1
|
Wu G, Wang M, Du Z, Li Z, Han T, Xie Z, Gu W. Tea polyphenol EGCG enhances the improvements of calorie restriction on hepatic steatosis and obesity while reducing its adverse outcomes in obese rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156744. [PMID: 40228342 DOI: 10.1016/j.phymed.2025.156744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/24/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Currently, calorie restriction (CR) is popular among young people as a way to lose weight and prevent obesity. However, CR can also cause a series of side effects, such as weight regain after resuming free eating. Tea polyphenol epigallocatechin-3-gallate (EGCG) has been widely recognized as antiobesity effects. However, whether EGCG can enhance the antiobesity effect of CR and reduce its adverse outcomes is still unclear. PURPOSE This study aimed to explore the enhancing effect and molecular mechanism of EGCG supplementation on CR in improving hepatic steatosis and obesity. METHODS The enhancing effect and molecular mechanism of EGCG supplementation on CR in alleviating hepatic steatosis and obesity were explored using a leptin receptor-knockout (LepR KO) rat model by performing biochemical, histochemistry, qPCR, plasma lipidomic, and gut microbiota analysis. RESULTS Our results showed that CR plus EGCG exhibited enhanced preventive effects in reducing blood glucose, insulin, TC, TG, LDL-C, and FFA levels in plasma, and protection against hepatic steatosis in LepR KO rats than CR alone. In addition, CR plus EGCG remarkably reduced oxidative stress and systemic inflammatory responses in LepR KO rats. Moreover, the combined intervention showed an enhanced improvement effect on the homeostasis of gut microbiota than CR alone, including increasing gut microbiota diversity and modulating microbiota composition. Plasma lipidomics analysis showed that CR plus EGCG significantly improved glycerophospholipid, glycerolipid and sphingolipid metabolism in LepR KO rats. Mechanistic studies showed that CR combined EGCG enhanced SIRT6 and suppressed SREBP1 and FAS expression in the livers of LepR KO rats than CR alone, thereby improving host lipid metabolism. CONCLUSION This study demonstrated that EGCG enhance the improvements of CR on hepatic steatosis and obesity in LepR KO rats, and reduce its adverse outcomes, especially in reducing hepatic lipogenesis and maintaining homeostasis of gut microbiota. This study provides a dietary strategy for preventing weight rebound following the transition from CR to a free diet by supplementing EGCG, suggesting that CR plus EGCG may offer a promising therapy for managing obesity in humans.
Collapse
Affiliation(s)
- Guohuo Wu
- Engineering Technology Research Center of Anti-Aging Chinese Herbal Medicine of Anhui Province, School of Biology and Food Engineering, Fuyang Normal University, Fuyang, China; State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Mengdi Wang
- Department of General Surgery, The First Affiliated Hospital of Fuyang Normal University, Fuyang, China
| | - Zhaofeng Du
- Engineering Technology Research Center of Anti-Aging Chinese Herbal Medicine of Anhui Province, School of Biology and Food Engineering, Fuyang Normal University, Fuyang, China; State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Zhuang Li
- Biotechnology Center, Anhui Agricultural University, Hefei, China
| | - Tingting Han
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| | - Wei Gu
- Laboratory Animal Center, Anhui Medical University, Hefei, China; State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
2
|
Chew V. Why weight loss is only half the battle: The epigenetic memory of adipose tissue. J Hepatol 2025; 82:938-939. [PMID: 39833020 DOI: 10.1016/j.jhep.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Affiliation(s)
- Valerie Chew
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore 169856, Singapore.
| |
Collapse
|
3
|
Sindi H, Almuzaini S, Mubarak A, Hakeem FF, Campus G, Fadel HT, Lingström P. Oral Health in Individuals After Bariatric Surgery: A Systematic Scoping Review. Obes Surg 2025; 35:1878-1899. [PMID: 40106168 PMCID: PMC12065770 DOI: 10.1007/s11695-025-07793-w] [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] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
This scoping review appraises the available literature that investigates oral health after bariatric surgery (BS). Forty-two records were included (33 original studies and 9 systematic reviews). Bariatric surgery had a negative impact on the oral health related quality of life (three out of six studies). Relatively few studies found improvement following BS, probing pocket depth (2/10), clinical attachment level (1/9), bleeding on probing (3/10) and plaque (3/8). Conversely, BS was negatively associated with the DMFT (4/6), tooth wear (4/6) and oral radiographic findings (1/1). There was conflicting evidence regarding salivary parameters. To conclude, the literature on oral health in bariatric surgery patients is continuously growing. Positive as well as negative associations between oral health parameters and bariatric surgery are observed.
Collapse
Affiliation(s)
- Hisham Sindi
- Department of Cariology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Preventive Dental Sciences, College of Dentistry, Taibah University, AlMadinah AlMunawwarah, Saudi Arabia.
| | - Sarah Almuzaini
- College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Arwa Mubarak
- College of Dentistry Hospital, Taibah University, AlMadinah AlMunawwarah, Saudi Arabia
| | - Faisal F Hakeem
- Department of Preventive Dental Sciences, College of Dentistry, Taibah University, AlMadinah AlMunawwarah, Saudi Arabia
- Centre for Epidemiology Versus Arthritis, The University of Manchester, Manchester, UK
| | - Guglielmo Campus
- Department of Cariology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hani T Fadel
- Department of Preventive Dental Sciences, College of Dentistry, Taibah University, AlMadinah AlMunawwarah, Saudi Arabia.
| | - Peter Lingström
- Department of Cariology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
4
|
Carlsson LM, Arnetorp I, Andersson-Assarsson JC, Jacobson P, Svensson PA, Taube M, Ahlin S, Kristensson FM, Karason K, Larsson I, Karlsson C, Pietiläinen KH, Näslund I, Carlsson B, Peltonen M, Sjöholm K. Health outcomes and their association with weight regain after substantial weight loss in Sweden: a prospective cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2025; 52:101261. [PMID: 40166366 PMCID: PMC11957514 DOI: 10.1016/j.lanepe.2025.101261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
Background The clinical implications of weight regain following weight loss remain uncertain. We analysed mortality, cardiovascular events, cancer, and microvascular disease in individuals with significant weight loss, comparing maintainers to regainers. Methods Using a prospective cohort design, we analysed 1346 participants who underwent bariatric surgery in the Swedish Obese Subjects (SOS) study, aged 37-60 years with BMI ≥34 (men) or ≥38 (women), recruited 1987-2001. Individuals who regained ≥20% of their 1-year weight loss after 4 years (regain group) were compared to those who regained less (maintenance group). The study was closed on December 31, 2020 with median follow-up of 27 years and 99.9% mortality tracking (ClinicalTrials.govNCT01479452). Findings Average weight loss after 1 year was 29.3 ± 11.7 kg and 31.9 ± 13.8 kg and average weight change from year 1 to year 4 was +12.7 ± 6.6 kg and -0.6 ± 7.3 kg in the regain and maintenance groups, respectively. During follow-up, regain and maintenance groups showed similar rates of total mortality and cancer, 12.4 (95% CI: 10.9-14.2) vs 12.4 (10.7-14.3), p = 0.740, and 11.3 (95% CI: 9.7-13.0) vs 10.4 (8.8-12.2) per 1000 person-years (p = 0.308), respectively. The regain group had, however, higher incidence of microvascular disease, 11.0 (95% CI: 9.5-12.8) vs 8.7 (7.3-10.4) per 1000 person-years (p = 0.024), and while not statistically significant, also higher incidence of major adverse cardiovascular events (myocardial infarction, stroke, and heart failure) 15.7 (95% CI: 13.8-17.8) vs 13.0 (11.2-15.1) per 1000 person-years (p = 0.055). Interpretation Weight regain was linked to increased vascular disease risk but we could not demonstrate an association with life expectancy. Funding The Swedish Research Council, the Swedish State under the agreement between the Swedish Government and the county councils, the Health & Medical Care Committee of the Region Västra Götaland, the Adlerbert Research Foundation, the Wilhelm and Martina Lundgren Foundation, the Royal Society of Arts and Sciences in Gothenburg, Academy of Finland, Finnish Medical Foundation, Gyllenberg Foundation, Novo Nordisk Foundation, Finnish Diabetes Research Foundation, Paulo Foundation and Sigrid Juselius Foundation.
Collapse
Affiliation(s)
- Lena M.S. Carlsson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Ida Arnetorp
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | - Peter Jacobson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Per-Arne Svensson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Institute of Health and Care Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Magdalena Taube
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sofie Ahlin
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Region of Västra Götaland, NU Hospital Group, Department of Clinical Physiology, Trollhättan, Sweden
| | - Felipe M. Kristensson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital/Östra, Department of Surgery, Gothenburg, Sweden
| | - Kristjan Karason
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Departments of Cardiology and Transplantation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ingrid Larsson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Karlsson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Late-Stage Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Healthy Weight Hub, Abdominal Centre, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Ingmar Näslund
- Faculty of Medicine and Health, Department of Surgery, Örebro University, Örebro, Sweden
| | - Björn Carlsson
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Kajsa Sjöholm
- Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
5
|
Na ES. Epigenetic Mechanisms of Obesity: Insights from Transgenic Animal Models. Life (Basel) 2025; 15:653. [PMID: 40283207 PMCID: PMC12028693 DOI: 10.3390/life15040653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Obesity is a chronic disease with prevalence rates that have risen dramatically over the past four decades. This increase is not due to changes in the human genome but rather to environmental factors that promote maladaptive physiological responses. Emerging evidence suggests that external influences, such as high-fat diets, modify the epigenome-the interface between genes and the environment-leading to persistent alterations in energy homeostasis. This review explores the role of epigenetic mechanisms in obesity, emphasizing insights from transgenic animal models and clinical studies. Additionally, we discuss the evolution of obesity research from homeostatic to allostatic frameworks, highlighting key neuroendocrine regulators of energy balance.
Collapse
Affiliation(s)
- Elisa S Na
- School of Social Work, Psychology, & Philosophy, Texas Woman's University, Denton, TX 76209, USA
| |
Collapse
|
6
|
van Baak MA, Mariman ECM. Physiology of Weight Regain after Weight Loss: Latest Insights. Curr Obes Rep 2025; 14:28. [PMID: 40163180 PMCID: PMC11958498 DOI: 10.1007/s13679-025-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE OF REVIEW This review summarizes the most recent research on the physiology of weight regain. It describes developments in areas that are currently being addressed and that may indicate promising directions for future research. RECENT FINDINGS Weight regain occurs independent of the way prior weight loss is achieved, i.e. by lifestyle, surgery or pharmacotherapy. Recent novel findings regarding weight regain belong to four areas. First, the immune obesity memory of which besides persistent immune cells promoting weight regain cells have been found that reduce weight regain. Second, the gut microbiome where autologous transplantation can limit weight regain. Third, the composition of the weight loss with the percentage of lost fat-free mass being inverse to the amount of regained weight independent of the weight loss procedure. Fourth, appetite control where after weight loss altered hypothalamic activity promoting hunger and weight regain persists, possibly mediated by altered neurotensin responses. In all four areas more conclusive evidence for their role in weight regain still needs to be obtained. Most studies on physiological mechanisms of weight regain are associative in nature and the number of intervention studies is very limited. To bring the field further, carefully designed intervention studies taking into account the dynamic character of weight loss and weight regain are needed.
Collapse
Affiliation(s)
- Marleen A van Baak
- Department of Human Biology, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and life Sciences+, Maastricht University, Maastricht, The Netherlands.
- Department of Human Biology, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University, PO Box 616, Maastricht, 6200MD, The Netherlands.
| | - Edwin C M Mariman
- Department of Human Biology, NUTRIM Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and life Sciences+, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
7
|
Lin Y, Shi J, Yu X, Sun J, Lixia S, Dou J, Zhang M, Li X, Tian Z, Deng H, Feng B, Su Q, Peng Y. Enhancing Diabetes Treatment: Comparing Pioglitazone/Metformin with Dapagliflozin Versus Basal Insulin/Metformin in Type 2 Diabetes. Drug Des Devel Ther 2025; 19:1795-1808. [PMID: 40098912 PMCID: PMC11911819 DOI: 10.2147/dddt.s512872] [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] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/01/2025] [Indexed: 03/19/2025] Open
Abstract
Aim The aim of this study was to compare the efficacy and safety of fixed-dose combination (FDC) of pioglitazone and metformin supplemented with dapagliflozin (test group) with those of basal insulin supplemented with metformin (control group) in patients with inadequately controlled type 2 diabetes mellitus (T2DM). Methods This 16-week, prospective, randomized, open-label study enrolled patients aged 18-75 years with glycated hemoglobin (HbA1c) levels between ≥ 8% and ≤ 11%. The primary endpoint was the proportion of patients who achieved HbA1c < 7% at week 16 without hypoglycemia or weight gain. The secondary endpoints included blood glucose, lipid profile, body weight, body mass index, inflammatory markers, bone Gla-protein, liver enzymes, and patient satisfaction. Results Among the full analysis set of 147 participants, no significant difference was observed in the primary endpoint between the test group and the control group. However, the test group had a higher percentage of patients who achieved HbA1c <7% at week 16 without hypoglycemia and experienced a weight loss of ≥3% (31.51% vs 13.51%, P=0.009). Patients in the test group whose BMI≥24 kg/m2 also achieved a substantial achievement rate (36.73% vs 15.79%, P=0.014). The test group also exhibited a greater reduction in body weight and improvements in 2-hour postprandial glucose level, systolic blood pressure, and lipid profile. Notably, combination therapy did not increase the risk of hypoglycemia or weight gain. Patients in the test group were more satisfied than those in the control group with continuing to accept pioglitazone/metformin FDC combined with dapagliflozin. Conclusion In the absence of contraindications, pioglitazone/metformin FDC supplemented with dapagliflozin may serve as a safe and effective alternative to basal insulin combined with metformin for rectifying inadequate glucose control, as the former enables metabolic improvements without compromising safety. Chinese Clinical Trial Registry Number CHiCTR2000036076. https://www.chictr.org.cn/showproj.html?proj=58825.
Collapse
Affiliation(s)
- Yi Lin
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Jianxia Shi
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xuemei Yu
- Central Hospital of Fengxian District, Shanghai, People’s Republic of China
| | - Jiao Sun
- Huadong Hospital Affiliated to Fudan University, Shanghai, People’s Republic of China
| | - Suo Lixia
- Shanghai Jiading Central Hospital, Shanghai Jiading Central Hospital, Shanghai, People’s Republic of China
| | - Jiaqing Dou
- Chaohu Hospital of Anhui Medical University, Chaohu, People’s Republic of China
| | - Min Zhang
- Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, People’s Republic of China
| | - Xiaohua Li
- Seventh People’s Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhufang Tian
- Xi‘an Central Hospital, Xi’an, Shanxi, People’s Republic of China
| | - Hongyan Deng
- Wuhan Fourth Hospital, Wuhan, People’s Republic of China
| | - Bo Feng
- Dongfang Hospital Affiliated to Tongji University, Shanghai, People’s Republic of China
| | - Qing Su
- Xinhua Hospital Affiliated to Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Yongde Peng
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
8
|
Nor Hanipah Z, Abdul Ghani R, Goon MDME. ACTION Malaysia-perception and barriers to obesity management among people with obesity and healthcare professionals in Malaysia. BMC Public Health 2025; 25:835. [PMID: 40033266 PMCID: PMC11874754 DOI: 10.1186/s12889-025-22052-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Timely weight loss conversations between healthcare professionals (HCPs) and people with obesity (PwO) can help in effective obesity management. The Awareness, Care, and Treatment in Obesity maNagement in the Asia Pacific region (ACTION APAC) studied the attitudes, perceptions, and behaviours toward obesity among PwO and HCPs in nine countries of South and Southeast Asia. The current study is a subgroup analysis based on the Malaysian population, known as ACTION Malaysia (ACTION-MY), and aims to explore the attitudes, perceptions, behaviours, and barriers to effective obesity management among both PwO and HCPs. METHODOLOGY An online survey in dual languages (Malay and English) was conducted between April 2022 and May 2022 among 1001 adult PwO and 200 HCPs (general practitioners, endocrinologists, obstetricians/gynaecologists, cardiologists, and other appropriate specialities). RESULTS The findings highlighted significant gaps in obesity awareness, with 57% of PwO misclassifying their weight status as normal or overweight. While 68% of PwO valued discussing weight management with HCPs, success rates remained low. On average, patients made three weight loss attempts in adulthood, with 63% regaining weight even after maintaining weight loss for six months or more. Key barriers included insufficient exercise, motivation deficits, and poor hunger control. Although 88% of HCPs recognised obesity as a chronic disease affecting overall health, patient disinterest, limited awareness of treatment options, and time constraints hindered effective intervention. Despite 70% of PwO trusting HCPs' medication recommendations, only 10% received weight loss prescriptions. CONCLUSIONS This study emphasises the need for enhanced communication between HCPs and PwO, along with comprehensive support that includes mental health services. Addressing the perception gap regarding weight management responsibility is crucial. The results suggest that culturally contextualised approaches to obesity management in Malaysia are essential. Our findings highlight the urgent need for developing treatment strategies and policies targeting identified barriers and establishing collaborative frameworks to enhance obesity management within Malaysia's healthcare system.
Collapse
Affiliation(s)
- Zubaidah Nor Hanipah
- Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| | - Rohana Abdul Ghani
- Department of Internal Medicine, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerFoRM), Universiti Teknologi MARA, Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Mohd Danial Mohd Efendy Goon
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerFoRM), Universiti Teknologi MARA, Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
- Novo Nordisk Pharma (Malaysia) Sdn. Bhd. Menara 1 Sentrum, Level 16, No. 201 Jalan Tun Sambanthan, Kuala Lumpur, 50470, Malaysia
| |
Collapse
|
9
|
Pahlavani M, Pham K, Kalupahana NS, Morovati A, Ramalingam L, Abidi H, Kiridana V, Moustaid-Moussa N. Thermogenic adipose tissues: Promising therapeutic targets for metabolic diseases. J Nutr Biochem 2025; 137:109832. [PMID: 39653156 DOI: 10.1016/j.jnutbio.2024.109832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
The ongoing increase in the prevalence of obesity and its comorbidities such as cardiovascular disease, type 2 diabetes (T2D) and dyslipidemia warrants discovery of novel therapeutic options for these metabolic diseases. Obesity is characterized by white adipose tissue expansion due to chronic positive energy balance as a result of excessive energy intake and/or reduced energy expenditure. Despite various efforts to prevent or reduce obesity including lifestyle and behavioral interventions, surgical weight reduction approaches and pharmacological methods, there has been limited success in significantly reducing obesity prevalence. Recent research has shown that thermogenic adipocyte (brown and beige) activation or formation, respectively, could potentially act as a therapeutic strategy to ameliorate obesity and its related disorders. This can be achieved through the ability of these thermogenic cells to enhance energy expenditure and regulate circulating levels of glucose and lipids. Thus, unraveling the molecular mechanisms behind the formation and activation of brown and beige adipocytes holds the potential for probable therapeutic paths to combat obesity. In this review, we provide a comprehensive update on the development and regulation of different adipose tissue types. We also emphasize recent interventions in harnessing therapeutic potential of thermogenic adipocytes by bioactive compounds and new pharmacological anti-obesity agents.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Sciences, Texas Woman's University, Dallas, Texas, USA
| | - Kenneth Pham
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Ashti Morovati
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Studies, Syracuse University, Syracuse, New York, USA
| | - Hussain Abidi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Vasana Kiridana
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Institute for One Health Innovation, Texas Tech University and Texas Tech Health Sciences Center, Lubbock, Texas, USA.
| |
Collapse
|
10
|
Zhang J, Li S, Cheng X, Tan X, Shi Y, Su G, Huang Y, Zhang Y, Xue R, Li J, Fan Q, Dong H, Deng Y, Zhang Y. Graphene-Based Far-Infrared Therapy Promotes Adipose Tissue Thermogenesis and UCP1 Activation to Combat Obesity in Mice. Int J Mol Sci 2025; 26:2225. [PMID: 40076847 PMCID: PMC11900916 DOI: 10.3390/ijms26052225] [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] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Hyperthermia (HT) has broad potential for disease treatment and health maintenance. Previous studies have shown that far-infrared rays (FIRs) at 8-10 μm can potentially reduce inflammation, oxidative stress, and gut microbiota imbalance. However, the effects of FIR HT on energy metabolism require further investigation. To investigate the effects of graphene-FIR HT therapy on diet-induced obesity and their regulatory mechanisms in energy metabolism disorders. After 8 weeks of hyperthermia, mice fed standard chow or a high-fat diet (HFD) underwent body composition analysis. Energy expenditure was measured using metabolic cages. The protein changes in adipose tissue were detected by molecular technology. Graphene-FIR therapy effectively mitigated body fat accumulation, improved dyslipidemia, and impaired liver function while enhancing insulin sensitivity. Furthermore, graphene-FIR therapy increased VO2, VCO2, and EE levels in HFD mice to exhibit enhanced metabolic activity. The therapy activated the AMPK/PGC-1α/SIRT1 pathway in adipose tissue, increasing the expression of uncoupling protein 1 (UCP1) and glucose transporter protein four (GLUT4), activating the thermogenic program in adipose tissue, and improving energy metabolism disorder in HFD mice. In short, graphene-FIR therapy represents a comprehensive approach to improving the metabolic health of HFD mice.
Collapse
Affiliation(s)
- Jinshui Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Shuo Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Xin Cheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Xiaocui Tan
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Yingxian Shi
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Guixin Su
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Yulong Huang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Yang Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Rui Xue
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Jingcao Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Qiongyin Fan
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Huajin Dong
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| | - Yun Deng
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Youzhi Zhang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (J.Z.); (S.L.); (X.C.); (X.T.); (Y.S.); (G.S.); (Y.H.); (Y.Z.); (R.X.); (J.L.); (Q.F.); (H.D.)
| |
Collapse
|
11
|
Liu C, Xin Y, Huang Y, Xu L, Zhou R, Wang Y, Wang W. Reduction of Hepatic Fat Content by Dulaglutide for the Treatment of Diabetes Mellitus: A Two-Centre Open, Single-Arm Trial. Endocrinol Diabetes Metab 2025; 8:e70021. [PMID: 39718468 DOI: 10.1002/edm2.70021] [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: 07/24/2024] [Revised: 10/07/2024] [Accepted: 11/06/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND With the elevated level of NAFLD prevalence, the incidence of diabetes, hypertension, metabolic syndrome and other diseases is also significantly elevated. GLP-1RA can exert weight loss, glucose-lowering effects and various nonglycaemic effects. However, the relationship between quantitative reduction in hepatic fat content and improvement of pancreatic islet function by GLP-1RA is unclear. METHODS This trial was a single-arm open cohort study. A total of 38 patients with T2DM and NAFLD were enrolled in the GLP-1RA treatment group. The included patients were tested for biochemical and blood glucose levels, adiponectin and FGF21 levels, and liver fat content was measured using MRI. Measure the above indicators again after at least 3 months of GLP-1RA treatment. Divided into Q1 (average decrease of 0.37%) and Q2 (average decrease of 8.6%) groups based on the degree of reduction in liver fat content. RESULTS Q2 group showed an average reduction in liver fat content of 8.6%, a decrease in glycated haemoglobin of 18.17%, a weight loss of 7.29% and an increase in fasting c-peptide release by 1.03%, 1-h and 2-h postprandial c-peptide release by 28.86% and 18.28% respectively. In contrast, Q1 group had an average reduction in liver fat content of 0.37%, a decrease in glycated haemoglobin of only 6.53%, a weight loss of 3.41%, a decrease in fasting c-peptide release by 1.91% and an increase in 1-h and 2-h postprandial c-peptide release by 19.18% and 11.66% respectively. CONCLUSION Reduction in liver fat content effectively improves pancreatic islet function secretion, particularly postprandial c-peptide secretion, especially in the first hour after a meal. This improvement leads to a decrease in glycated haemoglobin levels and promotes better compliance with blood glucose control.
Collapse
Affiliation(s)
- Chuanfeng Liu
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yu Xin
- Department of Endocrinology and Metabolic Diseases, Jiaozuo People's Hospital, Jiaozuo, China
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yajing Huang
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Xu
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruizhi Zhou
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Wang
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
de Faria MHS, Barroso LSS, Souza-Gomes AF, de Barros JLVM, Kakehasi AM, Vieira ELM, Silva ACSE, Nunes-Silva A. Strength Training can Modulate Urinary Adipokine Levels in Healthy Young Males. INTERNATIONAL JOURNAL OF EXERCISE SCIENCE 2025; 18:107-118. [PMID: 39916794 PMCID: PMC11798553 DOI: 10.70252/fxqy9475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Adipose tissue stores energy in fat-rich adipocytes, which can produce and release several adipokines and modulate body metabolism. Exercise may induce adipokine production in adipocytes; however, the relationship between the two remains unclear. Few studies have shown the relationship between adipokines and strength training. Thus, we aimed to evaluate the acute and chronic effects of strength training (ST) on urinary adiponectin, leptin, and resistin levels. Twelve untrained young men (23.42 ± 2.67 years) were included in this study. Body composition was evaluated at baseline and after completing of the training protocol using densitometry. Training protocol consisted of three exercises with three sets of 65% of one-repetition maximum (1MR) with a pause of 90 s between sets, each exercise lasting 5 s (2 s concentric / 3 s eccentric). The sessions were carried out three times a week for 10 weeks. Urine was collected during the pre- and post-training in the first and 30th session. Adipokine levels were determined by ELISA. Urinary levels of leptin acutely increased after the first ST session, and after the last ST session. Chronic changes in the leptin levels were also found when comparing the values before the last ST and before the first ST session. Urinary adiponectin levels changed in the comparison of values before and after the last session. There was a significant increase in the adiponectin levels when comparing values after the first and last ST sessions. The levels of resistin chronically increased. Strength training can induce acute and chronic changes in urinary levels of adipokines.
Collapse
Affiliation(s)
- Marcelo Henrique Salviano de Faria
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Laboratório de Inflamação e Imunologia do Exercício, Departamento de Educação Física e Esportes, Escola de Educação Física da Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Lucélia Scarabeli Silva Barroso
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- Laboratório de Inflamação e Imunologia do Exercício, Departamento de Educação Física e Esportes, Escola de Educação Física da Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Antonio Felipe Souza-Gomes
- Laboratório de Inflamação e Imunologia do Exercício, Departamento de Educação Física e Esportes, Escola de Educação Física da Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - João Luís Vieira Monteiro de Barros
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Adriana Maria Kakehasi
- Departamento do Aparelho Locomotor, Faculdade de Medicina, Universidade Federal de Minas Gerais, Brazil
| | - Erica Leandro Marciano Vieira
- Centre for Addiction and Mental Health, Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Albená Nunes-Silva
- Laboratório de Inflamação e Imunologia do Exercício, Departamento de Educação Física e Esportes, Escola de Educação Física da Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| |
Collapse
|
13
|
Dong Y, Dong J, Xiao H, Li Y, Wang B, Zhang S, Cui M. A gut microbial metabolite cocktail fights against obesity through modulating the gut microbiota and hepatic leptin signaling. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:9356-9367. [PMID: 39030978 DOI: 10.1002/jsfa.13758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/09/2024] [Accepted: 06/26/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Excessive body weight and obesity elevate the risk of chronic non-communicable diseases. The judicious application of the gut microbiome, encompassing both microorganisms and their derived compounds, holds considerable promise in the treatment of obesity. RESULTS In this study, we showed that a cocktail of gut microbiota-derived metabolites, comprising indole 3-propionic acid (IPA), sodium butyrate (SB) and valeric acid (VA), alleviated various symptoms of obesity in both male and female mice subjected to a high-fat diet (HFD). The 16S ribosomal RNA (rRNA) sequencing revealed that administering the cocktail via oral gavage retained the gut microbiota composition in obese mice. Fecal microbiota transplantation using cocktail-treated mice as donors mitigated the obesity phenotype of HFD-fed mice. Transcriptomic sequencing analysis showed that the cocktail preserved the gene expression profile of hepatic tissues in obese mice, especially up-regulated the expression level of leptin receptor. Gene delivery via in vivo fluid dynamics further validated that the anti-obesity efficacy of the cocktail was dependent on leptin signaling at least partly. The cocktail also inhibited the expression of appetite stimulators in hypothalamus. Together, the metabolite cocktail combated adiposity by retaining the gut microbiota configuration and activating the hepatic leptin signaling pathway. CONCLUSIONS Our findings provide a sophisticated regulatory network between the gut microbiome and host, and highlight a cocktail of gut microbiota-derived metabolites, including IPA, SB, and VA, might be a prospective intervention for anti-obesity in a preclinical setting. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yanxi Dong
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiali Dong
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huiwen Xiao
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yuan Li
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Bin Wang
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shuqin Zhang
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming Cui
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
14
|
Fortes YM, Souza-Gomes AF, Moreira ARS, Campos LN, de Moura SS, Barroso LSS, de Faria MHS, de Barros Fernandes H, de Miranda AS, Martins-Costa HC, Simões e Silva AC, Moreira JM, Nunes-Silva A. A single session of strength training changed plasma levels of resistin, but not leptin in overweight and obese men. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:324-330. [PMID: 39309458 PMCID: PMC11411330 DOI: 10.1016/j.smhs.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 09/25/2024] Open
Abstract
Obesity has a complex multifactorial etiology and is characterized by excessive accumulation of adipose tissue. Visceral adipose tissue has deleterious effects on health because it secretes large amounts of inflammatory cytokines. Nutritional calorie restriction associated with strength training may be useful in managing chronic systemic inflammation. This study aimed to evaluate the acute effect of a single strength-training session on plasma adipokine levels in sedentary, overweight, and obese young men. This study included twelve men (Age: [34.95 ± 9.77] years; Height: [174.16 ± 3.66] centimeter [cm]; Weight: [97.83 ± 12.87] kilogram (kg); body mass index [BMI]: [32.30 ± 4.51] kg/m2), who performed a single strength training session. The strength training protocol consisted of 4 sets of 12 repetitions in the following six exercises, 45° leg press, bench press, leg extension, machine row, leg curl, and shoulder press. Blood samples were collected before, immediately after, and 1-h subsequent after strength training. The plasma levels of resistin and leptin were measured. A significant decrease in resistin levels were found 1 h after the strength training session if compared to levels before the training session (pre-[before] [2 390 ± 1 199] picograms per milliliter [pg/mL] vs post-1 h [1-h subsequent] [1 523 ± 798],6 pg/mL, p = 0.002 8). The plasma leptin levels did not differ at any time point. In conclusion, a very well controlled single session of strength training significantly decreased the plasma levels of resistin without altering the concentration of leptin in overweight and obese individuals. This effect, at least in part, supports the benefits of exercise by reducing the low grade inflammation and insulin resistance in obesity.
Collapse
Affiliation(s)
- Yago Martins Fortes
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Antonio Felipe Souza-Gomes
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Alessandro Roberto Silveira Moreira
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Leo Nogueira Campos
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Samara Silva de Moura
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Lucélia Scarabeli Silva Barroso
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Interdisciplinary Medical Research Laboratory, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcelo Henrique Salviano de Faria
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Interdisciplinary Medical Research Laboratory, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Heliana de Barros Fernandes
- Neurobiology Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline Silva de Miranda
- Neurobiology Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Ana Cristina Simões e Silva
- Interdisciplinary Medical Research Laboratory, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Janaina Matos Moreira
- Department of Pediatrics, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Albená Nunes-Silva
- Laboratory of Inflammation and Exercise Immunology, Department of Physical Education, School of Physical Education, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Postgraduate Program in Health and Nutrition, Nutrition School, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| |
Collapse
|
15
|
Lin H, Wang Q, Gao A, Sun Y, Shen C, Chen Y, Wang Z, Xu X, Ni M, Chen Y, Zhang J, Luo Y, Lin X, Bi Y, Ning G, Wang W, Hong J, Gu W, Wang J, Liu R. Enteropancreatic hormone changes in caloric-restricted diet interventions associate with post-intervention weight maintenance. Clin Nutr 2024; 43:5-14. [PMID: 39418916 DOI: 10.1016/j.clnu.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/12/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND & AIMS To explore enteropancreatic hormone changes during isocaloric-restricted dietary interventions and their impact on post-intervention weight maintenance. METHODS 253 individuals with overweight/obesity and prediabetes were randomly assigned to 25% isocaloric-restricted diets: Control diet, Traditional Jiangnan diet or Mediterranean diet. Serum hormones and clinical indices were evaluated at 0, 3 and 6 months. Body weight values were collected again 6 months after completing interventions. RESULTS We observed decreased fasting and post-glucose load levels of glucagon, amylin, peptide YY, and glucagon-like peptide-1 (GLP-1) while increased ghrelin at three months after 25% calorie restriction (CR) of three dietary interventions, and most of these changes were sustained through the six month-treatment period. Interestingly, changes in appetite-inhibitory hormones glucagon, amylin and GLP-1 showed positive associations with body weight change while appetite-promoting hormone ghrelin showed an inverse association during intervention. Furthermore, subjects with more reduction in amylin and GLP-1, or more increase in ghrelin during intervention showed a greater increase in body weight after completing intervention. CONCLUSIONS CR intervention results in consistent hormone signatures regardless of dietary patterns. More changes in amylin, GLP-1 or ghrelin levels during CR are associated with poor weight maintenance after intervention, supporting that CR-induced hormone changes as biomarkers for predicting weight maintenance after intervention. TRIAL REGISTRATION Clinicaltrials.gov NCT03856762.
Collapse
Affiliation(s)
- Huibin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiaoling Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yingkai Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongrong Shen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifeng Wang
- 01life Institute, Shenzhen, Guangdong 518000, China
| | - Xiaoqiang Xu
- 01life Institute, Shenzhen, Guangdong 518000, China
| | - Mengshan Ni
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanru Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaogan Luo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
16
|
Malicka A, Ali A, MacCannell ADV, Roberts LD. Brown and beige adipose tissue-derived metabokine and lipokine inter-organ signalling in health and disease. Exp Physiol 2024. [PMID: 39591977 DOI: 10.1113/ep092008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024]
Abstract
Adipose tissue has an established endocrine function through the secretion of adipokines. However, a role for bioactive metabolites and lipids, termed metabokines and lipokines, is emerging in adipose tissue-mediated autocrine, paracrine and endocrine signalling and inter-organ communication. Traditionally seen as passive entities, metabolites are now recognized for their active roles in regulating cellular signalling and local and systemic metabolism. Distinct from white adipose tissue, specific endocrine functions have been attributed to thermogenic brown and beige adipose tissues. Brown and beige adipose tissues have been identified as sources of metabokines and lipokines, which influence diverse metabolic pathways, such as fatty acid β-oxidation, mitochondrial function and glucose homeostasis, across a range of tissues, including skeletal muscle, adipose tissue and heart. This review explores the intricate signalling mechanisms of brown and beige adipose tissue-derived metabokines and lipokines, emphasizing their roles in maintaining metabolic homeostasis and their potential dysregulation in metabolic diseases. Furthermore, we discuss the therapeutic potential of targeting these pathways, proposing that precise modulation of metabokine receptors and transporters could offer superior specificity and efficacy in comparison to conventional approaches, such as β-adrenergic signalling-stimulated activation of brown adipose tissue thermogenesis. Understanding the complex interactions between adipokines, metabokines and lipokines is essential for developing a systems-level approach to new interventions for metabolic disorders, underscoring the need for continued research in this rapidly evolving field.
Collapse
Affiliation(s)
- Anna Malicka
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Aysha Ali
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
17
|
Li L, Li R, Tian Q, Luo Y, Li R, Lin X, Ou Y, Guo T, Chen X, Pan A, Manson JE, Liu G. Effects of healthy low-carbohydrate diet and time-restricted eating on weight and gut microbiome in adults with overweight or obesity: Feeding RCT. Cell Rep Med 2024; 5:101801. [PMID: 39454570 PMCID: PMC11604488 DOI: 10.1016/j.xcrm.2024.101801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/02/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024]
Abstract
The effect of a healthy low-carbohydrate diet (HLCD) and time-restricted eating (TRE), alone or in combination, on body weight and gut microbiome beyond caloric restriction remains unclear. In this 12-week two-by-two factorial randomized trial with a 28-week follow-up among 96 participants with overweight or obesity, isocaloric-restricted feeding yields significant weight loss, ranging from 2.57 to 4.11 kg across different groups. Beyond caloric restriction, HLCD and TRE lead to additional reduction in body mass index. HLCD results in additional fat mass loss while TRE yields more lean mass loss. Additionally, HLCD leads to decreased fecal branched-chain amino acids, and TRE tends to yield an increased abundance of probiotic species involved in synthesizing short-chain fatty acids. Moreover, the effect of HLCD on reducing fat mass is sustained during the post-intervention follow-up. Overall, HLCD and TRE are effective in weight management and yield profound gut microbiome and metabolome alteration beyond caloric restriction. This study was registered at ChiCTR.org.cn (ChiCTR2200056363).
Collapse
Affiliation(s)
- Lin Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingying Tian
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaogan Luo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ruyi Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunjing Ou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Guo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environment Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
18
|
Hellberg A, Salamon D, Ujvari D, Rydén M, Hirschberg AL. Weight Changes Are Linked to Adipose Tissue Genes in Overweight Women with Polycystic Ovary Syndrome. Int J Mol Sci 2024; 25:11566. [PMID: 39519120 PMCID: PMC11547111 DOI: 10.3390/ijms252111566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Women with polycystic ovary syndrome (PCOS) have varying difficulties in achieving weight loss by lifestyle intervention, which may depend on adipose tissue metabolism. The objective was to study baseline subcutaneous adipose tissue gene expression as a prediction of weight loss by lifestyle intervention in obese/overweight women with PCOS. This is a secondary analysis of a randomized controlled trial where women with PCOS, aged 18-40 and body mass index (BMI) ≥ 27 were initially randomized to either a 4-month behavioral modification program or minimal intervention according to standard care. Baseline subcutaneous adipose tissue gene expression was related to weight change after the lifestyle intervention. A total of 55 obese/overweight women provided subcutaneous adipose samples at study entry. Weight loss was significant after behavioral modification (-2.2%, p = 0.0014), while there was no significant weight loss in the control group (-1.1%, p = 0.12). In microarray analysis of adipose samples, expression of 40 genes differed significantly between subgroups of those with the greatest weight loss or weight gain. 10 genes were involved in metabolic pathways including glutathione metabolism, gluconeogenesis, and pyruvate metabolism. Results were confirmed by real-time polymerase chain reaction (RT-PCR) in all 55 subjects. Expressions of GSTM5, ANLN, and H3C2 correlated with weight change (R = -0.41, p = 0.002; R = -0.31, p = 0.023 and R = -0.32, p = 0.016, respectively). GSTM5, involved in glutathione metabolism, was the strongest predictor of weight loss, and together with baseline waist-hip ratio (WHR) explained 31% of the variation in body weight change. This study shows that baseline subcutaneous adipose tissue genes play a role for body weight outcome in response to lifestyle intervention in overweight/obese women with PCOS.
Collapse
Affiliation(s)
- Anton Hellberg
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
| | - Daniel Salamon
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
| | - Dorina Ujvari
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
- Department of Microbiology, Tumor and Cell Biology, National Pandemic Centre, Centre for Translational Microbiome Research, Karolinska Institutet, 17165 Solna, Sweden
| | - Mikael Rydén
- Department of Medicine Huddinge (H7), Karolinska Institutet, C2-94 Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden;
| | - Angelica Lindén Hirschberg
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden; (D.S.); (D.U.); (A.L.H.)
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital Solna, 17176 Stockholm, Sweden
| |
Collapse
|
19
|
Huang J, Li Y, Chen M, Cai Z, Cai Z, Jiang Z. Comparing caloric restriction regimens for effective weight management in adults: a systematic review and network meta-analysis. Int J Behav Nutr Phys Act 2024; 21:108. [PMID: 39327619 PMCID: PMC11425986 DOI: 10.1186/s12966-024-01657-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Randomized controlled trials have confirmed the effectiveness of four prevalent caloric restriction regimens in reducing obesity-related health risks. However, there is no consensus on the optimal regimen for weight management in adults. METHODS We systematically searched PubMed, Embase, Web of Science, and Cochrane CENTRAL up to January 15, 2024, for randomized controlled trials (RCT) involving adults, evaluating the weight-loss effects of alternate day fasting (ADF), short-term fasting (STF), time-restricted eating (TRE), and continuous energy restriction (CER). The primary outcome was body weight, with secondary outcomes including BMI, fat mass, lean mass, waist circumference, fasting glucose, HOMA-IR, and adverse events. Bayesian network meta-analysis was conducted, ranking regimens using the surface under the cumulative ranking curve and the probability of being the best. Study quality was assessed using the Confidence in Network Meta-Analysis tool. RESULTS Data from 47 RCTs (representing 3363 participants) were included. ADF showed the most significant body weight loss (Mean difference (MD): -3.42; 95% Confidence interval (CI): -4.28 to -2.55), followed by TRE (MD: -2.25; 95% CI: -2.92 to -1.59). STF (MD: -1.87; 95% CI: -3.32 to -0.56) and CER (MD: -1.59; 95% CI: -2.42 to -0.79) rank third and fourth, respectively. STF lead to decline in lean mass (MD: -1.26; 95% CI: -2.16, -0.47). TRE showed benefits on fasting glucose (MD: -2.98; 95% CI: -4.7, -1.26). Subgroup analysis revealed all four caloric restriction regimens likely lead to modest weight loss after 1-3 months, with ADF ranked highest, but by 4-6 months, varying degrees of weight regain occur, particularly with CER, while interventions lasting 7-12 months may result in effective weight loss, with TRE potentially ranking first during both the 4-6 months and 7-12 months periods. ADF showing fewer and shorter-lasting physical symptoms. CONCLUSION All four included regiments were effective in reducing body weight, with ADF likely having the most significant impact. Each regimen likely leads to modest weight loss after 1-3 months, followed by weight regain by 4-6 months. However, interventions lasting 7-12 months achieve greater weight loss overall. TRIAL REGISTRATION PROSPERO: CRD42022382478.
Collapse
Affiliation(s)
- Jinming Huang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Maohua Chen
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhaolun Cai
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Cai
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Zhiyuan Jiang
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
Bozkurt O, Çamli A, Kocaadam‐Bozkurt B. Factors affecting food addiction: emotional eating, palatable eating motivations, and BMI. Food Sci Nutr 2024; 12:6841-6848. [PMID: 39554350 PMCID: PMC11561797 DOI: 10.1002/fsn3.4333] [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: 04/26/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 11/19/2024] Open
Abstract
Evaluating the factors leading to adult food addiction should shed light on potential preventive and treatment strategies for obesity and eating disorders. This research aimed to assess the relationship between food addiction, emotional eating, palatable eating motivations, and the factors that affected them. Five hundred twenty-two adults participated in this descriptive, cross-sectional study in Erzurum, Turkey. Participants completed a questionnaire that included a general information form, anthropometric measurements, Palatable Eating Motives Scale (PEMS), Yale Food Addiction Scale (YFAS), and Emotional Eater Questionnaire (EEQ). In total, 181 (34.7%) men and 341 (65.3%) women participated in the study. While 24.7% were overweight or obese, 65.7% had normal BMI (body mass index). Food addiction (FA) was determined in 18.2% of the participants. The FA group had significantly higher PEMS and EEQ scores (p < .001). The risk of FA was 3.18 times higher in women than in men (95% CI = 1.65, 6.13, p = .001). Significant positive associations between FA, BMI (OR = 1.06, 95% CI = 1.00, 1.11, p = .021), and EEQ (OR = 1.31, 95% CI = 1.23, 1.38, p = .000) were found. Emotional eating behavior and palatable eating motivations are more common in individuals with food addiction than nonfood addiction. Female gender, emotional eating, and high BMI values were determined as risk factors for food addiction.
Collapse
Affiliation(s)
- Osman Bozkurt
- Department of Nutrition and Dietetics, Faculty of Health SciencesErzurum Technical UniversityErzurumTurkey
| | - Ayşe Çamli
- Department of Nutrition and Dietetics, Faculty of Health SciencesErzurum Technical UniversityErzurumTurkey
| | - Betül Kocaadam‐Bozkurt
- Department of Nutrition and Dietetics, Faculty of Health SciencesErzurum Technical UniversityErzurumTurkey
| |
Collapse
|
21
|
Bian H, Qiao Y, Li Y, Wang Z, Zhao L, Li Z, Cheng B, Ding G. The Growth Performance and Nutrient Composition of Black Soldier Fly ( Hermetia illucens) Larvae Fed Slaughtered Bovine Blood. INSECTS 2024; 15:635. [PMID: 39336603 PMCID: PMC11432165 DOI: 10.3390/insects15090635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
The disposal of slaughterhouse blood poses significant environmental challenges due to its biological instability and high nutrient content. We used a gradient of 10% blood increments (0-100%) to feed BSFL, and the correlation between the proportion of bovine blood and the BSFL weight gain, mortality rate, fatty acid content, and amino acid content was researched. Results indicate a positive correlation between the bovine blood content and BSFL mortality, with survival rates above 95% for blood proportions below 60%. Larval weight exhibited a negative correlation as the bovine blood content increased. Nutritional analysis revealed that the crude protein content in BSFL increased proportionally with bovine blood (14.75-25.45 g/100 g), while the crude fat content decreased correspondingly (10.70-4.66 g/100 g). The sugar content remained relatively constant across groups. Fatty acid analysis showed increased levels of C16:0, C14:0, and C16:1 and decreased levels of C18:1, C18:2, and C18:3 with higher bovine blood contents. The amino acid content generally increased with higher blood proportions. This study highlights the bioconversion potential of BSFL for bovine blood and underscores the impact of protein, lipid, and sugar concentrations in feed on BSFL growth. These findings provide valuable insights for utilizing slaughterhouse waste in BSFL rearing, contributing to the development of more sustainable waste management and animal feed production methods.
Collapse
Affiliation(s)
- Hao Bian
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China; (H.B.); (Y.L.); (Z.W.); (L.Z.)
| | - Yuting Qiao
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Yantong Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China; (H.B.); (Y.L.); (Z.W.); (L.Z.)
| | - Zifan Wang
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China; (H.B.); (Y.L.); (Z.W.); (L.Z.)
| | - Lei Zhao
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China; (H.B.); (Y.L.); (Z.W.); (L.Z.)
| | - Zhiqiang Li
- School of Medicine, Northwest Minzu University, Lanzhou 730030, China;
| | - Bo Cheng
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Gongtao Ding
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China; (H.B.); (Y.L.); (Z.W.); (L.Z.)
| |
Collapse
|
22
|
Costa-E-Sousa RH, Brooks VL. The growing complexity of the control of the hypothalamic pituitary thyroid axis and brown adipose tissue by leptin. VITAMINS AND HORMONES 2024; 127:305-362. [PMID: 39864945 DOI: 10.1016/bs.vh.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
The balance between food intake and energy expenditure is precisely regulated to maintain adipose stores. Leptin, which is produced in and released from adipose in direct proportion to its size, is a major contributor to this control and initiates its homeostatic responses largely via binding to leptin receptors (LepR) in the hypothalamus. Decreases in hypothalamic LepR binding signals starvation, leading to hunger and reduced energy expenditure, whereas increases in hypothalamic LepR binding can suppress food intake and increase energy expenditure. However, large gaps persist in the specific hypothalamic sites and detailed mechanisms by which leptin increases energy expenditure, via the parallel activation of the hypothalamic pituitary thyroid (HPT) axis and brown adipose tissue (BAT). The purpose of this review is to develop a framework for the complex mechanisms and neurocircuitry. The core circuitry begins with leptin binding to receptors in the arcuate nucleus, which then sends projections to the paraventricular nucleus (to regulate the HPT axis) and the dorsomedial hypothalamus (to regulate BAT). We build on this core by layering complexities, including the intricate and unsettled regulation of arcuate proopiomelanocortin neurons by leptin and the changes that occur as the regulation of the HPT axis and BAT is engaged or modified by challenges such as starvation, hypothermia, obesity, and pregnancy.
Collapse
Affiliation(s)
- Ricardo H Costa-E-Sousa
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Virginia L Brooks
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
23
|
Dowker-Key PD, Jadi PK, Gill NB, Hubbard KN, Elshaarrawi A, Alfatlawy ND, Bettaieb A. A Closer Look into White Adipose Tissue Biology and the Molecular Regulation of Stem Cell Commitment and Differentiation. Genes (Basel) 2024; 15:1017. [PMID: 39202377 PMCID: PMC11353785 DOI: 10.3390/genes15081017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
White adipose tissue (WAT) makes up about 20-25% of total body mass in healthy individuals and is crucial for regulating various metabolic processes, including energy metabolism, endocrine function, immunity, and reproduction. In adipose tissue research, "adipogenesis" is commonly used to refer to the process of adipocyte formation, spanning from stem cell commitment to the development of mature, functional adipocytes. Although, this term should encompass a wide range of processes beyond commitment and differentiation, to also include other stages of adipose tissue development such as hypertrophy, hyperplasia, angiogenesis, macrophage infiltration, polarization, etc.… collectively, referred to herein as the adipogenic cycle. The term "differentiation", conversely, should only be used to refer to the process by which committed stem cells progress through distinct phases of subsequent differentiation. Recognizing this distinction is essential for accurately interpreting research findings on the mechanisms and stages of adipose tissue development and function. In this review, we focus on the molecular regulation of white adipose tissue development, from commitment to terminal differentiation, and examine key functional aspects of WAT that are crucial for normal physiology and systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Presley D. Dowker-Key
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Praveen Kumar Jadi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Nicholas B. Gill
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Katelin N. Hubbard
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Elshaarrawi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Naba D. Alfatlawy
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN 37996-0840, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996-0840, USA
| |
Collapse
|
24
|
Liu T, Liu Y, Yan T, Zhang B, Zhou L, Zhu W, Wang G, Kang J, Peng W, Shi L. Intermittent fasting, exercise, and dietary modification induce unique transcriptomic signatures of multiple tissues governing metabolic homeostasis during weight loss and rebound weight gain. J Nutr Biochem 2024; 130:109649. [PMID: 38642842 DOI: 10.1016/j.jnutbio.2024.109649] [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: 01/05/2024] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/22/2024]
Abstract
Obesity and its related metabolic diseases bring great challenges to public health. In-depth understanding on the efficacy of weight-loss interventions is critical for long-term weight control. Our study demonstrated the comparable efficacy of exercise (EX), intermittent fasting (IF), or the change of daily diet from an unhealthy to a normal chow (DR) for weight reduction, but largely divergently affected metabolic status and transcriptome of subcutaneous fat, scapular brown fat, skeletal muscles and liver in high-fat-high-fructose diet (HFHF) induced obese mice. EX and IF reduced systematic inflammation, improved glucose and lipid metabolism in liver and muscle, and amino acid metabolism and thermogenesis in adipose tissues. EX exhibited broad regulatory effects on TCA cycle, carbon metabolism, thermogenesis, propanoate-, fatty acid and amino acid metabolism across multiple tissues. IF prominently affected genes involved in mitophagy and autophagy in adipose tissues and core genes involved in butanoate metabolism in liver. DR, however, failed to improve metabolic homeostasis and biological dysfunctions in obese mice. Notably, by exploring potential inter-organ communication, we identified an obesity-resistant-like gene profile that were strongly correlated with HFHF induced metabolic derangements and could predict the degree of weight regain induced by the follow-up HFHF diet. Among them, 12 genes (e.g., Gdf15, Tfrc, Cdv3, Map2k4, and Nqo1) were causally associated with human metabolic traits, i.e., BMI, body fat mass, HbA1C, fasting glucose, and cholesterol. Our findings provide critical groundwork for improved understanding of the impacts of weight-loss interventions on host metabolism. The identified genes predicting weight regain may be considered regulatory targets for improving long-term weight control.
Collapse
Affiliation(s)
- Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Yuan Liu
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Tao Yan
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Baobao Zhang
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Lanqi Zhou
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Wanyu Zhu
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Guoze Wang
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jie Kang
- School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Wen Peng
- Nutrition and Health Promotion Center, Department of Public Health, Medical College, Qinghai University, Xining, Qinghai, China.
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
25
|
Anjom-Shoae J, Feinle-Bisset C, Horowitz M. Impacts of dietary animal and plant protein on weight and glycemic control in health, obesity and type 2 diabetes: friend or foe? Front Endocrinol (Lausanne) 2024; 15:1412182. [PMID: 39145315 PMCID: PMC11321983 DOI: 10.3389/fendo.2024.1412182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
It is well established that high-protein diets (i.e. ~25-30% of energy intake from protein) provide benefits for achieving weight loss, and subsequent weight maintenance, in individuals with obesity, and improve glycemic control in type 2 diabetes (T2D). These effects may be attributable to the superior satiating property of protein, at least in part, through stimulation of both gastrointestinal (GI) mechanisms by protein, involving GI hormone release and slowing of gastric emptying, as well as post-absorptive mechanisms facilitated by circulating amino acids. In contrast, there is evidence that the beneficial effects of greater protein intake on body weight and glycemia may only be sustained for 6-12 months. While both suboptimal dietary compliance and metabolic adaptation, as well as substantial limitations in the design of longer-term studies are all likely to contribute to this contradiction, the source of dietary protein (i.e. animal vs. plant) has received inappropriately little attention. This issue has been highlighted by outcomes of recent epidemiological studies indicating that long-term consumption of animal-based protein may have adverse effects in relation to the development of obesity and T2D, while plant-based protein showed either protective or neutral effects. This review examines information relating to the effects of dietary protein on appetite, energy intake and postprandial glycemia, and the relevant GI functions, as reported in acute, intermediate- and long-term studies in humans. We also evaluate knowledge relating to the relevance of the dietary protein source, specifically animal or plant, to the prevention, and management, of obesity and T2D.
Collapse
Affiliation(s)
- Javad Anjom-Shoae
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
| | - Michael Horowitz
- Adelaide Medical School and Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, SA, Australia
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
26
|
Mogna-Peláez P, Riezu-Boj JI, Milagro FI, Herrero JI, Elorz M, Benito-Boillos A, Tobaruela-Resola AL, Tur JA, Martínez JA, Abete I, Zulet MA. Inflammatory markers as diagnostic and precision nutrition tools for metabolic dysfunction-associated steatotic liver disease: Results from the Fatty Liver in Obesity trial. Clin Nutr 2024; 43:1770-1781. [PMID: 38861890 DOI: 10.1016/j.clnu.2024.05.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing public health concern. The disease is silent, and its diagnosis is often delayed. Inflammatory markers constitute an interesting tool to act as subrogate, non-invasive markers. This study aimed to evaluate the changes of inflammatory markers throughout a two-year dietary intervention in subjects presenting MASLD, to determine which of the markers are suitable to predict the disease, and act as a customizing tool for MASLD's dietary treatment. METHODS Ninety-eight subjects with MASLD and forty-five controls from the Fatty Liver in Obesity (FLiO) Study were analyzed. MASLD was diagnosed and graded by ultrasound. The MASLD subjects were randomly assigned to two different dietary strategies, the American Heart Association (AHA diet) or a dietary strategy based on the Mediterranean pattern, which was specially designed for the study (FLiO diet), and then followed for two years. Hepatic status was additionally assessed through Magnetic Resonance Imaging (MRI), elastography, and determination of transaminases. RESULTS & DISCUSSION Inflammatory markers improved throughout the intervention in the MASLD subjects and managed to reach similar levels to controls, especially at 6 and 12 months. Additionally, leptin, adiponectin, M30, and LECT2 managed to significantly diagnose the disease at all time marks of the intervention, making them candidates for subrogate non-invasive markers of the disease. Moreover, baseline chemerin, leptin, LECT2, and M65 were used to build a predictive score to achieve greater weight loss, and therefore, which strategy could be more useful for MASLD 's treatment. The predictive score was significantly able assign a specific diet to 55% of the study participants, meaning that the remaining 45% could achieve the same amount of weight loss following either diet equally. CONCLUSION Inflammatory markers constitute a potential non-invasive tool to be used in MASLD screening and could also constitute an interesting tool for MASLD's treatment customization, being able to predict the effectiveness of a dietary strategy based on the initial inflammatory state of each subject. TRIAL REGISTRATION www. CLINICALTRIALS gov (NCT03183193).
Collapse
Affiliation(s)
- Paola Mogna-Peláez
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
| | - José I Riezu-Boj
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Fermin I Milagro
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José I Herrero
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; Liver Unit, Clínica Universidad de Navarra, 31008 Pamplona, Spain; Biomedical Research Centre Network in Hepatic and Digestive Diseases (CIBERehd), 28029 Madrid, Spain
| | - Mariana Elorz
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; Department of Radiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Alberto Benito-Boillos
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; Department of Radiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ana Luz Tobaruela-Resola
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain
| | - Josep A Tur
- Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain; Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands-IUNICS & IDISBA, 07122 Palma, Spain
| | - J Alfredo Martínez
- Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain; Precision Nutrition and Cardiovascular Health Program, IMDEA Food, CEI UAM + CSIC, 28049 Madrid Spain
| | - Itziar Abete
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - M Angeles Zulet
- Department of Nutrition, Food Sciences and Physiology and Centre for Nutrition Research, Faculty of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; Biomedical Research Centre Network in Physiopathology of Obesity and Nutrition (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
27
|
Chao X, Guo L, Ye C, Liu A, Wang X, Ye M, Fan Z, Luan K, Chen J, Zhang C, Liu M, Zhou B, Zhang X, Li Z, Luo Q. ALKBH5 regulates chicken adipogenesis by mediating LCAT mRNA stability depending on m 6A modification. BMC Genomics 2024; 25:634. [PMID: 38918701 PMCID: PMC11197345 DOI: 10.1186/s12864-024-10537-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Previous studies have demonstrated the role of N6-methyladenosine (m6A) RNA methylation in various biological processes, our research is the first to elucidate its specific impact on LCAT mRNA stability and adipogenesis in poultry. RESULTS The 6 100-day-old female chickens were categorized into high (n = 3) and low-fat chickens (n = 3) based on their abdominal fat ratios, and their abdominal fat tissues were processed for MeRIP-seq and RNA-seq. An integrated analysis of MeRIP-seq and RNA-seq omics data revealed 16 differentially expressed genes associated with to differential m6A modifications. Among them, ELOVL fatty acid elongase 2 (ELOVL2), pyruvate dehydrogenase kinase 4 (PDK4), fatty acid binding protein 9 (PMP2), fatty acid binding protein 1 (FABP1), lysosomal associated membrane protein 3 (LAMP3), lecithin-cholesterol acyltransferase (LCAT) and solute carrier family 2 member 1 (SLC2A1) have ever been reported to be associated with adipogenesis. Interestingly, LCAT was down-regulated and expressed along with decreased levels of mRNA methylation methylation in the low-fat group. Mechanistically, the highly expressed ALKBH5 gene regulates LCAT RNA demethylation and affects LCAT mRNA stability. In addition, LCAT inhibits preadipocyte proliferation and promotes preadipocyte differentiation, and plays a key role in adipogenesis. CONCLUSIONS In conclusion, ALKBH5 mediates RNA stability of LCAT through demethylation and affects chicken adipogenesis. This study provides a theoretical basis for further understanding of RNA methylation regulation in chicken adipogenesis.
Collapse
Affiliation(s)
- Xiaohuan Chao
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lijin Guo
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Chutian Ye
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Aijun Liu
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaomeng Wang
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mao Ye
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhexia Fan
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Kang Luan
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiahao Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chunlei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Manqing Liu
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Bo Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiquan Zhang
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenhui Li
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China.
- College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Qingbin Luo
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China.
- College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
28
|
Lecoutre S, Rebière C, Marcelin G, Clément K. How does bariatric surgery remodel adipose tissue? ANNALES D'ENDOCRINOLOGIE 2024; 85:175-178. [PMID: 38871506 DOI: 10.1016/j.ando.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
This lecture delves into the pivotal role of adipose tissue in obesity and its response to weight loss, particularly via bariatric surgery. Adipose tissue, responsible for storing excess energy, undergoes significant changes during obesity, marked by inflammation and fibrosis. Bariatric surgery, serving as a model, allow the exploration of adipose tissue remodeling post-weight loss, inducing metabolic and fibro-inflammatory shifts. Despite successful weight loss, inflammation and fibrosis persist, as evidenced by changes in immune cells, altered cytokine profiles and the accumulation of extracellular matrix (ECM). Unfortunately, these lingering effects impair the normal adipose tissue function. In this context, adipose progenitors, an heterogenous resident population of mesenchymal stromal cells, display functions important to fibrosis development, capable of differentiating into myofibroblasts and contributing to ECM deposition. Particularly, a distinct subpopulation of adipose progenitors with high CD9 expression (CD9high) is associated with fibrosis and insulin resistance in human obesity. The persistence of fibrosis post-weight loss poses challenges, correlating with metabolic dysfunction despite improved glucose tolerance. A comprehensive understanding of the mechanisms driving adipose tissue remodeling and fibrosis post-weight loss is imperative for the development of effective treatments for obesity. The intricate interplay between adipose tissue, inflammation, and fibrosis underscores the necessity for further in-depth research to elucidate these mechanisms and formulate targeted therapies for obesity-related complications.
Collapse
Affiliation(s)
- Simon Lecoutre
- Research Unit: Nutrition and Obesities; Systemic Approaches, NutriOmics, Inserm, Sorbonne université, Paris, France
| | - Clémentine Rebière
- Research Unit: Nutrition and Obesities; Systemic Approaches, NutriOmics, Inserm, Sorbonne université, Paris, France; Nutrition Department, Pitié-Salpêtrière Hospital, Paris Public Hospitals, Paris, France
| | - Geneviève Marcelin
- Nutrition Department, Pitié-Salpêtrière Hospital, Paris Public Hospitals, Paris, France
| | - Karine Clément
- Research Unit: Nutrition and Obesities; Systemic Approaches, NutriOmics, Inserm, Sorbonne université, Paris, France.
| |
Collapse
|
29
|
Boyajian JL, Islam P, Abosalha A, Schaly S, Thareja R, Kassab A, Arora K, Santos M, Shum-Tim C, Prakash S. Probiotics, prebiotics, synbiotics and other microbiome-based innovative therapeutics to mitigate obesity and enhance longevity via the gut-brain axis. MICROBIOME RESEARCH REPORTS 2024; 3:29. [PMID: 39421246 PMCID: PMC11480732 DOI: 10.20517/mrr.2024.05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 05/11/2024] [Indexed: 10/19/2024]
Abstract
The global prevalence of obesity currently exceeds 1 billion people and is accompanied by an increase in the aging population. Obesity and aging share many hallmarks and are leading risk factors for cardiometabolic disease and premature death. Current anti-obesity and pro-longevity pharmacotherapies are limited by side effects, warranting the development of novel therapies. The gut microbiota plays a major role in human health and disease, with a dysbiotic composition evident in obese and aged individuals. The bidirectional communication system between the gut and the central nervous system, known as the gut-brain axis, may link obesity to unhealthy aging. Modulating the gut with microbiome-targeted therapies, such as biotics, is a novel strategy to treat and/or manage obesity and promote longevity. Biotics represent material derived from living or once-living organisms, many of which have therapeutic effects. Pre-, pro-, syn- and post-biotics may beneficially modulate gut microbial composition and function to improve obesity and the aging process. However, the investigation of biotics as next-generation therapeutics has only just begun. Further research is needed to identify therapeutic biotics and understand their mechanisms of action. Investigating the function of the gut-brain axis in obesity and aging may lead to novel therapeutic strategies for obese, aged and comorbid (e.g., sarcopenic obese) patient populations. This review discusses the interrelationship between obesity and aging, with a particular emphasis on the gut microbiome, and presents biotics as novel therapeutic agents for obesity, aging and related disease states.
Collapse
Affiliation(s)
- Jacqueline L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Ahmed Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta 31111, Egypt
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Amal Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Karan Arora
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Madison Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Cedrique Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal H3A 2B4, Quebec, Canada
| |
Collapse
|
30
|
Minari TP, Manzano CF, Tácito LHB, Yugar LBT, Sedenho-Prado LG, Rubio TDA, Pires AC, Vilela-Martin JF, Cosenso-Martin LN, Moreno H, Yugar-Toledo JC. The Impact of a Nutritional Intervention on Glycemic Control and Cardiovascular Risk Markers in Type 2 Diabetes. Nutrients 2024; 16:1378. [PMID: 38732624 PMCID: PMC11085322 DOI: 10.3390/nu16091378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
INTRODUCTION Nutritional management plays a crucial role in treating patients with type 2 diabetes (T2D), working to prevent and control the progression of chronic non-communicable diseases. OBJECTIVES To evaluate the effects of individualized nutritional interventions on weight, body mass index (BMI), waist circumference (WC), waist-to-hip ratio (WHR), fasting blood glucose (FBG), hemoglobin A1c (HbA1c), total cholesterol (TC), LDL cholesterol (LDL-C), HDL cholesterol (HDL-C), triglycerides (TGs), systolic blood pressure (SBP), diastolic blood pressure (DBP), and heart rate (HR)} over 12 months and subsequently at follow-up (15 months). METHODS This longitudinal experimental study (without randomization and blinding) enrolled 84 sedentary participants with T2D (both sexes, aged 18-80 years). They were divided into a control group of 40 participants who received only medical consultations, and an intervention group of 44 participants who received the same medical care along with a nutritional assessment. Consultations occurred quarterly from August 2020 to November 2022 (first-twelfth month), with six to nine patients per session. Subsequently, a follow-up was conducted from December 2022 to November 2023, during which the intervention group had only medical care (during the 12th-15th months). Personalized dietary planning was inspired by the Mediterranean/DASH diets adapted to Brazilian foods and socioeconomic cultures. STATISTICAL ANALYSIS Normal variables were compared between groups for each time point and also within each group across different time points using a two-way ANOVA (repeated measures for intragroup) followed by the Šídák post hoc test. Non-normal variables were compared between groups for each time point using Kruskal-Wallis followed by the Dunn post hoc test, and within each group across different time points using Friedman followed by the Dunn post hoc test. Data with a Gaussian distribution were presented as mean ± standard deviation (SD), and data with a non-Gaussian distribution were presented as median ± interquartile range (IQR). For all cases, α < 0.05 and p < 0.05 were adopted. RESULTS In the intervention group, significant reductions were observed between the first and twelfth month for all parameters (p < 0.05), (except for TC), along with an increase in HDL-C (p = 0.0105). Conversely, in the control group, there was a significant increase in HbA1c, weight, BMI, FBG, and WHR (p < 0.05) between the first and twelfth months. Regarding the comparison between groups, there was a significant difference for all analyzed parameters (p < 0.05) from the first to the twelfth month. In the follow-up, differences were also observed (p < 0.05), except for BMI (p > 0.05). CONCLUSION The individualized nutritional intervention improved eating habits, anthropometric, biochemical, and cardiovascular markers in T2D over 12 months, with sustained results during follow-up. The dietary plan inspired by the Mediterranean and DASH diets demonstrated good adaptation to the Brazilian food culture and the patients' socioeconomic contexts. Consistent monitoring and personalized nutritional management are essential for optimizing long-term outcomes. However, more clinical trials are necessary in order to optimize the level of evidence for longitudinal interventions.
Collapse
Affiliation(s)
- Tatiana Palotta Minari
- Department of Hypertension, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | - Carolina Freitas Manzano
- Department of Hypertension, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | - Lúcia Helena Bonalume Tácito
- Department of Endocrinology, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | | | | | - Tatiane de Azevedo Rubio
- Cardiovascular Pharmacology & Hypertension Laboratory, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas 13083-887, SP, Brazil
| | - Antônio Carlos Pires
- Department of Endocrinology, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | - José Fernando Vilela-Martin
- Department of Hypertension, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | - Luciana Neves Cosenso-Martin
- Department of Endocrinology, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| | - Heitor Moreno
- Cardiovascular Pharmacology & Hypertension Laboratory, School of Medical Sciences, State University of Campinas (UNICAMP), Campinas 13083-887, SP, Brazil
| | - Juan Carlos Yugar-Toledo
- Department of Hypertension, State Faculty of Medicine of São José do Rio Preto (FAMERP), São José do Rio Preto 15090-000, SP, Brazil
| |
Collapse
|
31
|
Yu Y, Ma Q, Groth S. Prepregnancy weight loss and maternal metabolic and inflammatory biomarkers during pregnancy: An analysis of National Health and Nutrition Examination Survey. J Obstet Gynaecol Res 2024; 50:809-820. [PMID: 38369640 DOI: 10.1111/jog.15904] [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: 07/29/2023] [Accepted: 01/31/2024] [Indexed: 02/20/2024]
Abstract
AIM Women with overweight or obesity are recommended to lose weight before conception to optimize pregnancy outcomes. However, the obstetrical implications of prepregnancy weight loss have been minimally examined. The objective of this study was to investigate the association between prepregnancy weight loss and maternal metabolic and inflammatory profiles during a subsequent pregnancy. METHODS This study was a retrospective analysis of National Health and Nutrition Examination Survey data (2003-2018). Participants were women who were pregnant at the time of assessment. Prepregnancy weight loss was described as percent weight change based on self-reported baseline (1 year before pregnancy) and prepregnancy weight. Metabolic (e.g., blood pressure [BP]) and inflammatory biomarkers (i.e., high-sensitivity C-reactive protein [hs-CRP]) were determined by standard medical tests. Statistical analyses included linear regressions with appropriate imputation, weighting, and variance estimation techniques. RESULTS Participants (N = 236) reported a mean percent weight loss of 4.6% (standard error [SE] = 0.3%) during the year before pregnancy. Regression models showed that prepregnancy weight loss was inversely associated with levels of total cholesterol (β = -1.24, p = 0.01), low-density lipoprotein-cholesterol (β = -0.79, p < 0.01), and high-density lipoprotein-cholesterol (β = -0.18, p < 0.01). The effect of prepregnancy weight loss on BP, insulin sensitivity, and hs-CRP was not significant, although there was a trend toward higher levels of diastolic BP (β = 0.24, p = 0.07) and hs-CRP (β = 0.10, p = 0.08). CONCLUSIONS This study found favorable changes in lipid profiles following prepregnancy weight loss. Due to limitations such as a relatively small sample size, self-reported weight measures, and missing data on several outcome variables, future studies are needed to confirm study findings.
Collapse
Affiliation(s)
- Yang Yu
- School of Nursing, University of Rochester, Rochester, New York, USA
| | - Qianheng Ma
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA
| | - Susan Groth
- School of Nursing, University of Rochester, Rochester, New York, USA
| |
Collapse
|
32
|
Lee J, Jung JH, Kang DW, Kim MH, Lim DJ, Kwon HS, Lee JM, Chang SA, Han K, Lee SH. Body Weight Variability and Risk of Suicide Mortality: A Nationwide Population-Based Study. Depress Anxiety 2024; 2024:7670729. [PMID: 40226695 PMCID: PMC11921691 DOI: 10.1155/2024/7670729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/19/2024] [Accepted: 04/18/2024] [Indexed: 04/15/2025] Open
Abstract
Background Suicide is a pressing global health concern, and identifying its risk factors is crucial for prevention. Body weight variability (BWV) has been increasingly recognized as a potential factor impacting physical and mental health outcomes. We aimed to explore the relationship between BWV and the risk of suicide mortality using a nationally representative database. Methods This population-based cohort study used data from the Korean National Health Insurance Database and included a total of 1,983,701 subjects. BWV was assessed using at least three health examination datasets and validated variability indices (variability independent of the mean (VIM), average successive variability, and coefficient of variation), and patients were divided into BWV quartiles (Q1-Q4). The primary endpoint was suicide-related death. Results During a median of 11.3 years of follow-up, 5,883 suicide deaths occurred. A higher baseline body weight was associated with a lower risk of suicide. However, greater BWV (VIM) was associated with a significantly greater risk of suicide (adjusted hazard ratio [95% confidence interval], 1.35 [1.26-1.45] in the Q4 group), even after adjusting for baseline body mass index (BMI). Similar results were observed regardless of obesity or BMI category. Consistent findings were observed when using different variability indices. Subgroup analyses according to sex, age, diabetes, and depression also supported these findings. Conclusion Our study highlights the importance of considering BWV as a potential risk factor for suicide.
Collapse
Affiliation(s)
- Jeongmin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Hyung Jung
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Dong Woo Kang
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min-Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong-Jun Lim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk-Sang Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Min Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Ah Chang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Seung-Hwan Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
33
|
Yoon DJ, Zhang J, Zapata RC, Ulivieri M, Libster AM, McMurray MS, Osborn O, Dulawa SC. The attenuation of activity-based anorexia by obese adipose tissue transplant is AgRP neuron-dependent. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590824. [PMID: 38712190 PMCID: PMC11071374 DOI: 10.1101/2024.04.23.590824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Anorexia nervosa (AN) is an eating disorder observed primarily in girls and women, and is characterized by a low body mass index, hypophagia, and hyperactivity. The activity-based anorexia (ABA) paradigm models aspects of AN, and refers to the progressive weight loss, hypophagia, and hyperactivity developed by rodents exposed to time-restricted feeding and running wheel access. Recent studies identified white adipose tissue (WAT) as a primary location of the 'metabolic memory' of prior obesity, and implicated WAT-derived signals as drivers of recidivism to obesity following weight loss. Here, we tested whether an obese WAT transplant could attenuate ABA-induced weight loss in normal female mice. Recipient mice received a WAT transplant harvested from normal chow-fed, or HFD-fed obese mice; obese fat recipient (OFR) and control fat recipient (CFR) mice were then tested for ABA. During ABA, OFR mice survived longer than CFR mice, defined as maintaining 75% of their initial body weight. Next, we tested whether agouti-related peptide (AgRP) neurons, which regulate feeding behavior and metabolic sensing, mediate this effect of obese WAT transplant. CFR and OFR mice received either control or neonatal AgRP ablation, and were assessed for ABA. OFR intact mice maintained higher body weights longer than CFR intact mice, and this effect was abolished by neonatal AgRP ablation; further, ablation reduced survival in OFR, but not CFR mice. In summary, obese WAT transplant communicates with AgRP neurons to increase body weight maintenance during ABA. These findings encourage the examination of obese WAT-derived factors as potential treatments for AN.
Collapse
Affiliation(s)
- Dongmin J. Yoon
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jie Zhang
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Rizaldy C. Zapata
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Martina Ulivieri
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Avraham M. Libster
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | | | - Olivia Osborn
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Stephanie C. Dulawa
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
34
|
Jensterle M, Ferjan S, Janez A. The maintenance of long-term weight loss after semaglutide withdrawal in obese women with PCOS treated with metformin: a 2-year observational study. Front Endocrinol (Lausanne) 2024; 15:1366940. [PMID: 38665260 PMCID: PMC11043580 DOI: 10.3389/fendo.2024.1366940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Background Withdrawal of semaglutide is frequently followed by weight regain due to compensatory biological changes that prevent the maintenance of long-term weight loss. There are some studies implying that metformin might attenuate weight regain. The weight trajectory after discontinuation of short-term semaglutide treatment in obese women with PCOS who continued metformin treatment has not yet been evaluated. Aims We explored changes in body weight, cardiometabolic and endocrine parameters in obese women with PCOS who continued treatment with metformin 2 years after discontinuation of short-term intervention with semaglutide. Methods 25 women with PCOS and obesity, aged 33.7 ± 5.3 years (mean ± SD), were treated with once-weekly subcutaneous semaglutide 1.0 mg as an adjunct to metformin 2000 mg/day and lifestyle intervention for 16 weeks. At week 16, semaglutide was discontinued. Treatment with metformin 2000 mg/day and promotion of lifestyle intervention were continued during the 2-year follow-up period. Weight change, cardiometabolic, and endocrine parameters were assessed 2 years after semaglutide discontinuation. Results During semaglutide treatment phase, weight decreased from 101 (90-106.8) kg to 92 (83.3-100.8) kg. Two years after semaglutide withdrawal, weight was 95 (77-104) kg. The net weight loss 2 years after discontinuation of semaglutide remained significant when compared to baseline (p=0.003). At the end of the study, 21 out of 25 subjects had lower body weight compared to baseline. Improvements in cardiometabolic parameters including decrease in total and LDL cholesterol, fasting glucose, and glucose after OGTT that had been seen during semaglutide-treatment phase reverted towards baseline two years after semaglutide cessation. Free testosterone levels significantly decreased during semaglutide treatment from 6.16 (4.07-9.71) to 4.12 (2.98-6.93) nmol/l, (p= 0.012) and did not significantly deteriorate after semaglutide discontinuation. Conclusion Two years after semaglutide withdrawal, women with PCOS who continued with metformin regained about one-third of the semaglutide-induced weight loss. At the end of the follow up, 84% of women had a lower body weight than at baseline.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Simona Ferjan
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Andrej Janez
- Department of Endocrinology, Diabetes and Metabolic Diseases, Division of Internal Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
35
|
Tolstik TV, Kirichenko TV, Markin AM, Bogatyreva AI, Markina YV, Kiseleva DG, Shaposhnikova NN, Starodubova AV, Orekhov AN. The association of TNF-alpha secretion and mtDNA copy number in CD14 + monocytes of patients with obesity and CHD. Front Mol Biosci 2024; 11:1362955. [PMID: 38572445 PMCID: PMC10987863 DOI: 10.3389/fmolb.2024.1362955] [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: 12/29/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
Introduction Mitochondrial dysfunction may be one of the causes of inflammatory activation of monocytes and macrophages, which leads to excessive secretion of inflammatory mediators and the development of chronic inflammation. Aims The study was aimed to evaluate the secretion of inflammatory cytokine tumor necrosis factor-α (TNF-α) in the primary culture of monocytes, and to analyze its relationship with the number of mitochondrial DNA (mtDNA) copies in the blood of patients with coronary heart disease (CHD) and obesity. Materials and methods 108 patients with obesity and concomitant CHD and a control group of 25 participants were included in the study. CD14+ monocytes were isolated by a standard method in a ficoll-urographin gradient, followed by separation using magnetic particles. The number of mtDNA copies was estimated using qPCR. Results It was demonstrated that the number of mtDNA copies was significantly increased in groups of patients with CHD and obesity + CHD in comparison with control group. mtDNA copy number positively correlated with basal and LPS-stimulated TNF-α secretion, the most significant correlation was found in the group of patients with CHD and obesity. Conclusion Thus, the change in mtDNA copy number in CD14+ monocytes which indicates the presence of mitochondrial dysfunction, confirm the direct involvement of mitochondria in the violation of the inflammatory response of monocytes revealed in this study as an increased secretion of inflammatory cytokine TNF-α.
Collapse
Affiliation(s)
| | - Tatiana V. Kirichenko
- Petrovsky National Research Center of Surgery, Moscow, Russia
- Chazov National Medical Research Center of Cardiology, Moscow, Russia
| | - Alexander M. Markin
- Petrovsky National Research Center of Surgery, Moscow, Russia
- Рeoples’ Friendship University of Russia Named After Patrice Lumumba (RUDN University), Moscow, Russia
| | | | | | - Diana G. Kiseleva
- Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | | | | |
Collapse
|
36
|
Li H, Zhang L, Li J, Wu Q, Qian L, He J, Ni Y, Kovatcheva-Datchary P, Yuan R, Liu S, Shen L, Zhang M, Sheng B, Li P, Kang K, Wu L, Fang Q, Long X, Wang X, Li Y, Ye Y, Ye J, Bao Y, Zhao Y, Xu G, Liu X, Panagiotou G, Xu A, Jia W. Resistant starch intake facilitates weight loss in humans by reshaping the gut microbiota. Nat Metab 2024; 6:578-597. [PMID: 38409604 DOI: 10.1038/s42255-024-00988-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 01/17/2024] [Indexed: 02/28/2024]
Abstract
Emerging evidence suggests that modulation of gut microbiota by dietary fibre may offer solutions for metabolic disorders. In a randomized placebo-controlled crossover design trial (ChiCTR-TTRCC-13003333) in 37 participants with overweight or obesity, we test whether resistant starch (RS) as a dietary supplement influences obesity-related outcomes. Here, we show that RS supplementation for 8 weeks can help to achieve weight loss (mean -2.8 kg) and improve insulin resistance in individuals with excess body weight. The benefits of RS are associated with changes in gut microbiota composition. Supplementation with Bifidobacterium adolescentis, a species that is markedly associated with the alleviation of obesity in the study participants, protects male mice from diet-induced obesity. Mechanistically, the RS-induced changes in the gut microbiota alter the bile acid profile, reduce inflammation by restoring the intestinal barrier and inhibit lipid absorption. We demonstrate that RS can facilitate weight loss at least partially through B. adolescentis and that the gut microbiota is essential for the action of RS.
Collapse
Affiliation(s)
- Huating Li
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong S.A.R., China.
- Department of Medicine, The University of Hong Kong, Hong Kong S.A.R., China.
| | - Lei Zhang
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Li
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong S.A.R., China
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Qian Wu
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingling Qian
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junsheng He
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong S.A.R., China
| | - Yueqiong Ni
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | | | - Rui Yuan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Shuangbo Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Li Shen
- Department of Clinical Nutrition, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingliang Zhang
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Li
- Department of Computing, The Hong Kong Polytechnic University, Hong Kong S.A.R., China
- School of Design, The Hong Kong Polytechnic University, Hong Kong S.A.R., China
| | - Kang Kang
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany
| | - Liang Wu
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qichen Fang
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxue Long
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yanli Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yaorui Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Jianping Ye
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Metabolic Disease Research Center, Zhengzhou University Affiliated Zhengzhou Central Hospital, Zhengzhou, China
| | - Yuqian Bao
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueliang Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, Germany.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong S.A.R., China.
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong S.A.R., China.
- Department of Medicine, The University of Hong Kong, Hong Kong S.A.R., China.
| | - Weiping Jia
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
37
|
Reinisch I, Michenthaler H, Sulaj A, Moyschewitz E, Krstic J, Galhuber M, Xu R, Riahi Z, Wang T, Vujic N, Amor M, Zenezini Chiozzi R, Wabitsch M, Kolb D, Georgiadi A, Glawitsch L, Heitzer E, Schulz TJ, Schupp M, Sun W, Dong H, Ghosh A, Hoffmann A, Kratky D, Hinte LC, von Meyenn F, Heck AJR, Blüher M, Herzig S, Wolfrum C, Prokesch A. Adipocyte p53 coordinates the response to intermittent fasting by regulating adipose tissue immune cell landscape. Nat Commun 2024; 15:1391. [PMID: 38360943 PMCID: PMC10869344 DOI: 10.1038/s41467-024-45724-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/02/2024] [Indexed: 02/17/2024] Open
Abstract
In obesity, sustained adipose tissue (AT) inflammation constitutes a cellular memory that limits the effectiveness of weight loss interventions. Yet, the impact of fasting regimens on the regulation of AT immune infiltration is still elusive. Here we show that intermittent fasting (IF) exacerbates the lipid-associated macrophage (LAM) inflammatory phenotype of visceral AT in obese mice. Importantly, this increase in LAM abundance is strongly p53 dependent and partly mediated by p53-driven adipocyte apoptosis. Adipocyte-specific deletion of p53 prevents LAM accumulation during IF, increases the catabolic state of adipocytes, and enhances systemic metabolic flexibility and insulin sensitivity. Finally, in cohorts of obese/diabetic patients, we describe a p53 polymorphism that links to efficacy of a fasting-mimicking diet and that the expression of p53 and TREM2 in AT negatively correlates with maintaining weight loss after bariatric surgery. Overall, our results demonstrate that p53 signalling in adipocytes dictates LAM accumulation in AT under IF and modulates fasting effectiveness in mice and humans.
Collapse
Affiliation(s)
- Isabel Reinisch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Helene Michenthaler
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Alba Sulaj
- Institute for Diabetes and Cancer, Helmholtz Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Endocrinology, Diabetology, Metabolism and Clinical Chemistry (Internal Medicine 1), Heidelberg University Hospital, Heidelberg, Germany
| | - Elisabeth Moyschewitz
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Jelena Krstic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Markus Galhuber
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Ruonan Xu
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Zina Riahi
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Tongtong Wang
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Nemanja Vujic
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Melina Amor
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Riccardo Zenezini Chiozzi
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Dagmar Kolb
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Core Facility Ultrastructure Analysis, Medical University of Graz, Graz, Austria
| | - Anastasia Georgiadi
- Institute for Diabetes and Cancer, Helmholtz Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lisa Glawitsch
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Michael Schupp
- Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wenfei Sun
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Hua Dong
- Stem Cell Biology and Regenerative Medicine Institute, University of Stanford, Stanford, CA, USA
| | - Adhideb Ghosh
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
- Functional Genomics Center Zurich, Eidgenössische Technische Hochschule Zürich (ETH), Zurich, Switzerland
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Laura C Hinte
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Ferdinand von Meyenn
- Laboratory of Nutrition and Metabolic Epigenetics, Institute for Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Endocrinology, Diabetology, Metabolism and Clinical Chemistry (Internal Medicine 1), Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Wolfrum
- Institute of Food Nutrition and Health, Department of Health Sciences and Technology, Eidgenössische Technische Hochschule Zürich (ETH), Schwerzenbach, Switzerland
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
38
|
Wang MN, Zhai MX, Wang YT, Dai QF, Liu L, Zhao LP, Xia QY, Li S, Li B. Mechanism of Acupuncture in Treating Obesity: Advances and Prospects. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1-33. [PMID: 38351701 DOI: 10.1142/s0192415x24500010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Obesity is a common metabolic syndrome that causes a significant burden on individuals and society. Conventional therapies include lifestyle interventions, bariatric surgery, and pharmacological therapies, which are not effective and have a high risk of adverse events. Acupuncture is an effective alternative for obesity, it modulates the hypothalamus, sympathetic activity and parasympathetic activity, obesity-related hormones (leptin, ghrelin, insulin, and CCK), the brain-gut axis, inflammatory status, adipose tissue browning, muscle blood flow, hypoxia, and reactive oxygen species (ROS) to influence metabolism, eating behavior, motivation, cognition, and the reward system. However, hypothalamic regulation by acupuncture should be further demonstrated in human studies using novel techniques, such as functional MRI (fMRI), positron emission tomography (PET), electroencephalogram (EEG), and magnetoencephalography (MEG). Moreover, a longer follow-up phase of clinical trials is required to detect the long-term effects of acupuncture. Also, future studies should investigate the optimal acupuncture therapeutic option for obesity. This review aims to consolidate the recent improvements in the mechanism of acupuncture for obesity as well as discuss the future research prospects and potential of acupuncture for obesity.
Collapse
Affiliation(s)
- Mi-Na Wang
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, P. R. China
- School of Traditional Chinese Medicine, School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, P. R. China
| | - Miao-Xin Zhai
- Yinghai Hospital, Daxing District, Beijing 100163, P. R. China
| | - Yi-Tong Wang
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, P. R. China
- School of Traditional Chinese Medicine, School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, P. R. China
| | - Qiu-Fu Dai
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, P. R. China
| | - Lu Liu
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, P. R. China
| | - Luo-Peng Zhao
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, P. R. China
| | - Qiu-Yu Xia
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, P. R. China
| | - Shen Li
- Department of Emergency, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, P. R. China
| | - Bin Li
- Department of Acupuncture and Moxibustion, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing 100010, P. R. China
| |
Collapse
|
39
|
Andruszko A, Szydłowski J, Grabarek BO, Mazur K, Sirek T, Ossowski P, Kozikowski M, Kaminiów K, Zybek-Kocik A, Banaszewski J. Impact of Nutritional Status of Patients with Head and Neck Squamous Cell Carcinoma on the Expression Profile of Ghrelin, Irisin, and Titin. Cancers (Basel) 2024; 16:437. [PMID: 38275878 PMCID: PMC10814803 DOI: 10.3390/cancers16020437] [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: 12/08/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
The goal of this paper was the evaluation of the changes in the expression profile of irisin, ghrelin, and titin in the carcinoma tissue and in the blood of patients with head and neck squamous cell carcinoma (HNSCC), including determining the profile of their expression in relation to patient nutrition. The study included 56 patients with diagnosed squamous cell carcinoma of HNSCC in the T3 and T4 stages of the disease. Healthy control tissue specimens were collected from an area 10 mm outside the histologically negative margin. In turn, the blood and serum from the control group came from healthy volunteers treated for non-oncologic reasons (n = 70). The molecular analysis allowed us to determine the profile of irisin, ghrelin, and titin methylation, evaluate their expression on the level of mRNA (quantitative Reverse Transcription Polymerase Chain Reaction; qRT-PCR) and protein (Enzyme-Linked Immunosorbent Assay Reaction; ELISA) in the carcinoma tissue and the margin of healthy tissue, as well as in serum of patients in the study and control groups. At the start of our observations, a Body Mass Index (BMI) < 18.5 was noted in 42 of the patients, while six months after the treatment a BMI < 18.5 was noted in 29 patients. We also noted a decrease in the expression of irisin, ghrelin, and titin both on the level of mRNA and protein, as well as a potential regulation of their expression via DNA methylation. There is no convincing evidence that the proteins assayed in the present work are specific with regard to HNSSC.
Collapse
Affiliation(s)
- Agata Andruszko
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Jarosław Szydłowski
- Department of Pediatric Otolaryngology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Beniamin Oskar Grabarek
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dąbrowa Górnicza, Poland; (B.O.G.); (P.O.); (K.K.)
- Gyncentrum, Laboratory of Molecular Biology and Virology, 40-851 Katowice, Poland
| | - Katarzyna Mazur
- Faculty of Health Sciences, The Higher School of Strategic Planning in Dąbrowa Górnicza, 41-300 Dabrowa Gornicza, Poland;
| | - Tomasz Sirek
- Department of Plastic Surgery, Faculty of Medicine, Academia of Silesia, 40-555 Katowice, Poland;
- Department of Plastic and Reconstructive Surgery, Hospital for Minimally Invasive and Reconstructive Surgery, 43-316 Bielsko-Biała, Poland
| | - Piotr Ossowski
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dąbrowa Górnicza, Poland; (B.O.G.); (P.O.); (K.K.)
| | - Mieszko Kozikowski
- Faculty of Medicine, Uczelnia Medyczna im. Marii Skłodowskiej-Curie, 00-136 Warszawa, Poland;
| | - Konrad Kaminiów
- Department of Medical and Health Sciences, Collegium Medicum, WSB University, 41-300 Dąbrowa Górnicza, Poland; (B.O.G.); (P.O.); (K.K.)
| | - Ariadna Zybek-Kocik
- Department of Metabolism Endocrinology and Internal Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Jacek Banaszewski
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| |
Collapse
|
40
|
Coutinho W, Halpern B. Pharmacotherapy for obesity: moving towards efficacy improvement. Diabetol Metab Syndr 2024; 16:6. [PMID: 38172940 PMCID: PMC10763391 DOI: 10.1186/s13098-023-01233-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/25/2023] [Indexed: 01/05/2024] Open
Abstract
Obesity is a chronic, recurring, progressive disease and a major public health problem associated with several other diseases that lead to disability, morbidity, and mortality. The prevalence of obesity has increased at pandemic levels, along with increasing weight-related comorbidities and deaths worldwide. Lifestyle interventions alone provide clinically significant long-term weight loss in only a small proportion of individuals, and bariatric surgery is not suitable or desirable for all patients. Historically, anti-obesity medications achieved a mean efficacy with weight loss between 5 and 10%, which significantly impacted several comorbidities and risk factors, but the average efficacy of these medications remained lower than that expected by both patients and health care professionals and eventually curbed long-term use. Moreover, there is no direct evidence on the impact of anti-obesity medications on cardiovascular outcomes. Semaglutide is a newer anti-obesity medication that changes the overall landscape, as phase 3 studies show a mean weight loss near the 15% threshold and significant proportions of patients with a weight loss of greater than 20%. In this review, we focus on the currently available anti-obesity medications, discuss the results of semaglutide, and present perspectives on the future of obesity treatment after semaglutide.
Collapse
Affiliation(s)
- Walmir Coutinho
- State Institute of Diabetes and Endocrinology, Rua Moncorvo Filho, 90, Rio de Janeiro, RJ, 20211-340, Brazil.
- Department of Medicine, Pontifical Catholic University of Rio de Janeiro, Rua Marquês de São Vicente, 225, Gávea, Rio de Janeiro, RJ, 22541-041, Brazil.
| | - Bruno Halpern
- Department of Endocrinology, Obesity Unit, Hospital das Clínicas Faculdade de Medicina da Universidade de São Paulo. Av. Dr. Enéas de Carvalho Aguiar, 255, 7Th Floor, Room 7037, São Paulo, SP, 05403-000, Brazil
| |
Collapse
|
41
|
Samuels JM, Paddu NU, Rekulapeli A, Madhar A, Srivastava G. High Prevalence of Positive Genetic Obesity Variants in Postoperative Bariatric Surgery Patients with Weight Regain Presenting for Medical Obesity Intervention. Obes Surg 2024; 34:170-175. [PMID: 37996769 DOI: 10.1007/s11695-023-06952-1] [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: 10/08/2023] [Revised: 11/02/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023]
Abstract
INTRODUCTION Genetic obesity susceptibility in postoperative bariatric surgery weight regain (PBSWR) remains largely unexplored. METHODS A retrospective case series of adult (N = 27) PBSWR patients who had undergone genetic obesity testing was conducted between Sept. 2020 and March 2022. PRIMARY OUTCOME frequency of genetic variants in patients experiencing weight regain following bariatric surgery. SECONDARY OUTCOMES prevalence of obesity-related comorbidities, nadir BMI achieved post-bariatric surgery, and percent total body weight loss (%TBWL) achieved with obesity pharmacotherapies. RESULTS Heterozygous mutations were identified in 22 (81%) patients, with the most prevalent mutations occurring in CEP290, RPGR1P1L, and LEPR genes (3 patients each). Median age was 56 years (interquartile range (IQR) 46.8-65.5), 88% female. Types of surgery were 67% RYGB, 19% SG, 4% gastric band, and 13% revisions. Median nadir BMI postoperatively was 34.0 kg/m2 (IQR 29.0-38.5). A high prevalence of metabolic derangements was noted; patients presented median 80 months (IQR 39-168.5) postoperative for medical weight management with 40% weight regain. BMI at initiation of anti-obesity medication (AOMs) was 41.7 kg/m2 (36.8-44.4). All received AOM and required at least 3 AOMs for weight regain. Semaglutide (N = 21), topiramate (N = 14), and metformin (N = 12) were most prescribed. Median %TBWL for the cohort at the first, second, and third visit was 1.7, 5.0, and 6.5 respectively. Fourteen (52%) achieved 5%TBWL, 10 (37%) achieved 10%TBWL, and 4 (15%) achieved 15%TBWL with combination AOMs and supervised medical intervention. CONCLUSION An unusually high prevalence of genetic obesity variants in PBSWR was found, warranting further research.
Collapse
Affiliation(s)
- Jason M Samuels
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37204, USA
- Vanderbilt Weight Loss Clinics, Vanderbilt University Medical Center, Nashville, TN, 37204, USA
| | - Nina U Paddu
- Vanderbilt Weight Loss Clinics, Vanderbilt University Medical Center, Nashville, TN, 37204, USA
- Department of Medicine, Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University School of Medicine, Nashville, TN, 37204, USA
| | - Akhil Rekulapeli
- Department of Medicine, Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University School of Medicine, Nashville, TN, 37204, USA
| | - Ayush Madhar
- Department of Medicine, Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University School of Medicine, Nashville, TN, 37204, USA
| | - Gitanjali Srivastava
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37204, USA.
- Vanderbilt Weight Loss Clinics, Vanderbilt University Medical Center, Nashville, TN, 37204, USA.
- Department of Medicine, Division of Diabetes, Endocrinology & Metabolism, Vanderbilt University School of Medicine, Nashville, TN, 37204, USA.
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN, 37204, USA.
| |
Collapse
|
42
|
Schultes B, Ernst B, Hallschmid M, Bueter M, Meyhöfer SM. The 'Behavioral Balance Model': A new perspective on the aetiology and therapy of obesity. Diabetes Obes Metab 2023; 25:3444-3452. [PMID: 37694802 DOI: 10.1111/dom.15271] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023]
Abstract
Obesity is a debilitating disease of global proportions that necessitates refined, concept-driven therapeutic approaches. Policy makers, the public and even health care professionals, but also individuals with obesity harbour many misconceptions regarding this disease, which leads to prejudice, negative attitudes, stigmatization, discrimination, self-blame, and failure to provide and finance adequate medical care. Decades of intensive, successful scientific research on obesity have only had a very limited effect on this predicament. We propose a science-based, easy-to-understand conceptual model that synthesizes the complex pathogenesis of obesity including biological, psychological, social, economic and environmental aspects with the aim to explain and communicate better the nature of obesity and currently available therapeutic modalities. According to our integrative 'Behavioral Balance Model', 'top-down cognitive control' strategies are implemented (often with limited success) to counterbalance the increased 'bottom-up drive' to gain weight, which is triggered by biological, psycho-social and environmental mechanisms in people with obesity. Besides offering a deeper understanding of obesity, the model also highlights why there is a strong need for multimodal therapeutic approaches that may not only increase top-down control but also reduce a pathologically increased bottom-up drive.
Collapse
Affiliation(s)
- Bernd Schultes
- Metabolic Center St. Gallen, friendlyDocs Ltd, St. Gallen, Switzerland
| | - Barbara Ernst
- Metabolic Center St. Gallen, friendlyDocs Ltd, St. Gallen, Switzerland
| | - Manfred Hallschmid
- Department of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen (IDM), Tübingen, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Marco Bueter
- Department of Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
- Department of Surgery, Spital Männedorf, Männedorf, Switzerland
| | - Sebastian M Meyhöfer
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Endocrinology & Diabetes, University of Lübeck, Lübeck, Germany
| |
Collapse
|
43
|
Yan J, Hu C. Bone marrow immune cells stop weight regain. Cell Metab 2023; 35:1845-1846. [PMID: 37939653 DOI: 10.1016/j.cmet.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 11/10/2023]
Abstract
Weight regain is a major challenge in the long-term management of obesity; however, the underlying mechanisms remain unclear. Zhou et al. found that bone-marrow-derived CD7+ monocytes respond to fluctuating nutritional stress and suppress weight regain by promoting beige fat thermogenesis.
Collapse
Affiliation(s)
- Jing Yan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, China.
| |
Collapse
|
44
|
Lisco G, De Tullio A, Iovino M, Disoteo O, Guastamacchia E, Giagulli VA, Triggiani V. Dopamine in the Regulation of Glucose Homeostasis, Pathogenesis of Type 2 Diabetes, and Chronic Conditions of Impaired Dopamine Activity/Metabolism: Implication for Pathophysiological and Therapeutic Purposes. Biomedicines 2023; 11:2993. [PMID: 38001993 PMCID: PMC10669051 DOI: 10.3390/biomedicines11112993] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Dopamine regulates several functions, such as voluntary movements, spatial memory, motivation, sleep, arousal, feeding, immune function, maternal behaviors, and lactation. Less clear is the role of dopamine in the pathophysiology of type 2 diabetes mellitus (T2D) and chronic complications and conditions frequently associated with it. This review summarizes recent evidence on the role of dopamine in regulating insular metabolism and activity, the pathophysiology of traditional chronic complications associated with T2D, the pathophysiological interconnection between T2D and chronic neurological and psychiatric disorders characterized by impaired dopamine activity/metabolism, and therapeutic implications. Reinforcing dopamine signaling is therapeutic in T2D, especially in patients with dopamine-related disorders, such as Parkinson's and Huntington's diseases, addictions, and attention-deficit/hyperactivity disorder. On the other hand, although specific trials are probably needed, certain medications approved for T2D (e.g., metformin, pioglitazone, incretin-based therapy, and gliflozins) may have a therapeutic role in such dopamine-related disorders due to anti-inflammatory and anti-oxidative effects, improvement in insulin signaling, neuroinflammation, mitochondrial dysfunction, autophagy, and apoptosis, restoration of striatal dopamine synthesis, and modulation of dopamine signaling associated with reward and hedonic eating. Last, targeting dopamine metabolism could have the potential for diagnostic and therapeutic purposes in chronic diabetes-related complications, such as diabetic retinopathy.
Collapse
Affiliation(s)
- Giuseppe Lisco
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Anna De Tullio
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Michele Iovino
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Olga Disoteo
- Diabetology Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari, 70124 Bari, Italy; (G.L.); (A.D.T.); (M.I.); (E.G.); (V.A.G.)
| |
Collapse
|
45
|
Zhou HY, Feng X, Wang LW, Zhou R, Sun H, Chen X, Lu RB, Huang Y, Guo Q, Luo XH. Bone marrow immune cells respond to fluctuating nutritional stress to constrain weight regain. Cell Metab 2023; 35:1915-1930.e8. [PMID: 37703873 DOI: 10.1016/j.cmet.2023.08.009] [Citation(s) in RCA: 4] [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: 03/01/2023] [Revised: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023]
Abstract
Weight regain after weight loss is a major challenge in the treatment of obesity. Immune cells adapt to fluctuating nutritional stress, but their roles in regulating weight regain remain unclear. Here, we identify a stem cell-like CD7+ monocyte subpopulation accumulating in the bone marrow (BM) of mice and humans that experienced dieting-induced weight loss. Adoptive transfer of CD7+ monocytes suppresses weight regain, whereas inducible depletion of CD7+ monocytes accelerates it. These cells, accumulating metabolic memories via epigenetic adaptations, preferentially migrate to the subcutaneous white adipose tissue (WAT), where they secrete fibrinogen-like protein 2 (FGL2) to activate the protein kinase A (PKA) signaling pathway and facilitate beige fat thermogenesis. Nevertheless, CD7+ monocytes gradually enter a quiescent state after weight loss, accompanied by increased susceptibility to weight regain. Notably, administration of FMS-like tyrosine kinase 3 ligand (FLT3L) remarkably rejuvenates CD7+ monocytes, thus ameliorating rapid weight regain. Together, our findings identify a unique bone marrow-derived metabolic-memory immune cell population that could be targeted to combat obesity.
Collapse
Affiliation(s)
- Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Li-Wen Wang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Heng Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xin Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ren-Bin Lu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan 410008, China.
| |
Collapse
|
46
|
van Baak MA, Mariman ECM. Obesity-induced and weight-loss-induced physiological factors affecting weight regain. Nat Rev Endocrinol 2023; 19:655-670. [PMID: 37696920 DOI: 10.1038/s41574-023-00887-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 09/13/2023]
Abstract
Weight regain after successful weight loss resulting from lifestyle interventions is a major challenge in the management of overweight and obesity. Knowledge of the causal mechanisms for weight regain can help researchers and clinicians to find effective strategies to tackle weight regain and reduce obesity-associated metabolic and cardiovascular complications. This Review summarizes the current understanding of a number of potential physiological mechanisms underlying weight regain after weight loss, including: the role of adipose tissue immune cells; hormonal and neuronal factors affecting hunger, satiety and reward; resting energy expenditure and adaptive thermogenesis; and lipid metabolism (lipolysis and lipid oxidation). We describe and discuss obesity-associated changes in these mechanisms, their persistence during weight loss and weight regain and their association with weight regain. Interventions to prevent or limit weight regain based on these factors, such as diet, exercise, pharmacotherapy and biomedical strategies, and current knowledge on the effectiveness of these interventions are also reviewed.
Collapse
Affiliation(s)
- Marleen A van Baak
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands.
| | - Edwin C M Mariman
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
47
|
Lee HJ, Jin BY, Park MR, Kim NH, Seo KS, Jeong YT, Wada T, Lee JS, Choi SH, Kim DH. Inhibition of adipose tissue angiogenesis prevents rebound weight gain after caloric restriction in mice fed a high-fat diet. Life Sci 2023; 332:122101. [PMID: 37730110 DOI: 10.1016/j.lfs.2023.122101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 08/30/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023]
Abstract
AIMS We investigated whether modulation of white adipose tissue (WAT) vasculature regulates rebound weight gain (RWG) after caloric restriction (CR) in mice fed a high-fat diet (HFD). MAIN METHODS We compared changes in energy balance, hypothalamic neuropeptide gene expression, and characteristics of WAT by RT-qPCR, ELISA, immunohistochemistry, and adipose-derived stromal vascular fraction spheroid sprouting assay in obese mice fed a HFD ad libitum (HFD-AL), mice under 40 % CR for 3 or 4 weeks, mice fed HFD-AL for 3 days after CR (CRAL), and CRAL mice treated with TNP-470, an angiogenic inhibitor. KEY FINDINGS WAT angiogenic genes were expressed at low levels, but WAT vascular density was maintained in the CR group compared to that in the HFD-AL group. The CRAL group showed RWG, fat regain, and hyperphagia with higher expression of angiogenic genes and reduced pericyte coverage of the endothelium in WAT on day 3 after CR compared to the CR group, indicating rapidly increased angiogenic activity after CR. Administration of TNP-470 suppressed RWG, fat regain, and hyperphagia only after CR compared to the CRAL group. Changes in circulating leptin levels and hypothalamic neuropeptide gene expression were correlated with changes in weight and fat mass, suggesting that TNP-470 suppressed hyperphagia independently of the hypothalamic melanocortin system. Additionally, TNP-470 increased gene expression related to thermogenesis, fuel utilization, and browning in brown adipose tissue (BAT) and WAT, indicating TNP-470-induced increase in thermogenesis. SIGNIFICANCE Modulation of the WAT vasculature attenuates RWG after CR by suppressing hyperphagia and increasing BAT thermogenesis and WAT browning.
Collapse
Affiliation(s)
- Hye-Jin Lee
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Bo-Yeong Jin
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Mi-Rae Park
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kwan Sik Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Yong Taek Jeong
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, Toyama 930-0194, Japan
| | - Jun-Seok Lee
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sang-Hyun Choi
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Dong-Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea; BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| |
Collapse
|
48
|
Perdomo CM, Avilés-Olmos I, Dicker D, Frühbeck G. Towards an adiposity-related disease framework for the diagnosis and management of obesities. Rev Endocr Metab Disord 2023; 24:795-807. [PMID: 37162651 PMCID: PMC10492748 DOI: 10.1007/s11154-023-09797-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 05/11/2023]
Abstract
Obesity is a complex disease that relapses frequently and associates with multiple complications that comprise a worldwide health priority because of its rising prevalence and association with numerous complications, including metabolic disorders, mechanic pathologies, and cancer, among others. Noteworthy, excess adiposity is accompanied by chronic inflammation, oxidative stress, insulin resistance, and subsequent organ dysfunction. This dysfunctional adipose tissue is initially stored in the visceral depot, overflowing subsequently to produce lipotoxicity in ectopic depots like liver, heart, muscle, and pancreas, among others. People living with obesity need a diagnostic approach that considers an exhaustive pathophysiology and complications assessment. Thus, it is essential to warrant a holistic diagnosis and management that guarantees an adequate health status, and quality of life. The present review summarizes the different complications associated with obesity, at the same time, we aim to fostering a novel framework that enhances a patient-centered approach to obesity management in the precision medicine era.
Collapse
Affiliation(s)
- Carolina M Perdomo
- Department of Endocrinology and Nutrition. Clínica, Universidad de Navarra, Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Icíar Avilés-Olmos
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Dror Dicker
- Department of Internal Medicine D, Rabin Medical Center, Hasharon Hospital, Petah Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition. Clínica, Universidad de Navarra, Pamplona, Spain.
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain.
- CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
49
|
Christiansen MR, Kilpeläinen TO, McCaffery JM. Abdominal Obesity Genetic Variants Predict Waist Circumference Regain After Weight Loss. Diabetes 2023; 72:1424-1432. [PMID: 37494631 DOI: 10.2337/db23-0131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/23/2023] [Indexed: 07/28/2023]
Abstract
Although many individuals are able to achieve weight loss, maintaining this loss over time is challenging. We aimed to study whether genetic predisposition to general or abdominal obesity predicts weight regain after weight loss. We examined the associations between genetic risk scores for higher BMI and higher waist-to-hip ratio adjusted for BMI (WHRadjBMI) with changes in weight and waist circumference up to 3 years after a 1-year weight loss program in participants (n = 822 women, n = 593 men) from the Look AHEAD (Action for Health in Diabetes) study who had lost ≥3% of their initial weight. Genetic predisposition to higher BMI or WHRadjBMI was not associated with weight regain after weight loss. However, the WHRadjBMI genetic score did predict an increase in waist circumference independent of weight change. To conclude, a genetic predisposition to higher WHRadjBMI predicts an increase in abdominal obesity after weight loss, whereas genetic predisposition to higher BMI is not predictive of weight regain. These results suggest that genetic effects on abdominal obesity may be more pronounced than those on general obesity during weight regain. ARTICLE HIGHLIGHTS Nearly all individuals who intentionally lose weight experience weight regain. Individuals with a higher genetic risk for abdominal adiposity experience increased regain in waist circumference after weight loss. Genetic predisposition to higher BMI does not predict weight regain after weight loss.
Collapse
Affiliation(s)
- Malene Revsbech Christiansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Jeanne M McCaffery
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT
| |
Collapse
|
50
|
Blaak EE, Goossens GH. Metabolic phenotyping in people living with obesity: Implications for dietary prevention. Rev Endocr Metab Disord 2023; 24:825-838. [PMID: 37581871 PMCID: PMC10492670 DOI: 10.1007/s11154-023-09830-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
Given the increasing number of people living with obesity and related chronic metabolic disease, precision nutrition approaches are required to increase the effectiveness of prevention strategies. This review addresses these approaches in different metabolic phenotypes (metabotypes) in obesity. Although obesity is typically associated with an increased cardiometabolic disease risk, some people with obesity are relatively protected against the detrimental effects of excess adiposity on cardiometabolic health, also referred to as 'metabolically healthy obesity' (MHO). Underlying mechanisms, the extent to which MHO is a transient state as well as lifestyle strategies to counteract the transition from MHO to metabolically unhealthy obesity (MUO) are discussed. Based on the limited resources that are available for dietary lifestyle interventions, it may be reasonable to prioritize interventions for people with MUO, since targeting high-risk patients for specific nutritional, lifestyle or weight-loss strategies may enhance the cost-effectiveness of these interventions. Additionally, the concept of tissue insulin resistant (IR) metabotypes is discussed, representing distinct etiologies towards type 2 diabetes (T2D) as well as cardiovascular disease (CVD). Recent evidence indicates that these tissue IR metabotypes, already present in individuals with obesity with a normal glucose homeostasis, respond differentially to diet. Modulation of dietary macronutrient composition according to these metabotypes may considerably improve cardiometabolic health benefits. Thus, nutritional or lifestyle intervention may improve cardiometabolic health, even with only minor or no weight loss, which stresses the importance of focusing on a healthy lifestyle and not on weight loss only. Targeting different metabotypes towards T2D and cardiometabolic diseases may lead to more effective lifestyle prevention and treatment strategies. Age and sex-related differences in tissue metabotypes and related microbial composition and functionality (fermentation), as important drivers and/or mediators of dietary intervention response, have to be taken into account. For the implementation of these approaches, more prospective trials are required to provide the knowledge base for precision nutrition in the prevention of chronic metabolic diseases.
Collapse
Affiliation(s)
- Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|