |
Collapse
|
130
|
Gao D, Jiao J, Wang Z, Huang X, Ni X, Fang S, Zhou Q, Zhu X, Sun L, Yang Z, Yuan H. The roles of cell-cell and organ-organ crosstalk in the type 2 diabetes mellitus associated inflammatory microenvironment. Cytokine Growth Factor Rev 2022; 66:15-25. [PMID: 35459618 DOI: 10.1016/j.cytogfr.2022.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a classic metaflammatory disease, and the inflammatory states of the pancreatic islet and insulin target organs have been well confirmed. However, abundant evidence demonstrates that there are countless connections between these organs in the presence of a low degree of inflammation. In this review, we focus on cell-cell crosstalk among local cells in the islet and organ-organ crosstalk among insulin-related organs. In contrast to that in acute inflammation, macrophages are the dominant immune cells causing inflammation in the islets and insulin target organs in T2DM. In the inflammatory microenvironment (IME) of the islet, cell-cell crosstalk involving local macrophage polarization and proinflammatory cytokine production impair insulin secretion by β-cells. Furthermore, organ-organ crosstalk, including the gut-brain-pancreas axis and interactions among insulin-related organs during inflammation, reduces insulin sensitivity and induces endocrine dysfunction. Therefore, this crosstalk ultimately results in a cascade leading to β-cell dysfunction. These findings could have broad implications for therapies aimed at treating T2DM.
Collapse
Affiliation(s)
- Danni Gao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China; Peking University Fifth School of Clinical Medicine, Beijing 100730, PR China
| | - Juan Jiao
- Department of Clinical Laboratory, the Seventh Medical Centre of Chinese PLA General Hospital, Beijing 100700, PR China
| | - Zhaoping Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Xiaolin Ni
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Sihang Fang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Qi Zhou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Xiaoquan Zhu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Liang Sun
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China
| | - Huiping Yuan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, PR China; Peking University Fifth School of Clinical Medicine, Beijing 100730, PR China.
| |
Collapse
|
131
|
Westmark CJ, Filon MJ, Maina P, Steinberg LI, Ikonomidou C, Westmark PR. Effects of Soy-Based Infant Formula on Weight Gain and Neurodevelopment in an Autism Mouse Model. Cells 2022; 11:1350. [PMID: 35456030 PMCID: PMC9025435 DOI: 10.3390/cells11081350] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
Mice fed soy-based diets exhibit increased weight gain compared to mice fed casein-based diets, and the effects are more pronounced in a model of fragile X syndrome (FXS; Fmr1KO). FXS is a neurodevelopmental disability characterized by intellectual impairment, seizures, autistic behavior, anxiety, and obesity. Here, we analyzed body weight as a function of mouse age, diet, and genotype to determine the effect of diet (soy, casein, and grain-based) on weight gain. We also assessed plasma protein biomarker expression and behavior in response to diet. Juvenile Fmr1KO mice fed a soy protein-based rodent chow throughout gestation and postnatal development exhibit increased weight gain compared to mice fed a casein-based purified ingredient diet or grain-based, low phytoestrogen chow. Adolescent and adult Fmr1KO mice fed a soy-based infant formula diet exhibited increased weight gain compared to reference diets. Increased body mass was due to increased lean mass. Wild-type male mice fed soy-based infant formula exhibited increased learning in a passive avoidance paradigm, and Fmr1KO male mice had a deficit in nest building. Thus, at the systems level, consumption of soy-based diets increases weight gain and affects behavior. At the molecular level, a soy-based infant formula diet was associated with altered expression of numerous plasma proteins, including the adipose hormone leptin and the β-amyloid degrading enzyme neprilysin. In conclusion, single-source, soy-based diets may contribute to the development of obesity and the exacerbation of neurological phenotypes in developmental disabilities, such as FXS.
Collapse
Affiliation(s)
- Cara J. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Molecular Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, USA
| | - Mikolaj J. Filon
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Undergraduate Research Program, University of Wisconsin, Madison, WI 53706, USA
| | - Patricia Maina
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Molecular Environmental Toxicology Summer Research Opportunities Program, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren I. Steinberg
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
- Undergraduate Research Program, University of Wisconsin, Madison, WI 53706, USA
| | - Chrysanthy Ikonomidou
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
| | - Pamela R. Westmark
- Department of Neurology, University of Wisconsin, Madison, WI 53706, USA; (M.J.F.); (P.M.); (L.I.S.); (C.I.); (P.R.W.)
| |
Collapse
|
132
|
Araújo MC, Soczek SHS, Pontes JP, Marques LAC, Santos GS, Simão G, Bueno LR, Maria-Ferreira D, Muscará MN, Fernandes ES. An Overview of the TRP-Oxidative Stress Axis in Metabolic Syndrome: Insights for Novel Therapeutic Approaches. Cells 2022; 11:cells11081292. [PMID: 35455971 PMCID: PMC9030853 DOI: 10.3390/cells11081292] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/19/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome (MS) is a complex pathology characterized by visceral adiposity, insulin resistance, arterial hypertension, and dyslipidaemia. It has become a global epidemic associated with increased consumption of high-calorie, low-fibre food and sedentary habits. Some of its underlying mechanisms have been identified, with hypoadiponectinemia, inflammation and oxidative stress as important factors for MS establishment and progression. Alterations in adipokine levels may favour glucotoxicity and lipotoxicity which, in turn, contribute to inflammation and cellular stress responses within the adipose, pancreatic and liver tissues, in addition to hepatic steatosis. The multiple mechanisms of MS make its clinical management difficult, involving both non-pharmacological and pharmacological interventions. Transient receptor potential (TRP) channels are non-selective calcium channels involved in a plethora of physiological events, including energy balance, inflammation and oxidative stress. Evidence from animal models of disease has contributed to identify their specific contributions to MS and may help to tailor clinical trials for the disease. In this context, the oxidative stress sensors TRPV1, TRPA1 and TRPC5, play major roles in regulating inflammatory responses, thermogenesis and energy expenditure. Here, the interplay between these TRP channels and oxidative stress in MS is discussed in the light of novel therapies to treat this syndrome.
Collapse
Affiliation(s)
- Mizael C. Araújo
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Suzany H. S. Soczek
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Jaqueline P. Pontes
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 565085-080, MA, Brazil;
| | - Leonardo A. C. Marques
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Gabriela S. Santos
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Laryssa R. Bueno
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Marcelo N. Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Elizabeth S. Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
- Correspondence:
| |
Collapse
|
133
|
Remodeling and Fibrosis of the Cardiac Muscle in the Course of Obesity-Pathogenesis and Involvement of the Extracellular Matrix. Int J Mol Sci 2022; 23:ijms23084195. [PMID: 35457013 PMCID: PMC9032681 DOI: 10.3390/ijms23084195] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/09/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity is a growing epidemiological problem, as two-thirds of the adult population are carrying excess weight. It is a risk factor for the development of cardiovascular diseases (hypertension, ischemic heart disease, myocardial infarct, and atrial fibrillation). It has also been shown that chronic obesity in people may be a cause for the development of heart failure with preserved ejection fraction (HFpEF), whose components include cellular hypertrophy, left ventricular diastolic dysfunction, and increased extracellular collagen deposition. Several animal models with induced obesity, via the administration of a high-fat diet, also developed increased heart fibrosis as a result of extracellular collagen accumulation. Excessive collagen deposition in the extracellular matrix (ECM) in the course of obesity may increase the stiffness of the myocardium and thereby deteriorate the heart diastolic function and facilitate the occurrence of HFpEF. In this review, we include a rationale for that process, including a discussion about possible putative factors (such as increased renin–angiotensin–aldosterone activity, sympathetic overdrive, hemodynamic alterations, hypoadiponectinemia, hyperleptinemia, and concomitant heart diseases). To address the topic clearly, we include a description of the fundamentals of ECM turnover, as well as a summary of studies assessing collagen deposition in obese individuals.
Collapse
|
134
|
Kim D, Howard AG, Blanco E, Burrows R, Correa-Burrows P, Memili A, Albala C, Santos JL, Angel B, Lozoff B, Justice AE, Gordon-Larsen P, Gahagan S, North KE. Dynamic relationships between body fat and circulating adipokine levels from adolescence to young adulthood: The Santiago Longitudinal Study. Nutr Metab Cardiovasc Dis 2022; 32:1055-1063. [PMID: 35181188 PMCID: PMC9107379 DOI: 10.1016/j.numecd.2022.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/07/2021] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Adipose tissue secretes adipokines such as adiponectin and leptin, playing important roles in energy metabolism. The longitudinal associations between such adipokines and body fat accumulation have not been established, especially during adolescence and young adulthood and in diverse populations. The study aims to assess the longitudinal association between body fat measured with dual X-ray absorptiometry and plasma adipokines from adolescence to young adulthood. METHODS AND RESULTS Among Hispanic/Latino participants (N = 537) aged 16.8 (SD: 0.3) years of the Santiago Longitudinal Study, we implemented structural equation modeling to estimate the sex-specific associations between adiposity (body fat percent (BF%) and proportion of trunk fat (PTF)) and adipokines (adiponectin and leptin levels) during adolescence (16 y) and these values after 6 years of follow-up (22 y). In addition, we further investigated whether the associations differed by baseline insulin resistance (IR) status. We found evidence for associations between 16 y BF% and 22 y leptin levels (β (SE): 0.58 (0.06) for females; 0.53 (0.05) for males), between 16 y PTF and 22 y adiponectin levels (β (SE): -0.31 (0.06) for females; -0.18 (0.06) for males) and between 16 y adiponectin levels and 22 y BF% (β (SE): 0.12 (0.04) for both females and males). CONCLUSION We observed dynamic relationships between adiposity and adipokines levels from late adolescence to young adulthood in a Hispanic/Latino population further demonstrating the importance of this period of the life course in the development of obesity.
Collapse
Affiliation(s)
- Daeeun Kim
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Annie Green Howard
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Estela Blanco
- Division of Academic General Pediatrics, Child Development and Community Health at the Center for Community Health, University of California at San Diego, San Diego, CA, USA; Department of Public Health, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Raquel Burrows
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | | | - Aylin Memili
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cecilia Albala
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - José L Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara Angel
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Betsy Lozoff
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Anne E Justice
- Department of Population Health Sciences, Geisinger, Danville, PA, USA
| | - Penny Gordon-Larsen
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, USA
| | - Sheila Gahagan
- Division of Academic General Pediatrics, Child Development and Community Health at the Center for Community Health, University of California at San Diego, San Diego, CA, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
135
|
Ramadan Intermittent Fasting Is Associated with Changes in Circulating Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) in Metabolically Healthy Obese Subjects. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58040503. [PMID: 35454343 PMCID: PMC9029631 DOI: 10.3390/medicina58040503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 11/16/2022]
Abstract
Background and Objectives: Dietary modification is the principal approach to the management of hyperlipidemia in adults. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key regulator of plasma cholesterol and a target for novel lipid-lowering pharmacotherapies. This study aimed to explore how circulating levels of PCSK9 changed during Ramadan intermittent fasting in metabolically healthy obese subjects. Materials and Methods: This cross-sectional study used convenience sampling to recruit 55 overweight and obese participants (22 females and 33 males) who observed Ramadan fasting. Body weight and composition, glucoregulatory factors, serum PCSK9 concentration, dietary intake, and physical activity were assessed 1 week before and at the end of Ramadan fasting. Results: The median (interquartile range) age was 35 (22) years, and body mass index was 30.2 (5.4). We found significant (p < 0.05) increases in serum levels of PCSK9, serum insulin, insulin resistance, and leptin at the end of Ramadan compared with pre-fasting levels. Significant (p < 0.05) reductions in body weight, waist circumference, systolic and diastolic blood pressure, total cholesterol, triglycerides, high-density lipoprotein cholesterol, and adiponectin were also observed at the end of Ramadan. Conclusions: Observing Ramadan fasting was associated with increased PCSK9 levels in metabolically healthy obese subjects. The complex relationships between PCSK9 and insulin resistance and dysregulation of adipokine secretion in relation to dietary and lifestyle modifications during Ramadan warrant further research.
Collapse
|
136
|
Liu M, Cardilla A, Ngeow J, Gong X, Xia Y. Studying Kidney Diseases Using Organoid Models. Front Cell Dev Biol 2022; 10:845401. [PMID: 35309912 PMCID: PMC8927804 DOI: 10.3389/fcell.2022.845401] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
The prevalence of chronic kidney disease (CKD) is rapidly increasing over the last few decades, owing to the global increase in diabetes, and cardiovascular diseases. Dialysis greatly compromises the life quality of patients, while demand for transplantable kidney cannot be met, underscoring the need to develop novel therapeutic approaches to stop or reverse CKD progression. Our understanding of kidney disease is primarily derived from studies using animal models and cell culture. While cross-species differences made it challenging to fully translate findings from animal models into clinical practice, primary patient cells quickly lose the original phenotypes during in vitro culture. Over the last decade, remarkable achievements have been made for generating 3-dimensional (3D) miniature organs (organoids) by exposing stem cells to culture conditions that mimic the signaling cues required for the development of a particular organ or tissue. 3D kidney organoids have been successfully generated from different types of source cells, including human pluripotent stem cells (hPSCs), adult/fetal renal tissues, and kidney cancer biopsy. Alongside gene editing tools, hPSC-derived kidney organoids are being harnessed to model genetic kidney diseases. In comparison, adult kidney-derived tubuloids and kidney cancer-derived tumoroids are still in their infancy. Herein, we first summarize the currently available kidney organoid models. Next, we discuss recent advances in kidney disease modelling using organoid models. Finally, we consider the major challenges that have hindered the application of kidney organoids in disease modelling and drug evaluation and propose prospective solutions.
Collapse
Affiliation(s)
- Meng Liu
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Angelysia Cardilla
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Joanne Ngeow
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- Cancer Genetics Service, National Cancer Centre Singapore, Singapore, Singapore
| | - Ximing Gong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- *Correspondence: Ximing Gong, ; Yun Xia,
| | - Yun Xia
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
- *Correspondence: Ximing Gong, ; Yun Xia,
| |
Collapse
|
137
|
Sudhakaran G, Guru A, Hari Deva Muthu B, Murugan R, Arshad A, Arockiaraj J. Evidence-based hormonal, mutational, and endocrine-disrupting chemical-induced zebrafish as an alternative model to study PCOS condition similar to mammalian PCOS model. Life Sci 2022; 291:120276. [PMID: 34990650 DOI: 10.1016/j.lfs.2021.120276] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022]
Abstract
Polycystic ovarian syndrome (PCOS) causes swollen ovaries in women at reproductive age due to hormonal disorder with small cysts on the outer edges. The cause of the disorder is still yet to be found. Multiple factors have increased PCOS prevalence, hyperandrogenism, oxidative stress, inflammation, and insulin resistance. Various animal PCOS models have been developed to imitate the pathophysiology of PCOS in humans. Zebrafish is one of the most versatile animal experimental models because of the transparency of the embryos, small size, and rapid growth. The zebrafish similarity to higher vertebrates made it a useful non-mammalian model for PCOS drug testing and screening. This review provides an insight into the usage of zebrafish, a non-mammalian model for PCOS, as an opportunity for evaluating future initiatives in such a research domain.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Ajay Guru
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - B Hari Deva Muthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Raghul Murugan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India
| | - Aziz Arshad
- International Institute of Aquaculture and Aquatic Sciences (I-AQUAS), Universiti Putra Malaysia, 71050 Port Dickson, Negeri Sembilan, Malaysia
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai 603 203, Tamil Nadu, India; Foundation for Aquaculture Innovations and Technology Transfer (FAITT), Thoraipakkam, Chennai 600 097, Tamil Nadu, India.
| |
Collapse
|
138
|
Maffei M, Giordano A. Leptin, the brain and energy homeostasis: From an apparently simple to a highly complex neuronal system. Rev Endocr Metab Disord 2022; 23:87-101. [PMID: 33822303 DOI: 10.1007/s11154-021-09636-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
Leptin, produced and secreted by white adipose tissue in tight relationship with adipose mass, informs the brain about the status of the energy stores serving as the main peripheral signal for energy balance regulation through interaction with a multitude of highly interconnected neuronal populations. Most obese patients display resistance to the anorectic effect of the hormone. The present review unravels the multiple levels of complexity that trigger hypothalamic response to leptin with the objective of highlighting those critical hubs that, mainly in the hypothalamic arcuate nucleus, may undergo obesity-induced alterations and create an obstacle to leptin action. Several mechanisms underlying leptin resistance have been proposed, possibly representing useful targets to empower leptin effects. Among these, a special focus is herein dedicated to detail how leptin gains access into the brain and how neuronal plasticity may interfere with leptin function.
Collapse
Affiliation(s)
- Margherita Maffei
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124, Pisa, Italy.
- Obesity and Lipodystrophy Center, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020, Ancona, Italy.
| |
Collapse
|
139
|
Abstract
Leptin is a hormone primarily produced by the adipose tissue in proportion to the size of fat stores, with a primary function in the control of lipid reserves. Besides adipose tissue, leptin is also produced by other tissues, such as the stomach, placenta, and mammary gland. Altogether, leptin exerts a broad spectrum of short, medium, and long-term regulatory actions at the central and peripheral levels, including metabolic programming effects that condition the proper development and function of the adipose organ, which are relevant for its main role in energy homeostasis. Comprehending how leptin regulates adipose tissue may provide important clues to understand the pathophysiology of obesity and related diseases, such as type 2 diabetes, as well as its prevention and treatment. This review focuses on the physiological and long-lasting regulatory effects of leptin on adipose tissue, the mechanisms and pathways involved, its main outcomes on whole-body physiological homeostasis, and its consequences on chronic diseases.
Collapse
Affiliation(s)
- Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Mariona Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Catalina Amadora Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Ana María Rodríguez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics, Biomarkers and Risk Evaluation), University of the Balearic Islands. CIBER de Fisiopatología de La Obesidad Y Nutrición (CIBEROBN). Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| |
Collapse
|
140
|
Hu Z, Ai N, Chen W, Wong QWL, Ge W. Leptin and Its Signaling Are Not Involved in Zebrafish Puberty Onset. Biol Reprod 2022; 106:928-942. [DOI: 10.1093/biolre/ioac005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/29/2021] [Accepted: 01/13/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Leptin is a peptide hormone secreted from the adipose tissues and its signaling plays a central role in metabolic regulation of growth, especially on fat mass. In addition, leptin is also involved in regulating reproduction in mammals. In teleosts, there are two leptin ligands (lepa and lepb) and one cognate leptin receptor (lepr); however, their functions are still elusive. In this study, we created null-function mutants for lepa, lepb and lepr in zebrafish using CRISPR/Cas9 method and analyzed their phenotypes with emphasis on puberty onset, one major function widely reported for leptin in mammals. We demonstrated that the loss of leptin ligands or their receptor resulted in no obesity from prepubertal stage to adulthood. We then focused on leptin involvement in controlling puberty onset. We first confirmed the somatic threshold for puberty onset in females and proposed a criterion and somatic threshold for male puberty onset. We examined gonadal development and sex maturation in different genotypic combinations including single mutants (lepa−/−, lepb−/− and lepr−/−), double mutants (lepa−/−;lepb−/−) and triple mutants (lepa−/−;lepb−/−;lepr−/−). Our results showed that once the fish reached the thresholds, the siblings of all genotypes displayed comparable gonadal development in both sexes without obvious signs of changed puberty onset. In conclusion, this comprehensive genetic study on the lep-lepr system demonstrated that in contrast to its counterpart in mammals, leptin system plays little role in controlling growth and reproduction especially puberty onset in zebrafish.
Collapse
|
141
|
Cowman W, Scroggins SM, Hamilton WS, Karras AE, Bowdler NC, Devor EJ, Santillan MK, Santillan DA. Association between plasma leptin and cesarean section after induction of labor: a case control study. BMC Pregnancy Childbirth 2022; 22:29. [PMID: 35031012 PMCID: PMC8759283 DOI: 10.1186/s12884-021-04372-6] [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: 05/26/2021] [Accepted: 12/30/2021] [Indexed: 11/17/2022] Open
Abstract
Background Obesity in pregnancy is common, with more than 50% of pregnant women being overweight or obese. Obesity has been identified as an independent predictor of dysfunctional labor and is associated with increased risk of failed induction of labor resulting in cesarean section. Leptin, an adipokine, is secreted from adipose tissue under the control of the obesity gene. Concentrations of leptin increase with increasing percent body fat due to elevated leptin production from the adipose tissue of obese individuals. Interestingly, the placenta is also a major source of leptin production during pregnancy. Leptin has regulatory effects on neuronal tissue, vascular smooth muscle, and nonvascular smooth muscle systems. It has also been demonstrated that leptin has an inhibitory effect on myometrial contractility with both intensity and frequency of contractions decreased. These findings suggest that leptin may play an important role in dysfunctional labor and be associated with the outcome of induction of labor at term. Our aim is to determine whether maternal plasma leptin concentration is indicative of the outcome of induction of labor at term. We hypothesize that elevated maternal plasma leptin levels are associated with a failed term induction of labor resulting in a cesarean delivery. Methods In this case-control study, leptin was measured in 3rd trimester plasma samples. To analyze labor outcomes, 174 women were selected based on having undergone an induction of labor (IOL), (115 women with successful IOL and 59 women with a failed IOL). Plasma samples and clinical information were obtained from the UI Maternal Fetal Tissue Bank (IRB# 200910784). Maternal plasma leptin and total protein concentrations were measured using commercially available assays. Bivariate analyses and logistic regression models were constructed using regression identified clinically significant confounding variables. All variables were tested at significance level of 0.05. Results Women with failed IOL had higher maternal plasma leptin values (0.5 vs 0.3 pg, P = 0.01). These women were more likely to have obesity (mean BMI 32 vs 27 kg/m2, P = 0.0002) as well as require multiple induction methods (93% vs 73%, p = 0.008). Logistic regression showed Bishop score (OR 1.5, p < 0.001), BMI (OR 0.92, P < 0.001), preeclampsia (OR 0.12, P = 0.010), use of multiple methods of induction (OR 0.22, P = 0.008) and leptin (OR 0.42, P = 0.017) were significantly associated with IOL outcome. Specifically, after controlling for BMI, Bishop Score, and preeclampsia, leptin was still predictive of a failed IOL with an odds ratio of 0.47 (P = 0.046). Finally, using leptin as a predictor for fetal outcomes, leptin was also associated with of fetal intolerance of labor, with an odds ratio of 2.3 (P = 0.027). This association remained but failed to meet statistical significance when controlling for successful (IOL) (OR 1.5, P = 0.50). Conclusions Maternal plasma leptin may be a useful tool for determining which women are likely to have a failed induction of labor and for counseling women about undertaking an induction of labor versus proceeding with cesarean delivery.
Collapse
Affiliation(s)
- Whitney Cowman
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA.,Present Address: Department of Obstetrics & Gynecology, Iowa Methodist Medical Center, 1200 Pleasant Street, Des Moines, IA, 50309, USA
| | - Sabrina M Scroggins
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Wendy S Hamilton
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Alexandra E Karras
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Noelle C Bowdler
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Eric J Devor
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Mark K Santillan
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA
| | - Donna A Santillan
- Department of Obstetrics & Gynecology, University of Iowa Hospitals & Clinics, 200 Hawkins Drive, 463 MRF, Iowa City, IA, 52242, USA.
| |
Collapse
|
142
|
Lui PPY, Yung PSH. Inflammatory mechanisms linking obesity and tendinopathy. J Orthop Translat 2022; 31:80-90. [PMID: 34976728 PMCID: PMC8666605 DOI: 10.1016/j.jot.2021.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic tendinopathy is a debilitating tendon disorder with disappointing treatment outcomes. This review focuses on the potential roles of chronic low-grade inflammation in promoting tendinopathy in obesity. A systematic literature search was performed to identify all clinical studies supporting the actions of obesity-associated inflammatory mediators in the development of tendinopathy. The mechanisms of obesity-induced chronic inflammation in adipose tissue are firstly reviewed. Common inflammatory mediators potentially linking obesity and the development of tendinopathy, and their association with mechanical overuse, are discussed, along with pre-clinical evidences and a systematic literature search on clinical studies. The potential contribution of local adipose tissues in the promotion of inflammation, pain and tendon degeneration is then discussed. The future research directions are proposed. Translational potential statement Better understanding of the roles of obesity-associated inflammatory mediators on tendons will clarify the pathophysiological drivers of tendinopathy in patients with obesity and identify possible treatment targets. Further studies on the mechanisms of obesity-induced chronic inflammation on tendon are a promising direction for the treatment of tendinopathy.
Collapse
Affiliation(s)
- Pauline Po Yee Lui
- Corresponding author. Room 74037, 5/F, Lui Che Woo Clinical Sciences Building, Prince of Wales Hospital, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, China.
| | | |
Collapse
|
143
|
Lizcano F. Roles of estrogens, estrogen-like compounds, and endocrine disruptors in adipocytes. Front Endocrinol (Lausanne) 2022; 13:921504. [PMID: 36213285 PMCID: PMC9533025 DOI: 10.3389/fendo.2022.921504] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Women are subject to constitutional changes after menopause, which increases conditions and diseases prone to cardiovascular risks such as obesity and diabetes mellitus. Both estrogens and androgens influence the individual's metabolic mechanism, which controls the fat distribution and the hypothalamic organization of the regulatory centers of hunger and satiety. While androgens tend to accumulate fat in the splanchnic and the visceral region with an increase in cardiovascular risk, estrogens generate more subcutaneous and extremity distribution of adipose tissue. The absence of estrogen during menopause seems to be the main factor that gives rise to the greater predisposition of women to suffer cardiovascular alterations. However, the mechanisms by which estrogens regulate the energy condition of people are not recognized. Estrogens have several mechanisms of action, which mainly include the modification of specific receptors that belong to the steroid receptor superfamily. The alpha estrogen receptors (ERα) and the beta receptors (ERβ) have a fundamental role in the metabolic control of the individual, with a very characteristic corporal distribution that exerts an influence on the metabolism of lipids and glucose. Despite the significant amount of knowledge in this field, many of the regulatory mechanisms exerted by estrogens and ER continue to be clarified. This review will discuss the role of estrogens and their receptors on the central regulation of caloric expenditure and the influence they exert on the differentiation and function of adipocytes. Furthermore, chemical substances with a hormonal activity that cause endocrine disruption with affectation on estrogen receptors will be considered. Finally, the different medical therapies for the vasomotor manifestations of menopause and their role in reducing obesity, diabetes, and cardiovascular risk will be analyzed.
Collapse
|
144
|
Nasr MJC, Geerling E, Pinto AK. Impact of Obesity on Vaccination to SARS-CoV-2. Front Endocrinol (Lausanne) 2022; 13:898810. [PMID: 35795152 PMCID: PMC9252434 DOI: 10.3389/fendo.2022.898810] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/17/2022] [Indexed: 12/02/2022] Open
Abstract
To combat the immense toll on global public health induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), new vaccines were developed. While these vaccines have protected the populations who received them from severe SARS-CoV-2 infection, the effectiveness and durability of these vaccines in individuals with obesity are not fully understood. Our uncertainty of the ability of these novel vaccines to induce protective immunity in humans with obesity stems from historical data that revealed obesity-associated immune defects to influenza vaccines. This review analyzes the efficacy of SARS-CoV-2 vaccines in humans with obesity. According to the vaccine safety and efficacy information for the Pfizer, Moderna, and Johnson & Johnson formulations, these vaccines showed a similar efficacy in both individuals with and without obesity. However, clinical trials that assess BMI and central obesity showed that induced antibody titers are lower in individuals with obesity when compared to healthy weight subjects, highlighting a potential early waning of vaccine-induced antibodies linked to obesity rates. Thus, the desired protective effects of SARS-CoV-2 vaccination were potentially diminished in humans with obesity when compared to the healthy weight population, but further studies outlining functional implications of the link between obesity and lower antibody titers need to be conducted to understand the full impact of this immune phenomenon. Further, additional research must be completed to truly understand the immune responses mounted against SARS-CoV-2 in patients with obesity, and whether these responses differ from those elicited by previously studied influenza viruses.
Collapse
|
145
|
Çakır I, Hadley CK, Pan PL, Bagchi RA, Ghamari-Langroudi M, Porter DT, Wang Q, Litt MJ, Jana S, Hagen S, Lee P, White A, Lin JD, McKinsey TA, Cone RD. Histone deacetylase 6 inhibition restores leptin sensitivity and reduces obesity. Nat Metab 2022; 4:44-59. [PMID: 35039672 PMCID: PMC8892841 DOI: 10.1038/s42255-021-00515-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/07/2021] [Indexed: 01/05/2023]
Abstract
The adipose tissue-derived hormone leptin can drive decreases in food intake while increasing energy expenditure. In diet-induced obesity, circulating leptin levels rise proportionally to adiposity. Despite this hyperleptinemia, rodents and humans with obesity maintain increased adiposity and are resistant to leptin's actions. Here we show that inhibitors of the cytosolic enzyme histone deacetylase 6 (HDAC6) act as potent leptin sensitizers and anti-obesity agents in diet-induced obese mice. Specifically, HDAC6 inhibitors, such as tubastatin A, reduce food intake, fat mass, hepatic steatosis and improve systemic glucose homeostasis in an HDAC6-dependent manner. Mechanistically, peripheral, but not central, inhibition of HDAC6 confers central leptin sensitivity. Additionally, the anti-obesity effect of tubastatin A is attenuated in animals with a defective central leptin-melanocortin circuitry, including db/db and MC4R knockout mice. Our results suggest the existence of an HDAC6-regulated adipokine that serves as a leptin-sensitizing agent and reveals HDAC6 as a potential target for the treatment of obesity.
Collapse
Affiliation(s)
- Işın Çakır
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
| | - Colleen K Hadley
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- College of Literature, Science and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Pauline Lining Pan
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Rushita A Bagchi
- Department of Medicine, Division of Cardiology and the Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Masoud Ghamari-Langroudi
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | | | - Qiuyu Wang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Michael J Litt
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Somnath Jana
- Chemical Synthesis Core, Vanderbilt Institute of Chemical Biology, Nashville, TN, USA
| | - Susan Hagen
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Pil Lee
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Andrew White
- Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Jiandie D Lin
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Cell & Developmental Biology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology and the Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
146
|
Bariatric Surgery Leads to a Reduction in Antibodies to Apolipoprotein A-1: a Prospective Cohort Study. Obes Surg 2021; 32:355-364. [PMID: 34888742 PMCID: PMC8794910 DOI: 10.1007/s11695-021-05738-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 10/31/2022]
Abstract
PURPOSE Autoantibodies against apolipoprotein A-1 have been associated with cardiovascular disease, poorer CV outcomes and all-cause mortality in obese individuals. The impact of bariatric surgery (BS) on the presence of circulating anti-apoA-1 IgG antibodies is unknown. This study aimed to determine the effect of bariatric surgery on auto-antibodies titres against Apolipoprotein A-1 (anti-apoA-1 IgG), looking for changes associated with lipid parameters, insulin resistance, inflammatory profile and percentage of excess body mass index loss (%EBMIL). MATERIALS AND METHODS We assessed 55 patients (40 women) before, 6 and 12 months post-operatively. Baseline and post-operative clinical history and measurements of body mass index (BMI), serum cholesterol, triglycerides, high- and low-density lipoprotein cholesterol (HDL-C and LDL-C), apoA-1, highly sensitive C-reactive protein (hsCRP), fasting glucose (FG), glycated haemoglobin (HbA1c) and HOMA-IR were taken at each point. Human anti-apoA-1 IgG were measured by ELISA. RESULTS The mean age of participants was 50 years. BS significantly improved BMI, %EBMIL triglycerides, HDL-C, apoA-1, hsCRP, HBA1c, FG and HOMA-IR. Baseline anti-apoA-1 IgG seropositivity was 25% and was associated with lower apoA-1 and higher hsCRP levels. One year after BS, anti-apoA-1 IgG seropositivity decreased to 15% (p = 0.007) and median anti-apoA-1 IgG values decreased from 0.70 (0.56-0.84) to 0.47 (0.37-0.61) AU (p < 0.001). Post-operative anti-apoA-1 IgG levels were significantly associated with a decreased post-surgical %EBMIL at 1 year. CONCLUSION Bariatric surgery results in significant reduction in anti-apoA-1 IgG levels, which may adversely influence weight loss. The exact mechanisms underpinning these results are elusive and require further study before defining any clinical recommendations.
Collapse
|
147
|
García-Estevez L, González-Martínez S, Moreno-Bueno G. The Leptin Axis and Its Association With the Adaptive Immune System in Breast Cancer. Front Immunol 2021; 12:784823. [PMID: 34868066 PMCID: PMC8634160 DOI: 10.3389/fimmu.2021.784823] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022] Open
Abstract
Adipose tissue secretes various peptides, including leptin. This hormone acts through the leptin receptor (Ob-R), which is expressed ubiquitously on the surface of various cells, including breast cancer cells and immune cells. Increasing evidence points to an interaction between the tumor microenvironment, tumor cells, and the immune system. Leptin plays an important role in breast cancer tumorigenesis and may be implicated in activation of the immune system. While breast cancer cannot be considered an immunogenic cancer, the triple-negative subtype is an exception. Specific immune cells - tumor infiltrating lymphocytes - are involved in the immune response and act as predictive and prognostic factors in certain breast cancer subtypes. The aim of this article is to review the interaction between adipose tissue, through the expression of leptin and its receptor, and the adaptive immune system in breast cancer.
Collapse
Affiliation(s)
- Laura García-Estevez
- Breast Cancer Department, MD Anderson Cancer Center, Madrid, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain
| | - Silvia González-Martínez
- Pathology Department, Hospital Ramón y Cajal, Madrid, Spain.,Fundación Contigo Contra el Cáncer de la Mujer, Madrid, Spain
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain.,Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), IdiPaz, Madrid, Spain
| |
Collapse
|
148
|
Bean C, Audano M, Varanita T, Favaretto F, Medaglia M, Gerdol M, Pernas L, Stasi F, Giacomello M, Herkenne S, Muniandy M, Heinonen S, Cazaly E, Ollikainen M, Milan G, Pallavicini A, Pietiläinen KH, Vettor R, Mitro N, Scorrano L. The mitochondrial protein Opa1 promotes adipocyte browning that is dependent on urea cycle metabolites. Nat Metab 2021; 3:1633-1647. [PMID: 34873337 DOI: 10.1038/s42255-021-00497-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022]
Abstract
White to brown/beige adipocytes conversion is a possible therapeutic strategy to tackle the current obesity epidemics. While mitochondria are key for energy dissipation in brown fat, it is unknown if they can drive adipocyte browning. Here, we show that the mitochondrial cristae biogenesis protein optic atrophy 1 (Opa1) facilitates cell-autonomous adipocyte browning. In two cohorts of patients with obesity, including weight discordant monozygotic twin pairs, adipose tissue OPA1 levels are reduced. In the mouse, Opa1 overexpression favours white adipose tissue expandability as well as browning, ultimately improving glucose tolerance and insulin sensitivity. Transcriptomics and metabolomics analyses identify the Jumanji family chromatin remodelling protein Kdm3a and urea cycle metabolites, including fumarate, as effectors of Opa1-dependent browning. Mechanistically, the higher cyclic adenosine monophosphate (cAMP) levels in Opa1 pre-adipocytes activate cAMP-responsive element binding protein (CREB), which transcribes urea cycle enzymes. Flux analyses in pre-adipocytes indicate that Opa1-dependent fumarate accumulation depends on the urea cycle. Conversely, adipocyte-specific Opa1 deletion curtails urea cycle and beige differentiation of pre-adipocytes, and is rescued by fumarate supplementation. Thus, the urea cycle links the mitochondrial dynamics protein Opa1 to white adipocyte browning.
Collapse
Affiliation(s)
- Camilla Bean
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Tatiana Varanita
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Marta Medaglia
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Gerdol
- Department of Life Science, University of Trieste, Trieste, Italy
| | - Lena Pernas
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Fabio Stasi
- Department of Medicine, University of Padova, Padova, Italy
| | | | - Stèphanie Herkenne
- Department of Biology, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Maheswary Muniandy
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emma Cazaly
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | | | | | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Obesity Centre, Abdominal Centre, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Roberto Vettor
- Department of Medicine, University of Padova, Padova, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Luca Scorrano
- Department of Biology, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
149
|
Impact of Bariatric Surgery on Adipose Tissue Biology. J Clin Med 2021; 10:jcm10235516. [PMID: 34884217 PMCID: PMC8658722 DOI: 10.3390/jcm10235516] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery (BS) procedures are actually the most effective intervention to help subjects with severe obesity achieve significant and sustained weight loss. White adipose tissue (WAT) is increasingly recognized as the largest endocrine organ. Unhealthy WAT expansion through adipocyte hypertrophy has pleiotropic effects on adipocyte function and promotes obesity-associated metabolic complications. WAT dysfunction in obesity encompasses an altered adipokine secretome, unresolved inflammation, dysregulated autophagy, inappropriate extracellular matrix remodeling and insufficient angiogenic potential. In the last 10 years, accumulating evidence suggests that BS can improve the WAT function beyond reducing the fat depot sizes. The causal relationships between improved WAT function and the health benefits of BS merits further investigation. This review summarizes the current knowledge on the short-, medium- and long-term outcomes of BS on the WAT composition and function.
Collapse
|
150
|
Quaresma PGF, Wasinski F, Mansano NS, Furigo IC, Teixeira PDS, Gusmao DO, Frazao R, Donato J. Leptin Receptor Expression in GABAergic Cells is Not Sufficient to Normalize Metabolism and Reproduction in Mice. Endocrinology 2021; 162:6353267. [PMID: 34402859 DOI: 10.1210/endocr/bqab168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Previous studies indicate that leptin receptor (LepR) expression in GABAergic neurons is necessary for the biological effects of leptin. However, it is not clear whether LepR expression only in GABAergic neurons is sufficient to prevent the metabolic and neuroendocrine imbalances caused by LepR deficiency. In the present study, we produced mice that express the LepR exclusively in GABAergic cells (LepRVGAT mice) and compared them with wild-type (LepR+/+) and LepR-deficient (LepRNull/Null) mice. Although LepRVGAT mice showed a pronounced reduction in body weight and fat mass, as compared with LepRNull/Null mice, male and female LepRVGAT mice exhibited an obese phenotype relative to LepR+/+ mice. Food intake was normalized in LepRVGAT mice; however, LepRVGAT mice still exhibited lower energy expenditure in both sexes and reduced ambulatory activity in the females, compared with LepR+/+ mice. The acute anorexigenic effect of leptin and hedonic feeding were normalized in LepRVGAT mice despite the hyperleptinemia they present. Although LepRVGAT mice showed improved glucose homeostasis compared with LepRNull/Null mice, both male and female LepRVGAT mice exhibited insulin resistance. In contrast, LepR expression only in GABAergic cells was sufficient to normalize the density of agouti-related peptide (AgRP) and α-MSH immunoreactive fibers in the paraventricular nucleus of the hypothalamus. However, LepRVGAT mice exhibited reproductive dysfunctions, including subfertility in males and alterations in the estrous cycle of females. Taken together, our findings indicate that LepR expression in GABAergic cells, although critical to the physiology of leptin, is insufficient to normalize several metabolic aspects and the reproductive function in mice.
Collapse
Affiliation(s)
- Paula G F Quaresma
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Naira S Mansano
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Isadora C Furigo
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Pryscila D S Teixeira
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Daniela O Gusmao
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| | - Renata Frazao
- Departamento de Anatomia, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Jose Donato
- Universidade de São Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, São Paulo, SP, 05508-000, Brazil
| |
Collapse
|