1
|
Suh D, Lee SM. The Impact of Obesity on Readmission and Healthcare Costs in Patients with Skin and Subcutaneous Tissue Infections. Risk Manag Healthc Policy 2025; 18:1579-1590. [PMID: 40386074 PMCID: PMC12085134 DOI: 10.2147/rmhp.s516684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025] Open
Abstract
Purpose Obesity is a global public health issue linked to worsened skin and subcutaneous tissue infections (SSTIs), complicating clinical management and increasing healthcare costs. This study aimed to evaluate obesity's influence on hospitalization duration, readmission rates, and healthcare costs among patients with SSTIs, with an emphasis on sex-specific patterns. Patients and Methods This retrospective cohort study analyzed data from South Korea's national healthcare database. The study population comprised adults hospitalized with SSTIs between 2015 and 2020. Obesity measures included body mass index (BMI) and waist circumference (WC), categorized by standard thresholds. Statistical analyses included Cox proportional hazards models for hospitalization duration, while multivariable logistic regression evaluated readmission risks. Healthcare costs were analyzed using generalized linear models, with sex-stratified analysis to examine clinical and economic outcome disparities. Results Male patients demonstrated an inverse relationship between BMI and hospitalization duration and costs, with minimal WC influence. Conversely, female patients exhibited positive associations between both obesity measures and hospitalization outcomes. SSTI-related readmissions within two years increased with rising BMI and WC across both sexes (p < 0.001). Estimated readmission costs showed significant sex-specific variations, increasing 55% among males with WC ≥ 100 cm versus < 80 cm and 132% among females with WC ≥ 95 cm versus < 75 cm. Conclusion Obesity substantially impacts SSTI clinical severity and economic costs, with distinct sex-specific disparities. Implementing tailored antimicrobial regimens, weight management strategies, and sex-specific treatment protocols is essential for outcome optimization and cost reduction. Future research should prioritize sex-specific interventions and resource allocation strategies in SSTI management.
Collapse
Affiliation(s)
- David Suh
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Seung-Mi Lee
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Republic of Korea
| |
Collapse
|
2
|
Schwartz L, Salamon K, Simoni A, Cotzomi-Ortega I, Sanchez-Zamora Y, Linn-Peirano S, John P, Ruiz-Rosado JDD, Jackson AR, Wang X, Spencer JD. Obesity promotes urinary tract infection by disrupting urothelial immune defenses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647270. [PMID: 40236097 PMCID: PMC11996552 DOI: 10.1101/2025.04.04.647270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Obesity is a significant public health concern that is associated with numerous health risks. Infections are a major complication of obesity, but the mechanisms responsible for increased infection risk are poorly defined. Here, we use a diet induced obesity mouse model and investigate how obesity impacts urinary tract infection (UTI) susceptibility and bladder immune defenses. Our results show that high-fat diet fed female and male mice exhibit increased susceptibility to uropathogenic E. coli (UPEC) following experimental UTI. Transcriptomic analysis of bladder urothelial cells shows that obesity alters gene expression in a sex-specific manner, with distinct differentially expressed genes in male and female mice, but shared activation of focal adhesion and extracellular matrix signaling. Western blot and immunostaining confirm activation of focal adhesion kinase, a central component of the focal adhesion pathway, in the bladders of obese female and male mice. Mechanistically, experiments using primary human urothelial cells demonstrate that focal adhesion kinase overexpression promotes UPEC invasion. These findings demonstrate that obesity enhances UTI susceptibility by activating focal adhesion kinase and promoting bacterial invasion of the urothelium. Together, they explain how obesity promotes UTI vulnerability and identify modifiable targets for managing obesity-associated UTI. Significance Statement Obesity is associated with an increased risk of urinary tract infections (UTIs), but the underlying mechanisms promoting infection susceptibility remain poorly understood. Here, we show that diet-induced obesity drives sex-specific changes in bladder urothelial gene expression, including distinct immune responses in male and female mice. Despite these differences, both sexes exhibit activation of focal adhesion kinase (FAK). FAK overexpression promotes bacterial invasion into human bladder cells. These findings provide a mechanistic explanation for obesity-associated UTI susceptibility and suggest that targeting FAK signaling could offer a therapeutic strategy to prevent UTIs, with implications for personalized interventions in obesity.
Collapse
|
3
|
Pessoa VFDS, Hecht M, Nitz N, Hagström L. Adipose Tissue in Chagas Disease: A Neglected Component of Pathogenesis. Pathogens 2025; 14:339. [PMID: 40333112 PMCID: PMC12030347 DOI: 10.3390/pathogens14040339] [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: 12/31/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 05/09/2025] Open
Abstract
Chagas disease (CD), caused by the protozoan T. cruzi, is a serious public health issue with high morbidity and mortality rates. Approximately 7 million people are infected, mostly in Latin America. The pathogenesis is multifactorial and poorly elucidated, particularly regarding the role of adipose tissue (AT). This review aims to explore the complex relationship between T. cruzi and AT, focusing on the possible role of this tissue in CD, as well as to explore the impact of diet on the progression of the disease. T. cruzi infects adipocytes, affecting their function. Chronic infection alters adipose physiology, contributing to systemic inflammation and metabolic disturbances. Adipokines are dysregulated, while markers of inflammation and oxidative stress increase within AT during CD. Additionally, the immune response and clinical aspects of CD may be influenced by the host's diet. High-fat diets (HFDs) impact parasite burden and cardiac pathology in murine models. The complex interaction among T. cruzi infection, AT dysfunction, and dietary factors underscore the complexity of CD pathogenesis. Despite accumulating evidence suggesting the role of AT in CD, further research is needed to elucidate its clinical implications. Understanding the bidirectional relationship between AT and T. cruzi infection may offer insights into disease progression and potential therapeutic targets, highlighting the importance of considering adipose physiology in CD management strategies.
Collapse
Affiliation(s)
- Vitória França dos Santos Pessoa
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
| | - Nadjar Nitz
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
| | - Luciana Hagström
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
- Faculty of Physical Education, University of Brasília, Brasília 70910-900, Brazil
| |
Collapse
|
4
|
Tulio EF, Lucini F, de Lima AC, Garoni Martins do Carmo ND, Barbosa MDS, de Almeida de Souza GH, Rossato L. Candida infections in COVID-19 patients: A review of prevalence, risk factors, and mortality. Indian J Med Microbiol 2025; 55:100831. [PMID: 40157425 DOI: 10.1016/j.ijmmb.2025.100831] [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: 09/05/2024] [Revised: 01/14/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Candida spp. infections have increasingly been reported among COVID-19 patients, yet the epidemiological factors, diagnostic methods, and outcomes associated with these infections remain poorly understood. These infections, particularly in ICU settings, present significant challenges due to high mortality rates and rising antifungal resistance. This study aimed to investigate the occurrence, risk factors, treatment, and outcomes of Candida albicans and non-albicans Candida in COVID-19 patients, providing clinical and epidemiological insights. METHODS A review following PRISMA guidelines was conducted. Searches were performed in PubMed, Embase, and BVS databases, covering articles published from January 2020 to May 2024. Inclusion criteria included case reports or case series providing detailed information on Candida spp. in COVID-19 patients. Data extraction focused on patient demographics, underlying diseases, antifungal and antibiotic therapies, antifungal susceptibility, resistance profiles, and outcomes. Statistical analyses were conducted using SPSS software. RESULTS The review included 67 studies, totaling 223 COVID-19 patients. Male patients were predominant. Common comorbidities included hypertension, cancer, and dyslipidemia. Echinocandins were the primary antifungal treatment. Non-albicans Candida exhibited a higher resistance rate (47.10 %) compared to C. albicans (2.35 %). Overall mortality rates were high, at 60.50 % for C. albicans and 62.30 % for non-albicans. Significant risk factors for mortality included age, central venous catheter use, ICU admission, and corticosteroid therapy. CONCLUSIONS The study identified critical risk factors and clinical characteristics in COVID-19 patients with Candida infections. The high incidence of antifungal resistance among non-albicans and high mortality rates highlight the need for vigilant monitoring and targeted antifungal strategies to improve outcomes.
Collapse
Affiliation(s)
- Eduardo Franco Tulio
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil.
| | - Fabíola Lucini
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil.
| | - Allan Carminatti de Lima
- Medical Student, Faculty of Medicine, Universidade Federal of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil.
| | | | - Marcelo Dos Santos Barbosa
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil.
| | | | - Luana Rossato
- Health Sciences Research Laboratory, Universidade Federal of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil.
| |
Collapse
|
5
|
Wiewiórska-Krata N, Foroncewicz B, Mucha K, Zagożdżon R. Cell therapies for immune-mediated disorders. Front Med (Lausanne) 2025; 12:1550527. [PMID: 40206475 PMCID: PMC11980423 DOI: 10.3389/fmed.2025.1550527] [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: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 04/11/2025] Open
Abstract
Immune-mediated disorders are a broad range of diseases, arising as consequence of immune defects, exaggerated/misguided immune response or a mixture of both conditions. Their frequency is on a rise in the developed societies and they pose a significant challenge for diagnosis and treatment. Traditional pharmacological, monoclonal antibody-based or polyclonal antibody replacement-based therapies aiming at modulation of the immune responses give very often dissatisfactory results and/or are burdened with unacceptable adverse effects. In recent years, a new group of treatment modalities has emerged, utilizing cells as living drugs, especially with the use of the up-to-date genetic engineering. These modern cellular therapies are designed to offer a high potential for more targeted, safe, durable, and personalized treatment options. This work briefly reviews the latest advances in the treatment of immune-mediated disorders, mainly those related to exaggeration of the immune response, with such cellular therapies as hematopoietic stem cells (HSCs), mesenchymal stromal cells (MSCs), regulatory T cells (Tregs), chimeric antigen receptor (CAR) T cells and others. We highlight the main features of these therapies as new treatment options for taming the dysregulated immune system. Undoubtfully, in near future such therapies can provide lasting remissions in a range of immune-mediated disorders with reduced treatment burden and improved quality of life for the patients.
Collapse
Affiliation(s)
- Natalia Wiewiórska-Krata
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- ProMix Center (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Foroncewicz
- ProMix Center (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Mucha
- ProMix Center (ProteogenOmix in Medicine), Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Radosław Zagożdżon
- Laboratory of Cellular and Genetic Therapies, Center for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
6
|
Theodorakis N, Nikolaou M. The Human Energy Balance: Uncovering the Hidden Variables of Obesity. Diseases 2025; 13:55. [PMID: 39997062 PMCID: PMC11854607 DOI: 10.3390/diseases13020055] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Obesity has emerged as a global epidemic, creating an increased burden of weight-related diseases and straining healthcare systems worldwide. While the fundamental principle of energy balance-caloric intake versus expenditure-remains central to weight regulation, real-world outcomes often deviate from simplistic predictions due to a multitude of physiological and environmental factors. Genetic predispositions, variations in basal metabolic rates, adaptive thermogenesis, physical activity, and nutrient losses via fecal and urinary excretion contribute to interindividual differences in energy homeostasis. Additionally, factors such as meal timing, macronutrient composition, gut microbiota dynamics, and diet-induced thermogenesis (DIT) further modulate energy utilization and metabolic efficiency. This Perspective explores key physiological determinants of the energy balance, while also highlighting the clinical significance of thrifty versus spendthrifty metabolic phenotypes. Key strategies for individualized weight management include precision calorimetry, circadian-aligned meal timing, the use of protein- and whole food diets to enhance DIT, and increases in non-exercise activity, as well as mild cold exposure and the use of thermogenic agents (e.g., capsaicin-like compounds) to stimulate brown adipose tissue activity. A comprehensive, personalized approach to obesity management that moves beyond restrictive caloric models is essential to achieving sustainable weight control and improving long-term metabolic health. Integrating these multifactorial insights into clinical practice will enhance obesity treatment strategies, fostering more effective and enduring interventions.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- NT-CardioMetabolics, Clinic for Metabolism and Athletic Performance, 47 Tirteou Str., 17564 Palaio Faliro, Greece
- Department of Cardiology & Preventive Cardiology Outpatient Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece;
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Maria Nikolaou
- Department of Cardiology & Preventive Cardiology Outpatient Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece;
| |
Collapse
|
7
|
Lobato S, Salomón-Soto VM, Espinosa-Méndez CM, Herrera-Moreno MN, García-Solano B, Pérez-González E, Comba-Marcó-del-Pont F, Montesano-Villamil M, Mora-Ramírez MA, Mancilla-Simbro C, Álvarez-Valenzuela R. Molecular Pathways Linking High-Fat Diet and PM 2.5 Exposure to Metabolically Abnormal Obesity: A Systematic Review and Meta-Analysis. Biomolecules 2024; 14:1607. [PMID: 39766314 PMCID: PMC11674716 DOI: 10.3390/biom14121607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Obesity, influenced by environmental pollutants, can lead to complex metabolic disruptions. This systematic review and meta-analysis examined the molecular mechanisms underlying metabolically abnormal obesity caused by exposure to a high-fat diet (HFD) and fine particulate matter (PM2.5). Following the PRISMA guidelines, articles from 2019 to 2024 were gathered from Scopus, Web of Science, and PubMed, and a random-effects meta-analysis was performed, along with subgroup analyses and pathway enrichment analyses. This study was registered in the Open Science Framework. Thirty-three articles, mainly case-control studies and murine models, were reviewed, and they revealed that combined exposure to HFD and PM2.5 resulted in the greatest weight gain (82.835 g, p = 0.048), alongside increases in high-density lipoproteins, insulin, and the superoxide dismutase. HFD enriched pathways linked to adipocytokine signaling in brown adipose tissue, while PM2.5 impacted genes associated with fat formation. Both exposures downregulated protein metabolism pathways in white adipose tissue and activated stress-response pathways in cardiac tissue. Peroxisome proliferator-activated receptor and AMP-activated protein kinase signaling pathways in the liver were enriched, influencing non-alcoholic fatty liver disease. These findings highlight that combined exposure to HFD and PM2.5 amplifies body weight gain, oxidative stress, and metabolic dysfunction, suggesting a synergistic interaction with significant implications for metabolic health.
Collapse
Affiliation(s)
- Sagrario Lobato
- Departamento de Investigación en Salud, Servicios de Salud del Estado de Puebla, 603 North 6th Street, Centro Colony, Puebla 72000, Mexico;
- Clínica de Medicina Familiar con Especialidades y Quirófano ISSSTE, 27 North Street 603, Santa Maria la Rivera Colony, Puebla 72045, Mexico
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato 80600, Mexico; (V.M.S.-S.); (M.N.H.-M.); (C.M.-S.); (R.Á.-V.)
| | - Víctor Manuel Salomón-Soto
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato 80600, Mexico; (V.M.S.-S.); (M.N.H.-M.); (C.M.-S.); (R.Á.-V.)
| | - Claudia Magaly Espinosa-Méndez
- Facultad de Cultura Física, Benemérita Universidad Autónoma de Puebla, San Claudio Avenue and 22nd South Boulevard, Ciudad Universitaria Colony, Puebla 72560, Mexico;
| | - María Nancy Herrera-Moreno
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato 80600, Mexico; (V.M.S.-S.); (M.N.H.-M.); (C.M.-S.); (R.Á.-V.)
- Departamento de Medio Ambiente, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional Unidad Sinaloa, Instituto Politécnico Nacional, Juan de Dios Bátiz Boulevard 250, San Joachin Colony, Guasave 81049, Mexico
| | - Beatriz García-Solano
- Facultad de Enfermería, Benemérita Universidad Autónoma de Puebla, 25th Avenue West 1304, Los Volcanes Colony, Puebla 74167, Mexico
| | - Ernestina Pérez-González
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato 80600, Mexico; (V.M.S.-S.); (M.N.H.-M.); (C.M.-S.); (R.Á.-V.)
- Departamento de Medio Ambiente, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional Unidad Sinaloa, Instituto Politécnico Nacional, Juan de Dios Bátiz Boulevard 250, San Joachin Colony, Guasave 81049, Mexico
| | - Facundo Comba-Marcó-del-Pont
- Facultad de Cultura Física, Benemérita Universidad Autónoma de Puebla, San Claudio Avenue and 22nd South Boulevard, Ciudad Universitaria Colony, Puebla 72560, Mexico;
| | - Mireya Montesano-Villamil
- Subsecretaría de Servicios de Salud Zona B, Servicios de Salud del Estado de Puebla, 603 North 6th Street, Centro Colony, Puebla 72000, Mexico;
| | - Marco Antonio Mora-Ramírez
- Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, San Claudio Avenue 1814, Ciudad Universitaria Colony, Puebla 72560, Mexico;
| | - Claudia Mancilla-Simbro
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato 80600, Mexico; (V.M.S.-S.); (M.N.H.-M.); (C.M.-S.); (R.Á.-V.)
- HybridLab, Fisiología y Biología Molecular de Células Excitables, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Prolongation of 14th South Street 6301, Ciudad Universitaria Colony, Puebla 72560, Mexico
| | - Ramiro Álvarez-Valenzuela
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato 80600, Mexico; (V.M.S.-S.); (M.N.H.-M.); (C.M.-S.); (R.Á.-V.)
| |
Collapse
|
8
|
Guo W, Wu D, Li L, Lewis ED, Meydani SN. Increased Fruit and Vegetable Consumption Prevents Dysregulated Immune and Inflammatory Responses in High-Fat Diet-Induced Obese Mice. J Nutr 2024; 154:3144-3150. [PMID: 39154866 DOI: 10.1016/j.tjnut.2024.08.009] [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: 04/12/2024] [Revised: 07/29/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Obesity is often associated with impaired immune responses, including enlarged spleen, increased inflammation, and impaired T-cell-mediated function, which may lead to increased susceptibility to infections. Bioactive compounds found in various fruits and vegetables (F&V) have been shown to have strong anti-inflammatory effects. However, few prospective studies have examined the effects of F&V on preventing obesity-associated dysregulation of immune and inflammatory responses. OBJECTIVE The objective of this was to determine the impact of different levels of a mixture of F&V incorporated in a high-fat diet (HFD) on immune function changes in a diet-induced obesity animal model. METHODS Six-wk-old male C57BL/6J mice were randomly assigned to 1 of 5 groups (n = 12/group): matched low-fat control (LF, 10% kcal fat) or HFD (45% kcal fat) supplemented with 0%, 5%, 10%, or 15% (wt/wt) freeze-dried powder of the most consumed F&V (human equivalent of 0, 3, 5-7, 8-9 servings/d, respectively) for 20 wk. Spleen weight was recorded, and the immunophenotype of splenocytes was evaluated by flow cytometry. Ex vivo splenic lymphocyte proliferation was assessed by thymidine incorporation and serum cytokines concentrations were measured by Meso Scale Discovery. RESULTS Mice fed the HFD exhibited significantly higher spleen weight, decreased splenic CD8+ lymphocytes, suppressed T lymphocyte proliferation, and reduced serum IL-1ß and IFN-γ concentrations compared with those fed the LF diet. Feeding mice with the HFD supplemented with 10% or 15% F&V restored HFD-associated changes of these affected biomarkers compared with those fed HFD only. Furthermore, a significant correlation was found between immunologic markers and F&V level. CONCLUSIONS These results suggest that increased consumption of F&V has beneficial effects in preventing HFD-associated dysregulation of immune function.
Collapse
Affiliation(s)
- Weimin Guo
- Nutritional Immunology Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States.
| | - Dayong Wu
- Nutritional Immunology Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Lijun Li
- Nutritional Immunology Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Erin D Lewis
- Nutritional Immunology Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States
| | - Simin Nikbin Meydani
- Nutritional Immunology Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, United States.
| |
Collapse
|
9
|
Jarkas DA, Villeneuve AH, Daneshmend AZB, Villeneuve PJ, McQuaid RJ. Sex differences in the inflammation-depression link: A systematic review and meta-analysis. Brain Behav Immun 2024; 121:257-268. [PMID: 39089535 DOI: 10.1016/j.bbi.2024.07.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024] Open
Abstract
Major Depressive Disorder (MDD) is a heterogeneous disorder that affects twice as many women than men. Precluding advances in more tailored and efficacious treatments for depression is the lack of reliable biomarkers. While depression is linked to elevations in inflammatory immune system functioning, this relationship is not evident among all individuals with depression and may vary based on symptom subtypes and/or sex. This systematic review and meta-analysis examined whether inflammatory immune peripheral markers of depression are sex-specific. PRISMA guidelines were followed for the systematic review, and a comprehensive search strategy that identified studies from PubMed and PsycInfo was applied. Studies were included if they reported C-reactive protein (CRP), interleukin (IL)-6, tumor necrosis factor (TNF)-α and/or IL-1β for males and/or females among depressed and healthy adults. We identified 23 studies that satisfied these inclusion criteria. Random-effects meta-analysis models were fit, and measures of association were summarized between levels of circulating markers of inflammation in depressed and healthy males and females. Sex-based analyses revealed elevated levels of CRP among females with depression (Cohen's d = 0.19) relative to their healthy counterparts (p = 0.02), an effect not apparent among males (Cohen's d = -0.01). Similarly, levels of IL-6 were increased among females with depression compared to healthy controls (Cohen's d = 0.51; p = 0.04), but once again this was not found among males (Cohen's d = 0.16). While TNF-α levels were elevated among individuals with depression compared to controls (p = 0.01), no statistically significant sex differences were found. The meta-analysis for IL-1β resulted in only three articles, and thus, results are presented in the supplemental section. This meta-analysis advances our understanding of the unique involvement of inflammatory biomarkers in depression among men and women, which may help inform more tailored sex-specific treatment approaches in the future.
Collapse
Affiliation(s)
- Dana A Jarkas
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada.
| | - Ally H Villeneuve
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada
| | - Ayeila Z B Daneshmend
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada
| | - Paul J Villeneuve
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Robyn J McQuaid
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, Ontario, Canada
| |
Collapse
|
10
|
Primaguna MR, Rasyid H, Aman M, Bakri S, Kasim H, Iskandar H, Dwiyanti R, Junita AR, Ridwan R, Noviyanthi RA, Purnamasar NI, Hatta M. The Strong Effect of Propolis in Suppressing NF-κB, CysC, and ACE2 on a High-fat Diet. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2024; 17:1539-1554. [DOI: 10.13005/bpj/2963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Background: A high fat diet (HFD)is one of the main causes of obesity and is closely linked to metabolic disorders brought on by stress and malfunctioning tissues. Propolis (Trigona Honey) is considered to be helpful in treating inflammatory diseases because it has also been demonstrated to have anti-inflammatory and anti-free radical properties. This study to demonstrate how much propolis supplementation affects BW, NF-κB, CysC, and ACE2 levels in Wistar rats (Rattus norvegicus) fed a HFD. Methods: Post-test and control group designs in an experimental setup. A total of twenty-four rats were randomly assigned to four groups of six. Group I received a normal diet for sixteen weeks (ND), Group II received a high fat diet (HFD) for sixteen weeks (HFD), Group III received an HFD for sixteen weeks plus propolis for eight weeks (HFD-8), and Group IV received an HFD and propolis for sixteen weeks (HFD-16). Using the Enzyme-Linked Immunosorbent Assay (ELISA), body weight (BW), serum NF-κB, Cys C, and ACE2 levels were measured before treatment (week 0), after 8 weeks of HFD (HFD-8) (week 8), and after 16 weeks of HFD (HFD-16). Results: The mean starting weight in the ND, HFD, HFD-8, and HFD-16 groups did not differ significantly (p > 0.001). By week eight, the HFD group's body weight had increased considerably (254.83 grams vs. 202.0 grams) in comparison to the ND group (p<0.001). The HFD and HFD-8 groups' body weight increased significantly at week 16 in comparison to the ND group (334.83 grams and 269.50 grams vs. 208.67 grams) (p<0.001). At week 16, there was no discernible difference in mean BW between ND and HFD-16 (p > 0.001). There was no significant difference found in the mean initial NF-κB levels between the ND, HFD, HFD-8, and HFD-16 groups (p > 0.001). At week 8, NF-κB levels in the HFD group were significantly higher (5,038 ng/ml vs. 3,655 ng/ml) (p<0.001) than in the ND group. At week 16, NF-κB levels in the HFD and HFD-8 groups were notably higher than those in the ND group (p<0.001), at 6,136 ng/ml and 4,378 ng/ml, respectively, compared to 3,775 ng/ml. Between ND and HFD-16, there was no significant distinction in the mean NF-κB levels at week 16 (p>0.001). There was no significant difference observed in the mean CysC and ACE2 between the ND, HFD, HFD-8, and HFD-16 groups (p > 0.001). CysC and ACE2 levels in the HFD group were significantly higher than those in the ND group at week 8, and in the HFD and HFD-8 groups, they were significantly higher than those in the ND group at week 16. When propolis is administered for eight weeks, the rise in BW, NF-κB, CysC, and ACE2 is suppressed until the eighth week, at which point it increases once more until the sixteenth week. Propolis administration, however, will halt the rise in BW, NF-κB, CysC, and ACE2 until the sixteenth week. Conclusion: Propolis administration for 16 weeks can suppress the increase in BW, LI, RI, NF-κB, CysC and ACE2 levels in rats given a high fat diet (HFD).
Collapse
Affiliation(s)
- Muhammad Reza Primaguna
- 1Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Haerani Rasyid
- 2Nephrology and Hypertension Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Makbul Aman
- 3Endocrine and Metabolic Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Syakib Bakri
- 2Nephrology and Hypertension Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Hasyim Kasim
- 2Nephrology and Hypertension Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Harun Iskandar
- 1Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Ressy Dwiyanti
- 4Department of Microbiology, Faculty of Medicine, Tadulako University, Palu, Indonesia
| | - Ade Rifka Junita
- 5Department of Biology and Immunology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Ridwan Ridwan
- 5Department of Biology and Immunology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Rizki Amelia Noviyanthi
- 5Department of Biology and Immunology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Nur Indah Purnamasar
- 7Department of Obstetrics and Gynecology, Faculty of Medicine, Haluoleo University, Kendari, Indonesia
| | - Mochammad Hatta
- 5Department of Biology and Immunology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
11
|
Kolinski L, Barrett TM, Kramer RA, Nunn CL. How market integration impacts human disease ecology. Evol Med Public Health 2024; 12:229-241. [PMID: 39524484 PMCID: PMC11544622 DOI: 10.1093/emph/eoae026] [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: 04/13/2024] [Revised: 08/16/2024] [Indexed: 11/16/2024] Open
Abstract
Market integration (MI), or the shift from subsistence to market-based livelihoods, profoundly influences health, yet its impacts on infectious diseases remain underexplored. Here, we synthesize the current understanding of MI and infectious disease to stimulate more research, specifically aiming to leverage concepts and tools from disease ecology and related fields to generate testable hypotheses. Embracing a One Health perspective, we examine both human-to-human and zoonotic transmission pathways in their environmental contexts to assess how MI alters infectious disease exposure and susceptibility in beneficial, detrimental and mixed ways. For human-to-human transmission, we consider how markets expand contact networks in ways that facilitate infectious disease transmission while also increasing access to hygiene products and housing materials that likely reduce infections. For zoonotic transmission, MI influences exposures to pathogens through agricultural intensification and other market-driven processes that may increase or decrease human encounters with disease reservoirs or vectors in their shared environments. We also consider how MI-driven changes in noncommunicable diseases affect immunocompetence and susceptibility to infectious disease. Throughout, we identify statistical, survey and laboratory methods from ecology and the social sciences that will advance interdisciplinary research on MI and infectious disease.
Collapse
Affiliation(s)
- Lev Kolinski
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
| | - Tyler M Barrett
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
| | - Randall A Kramer
- Nicholas School of the Environment, Duke University, Durham, NC, USA
- Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Charles L Nunn
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
- Duke Global Health Institute, Duke University, Durham, NC, USA
| |
Collapse
|
12
|
Soponkanabhorn T, Suratannon N, Buranapraditkun S, Tubjareon C, Prachuapthunyachart S, Eiamkulbutr S, Chongsrisawat V. Cellular immune response to a single dose of live attenuated hepatitis a virus vaccine in obese children and adolescents. Heliyon 2024; 10:e36610. [PMID: 39258209 PMCID: PMC11385843 DOI: 10.1016/j.heliyon.2024.e36610] [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: 08/23/2023] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024] Open
Abstract
Background Limited data are currently available regarding the cellular immune response to a live attenuated hepatitis A virus (HAV) vaccine, especially in children with obesity. The objective of this retrospective cohort study was to compare the activation of antigen-specific interferon (IFN)-γ+ T cells in obese children and adolescents with healthy individuals before and after immunization with a single dose of live attenuated HAV vaccine. Methods Blood samples were obtained from the 2021 study by Dumrisilp et al. investigating the immunogenicity of the live attenuated hepatitis A vaccine in children and young adults. Prior to enrollment, all 212 subjects had never received any HAV vaccine and tested negative for anti-HAV antibodies. The participants were vaccinated with a freeze-dried, live attenuated HAV vaccine of the H2 strain. In this study, we analyzed the stored peripheral blood mononuclear cells (PBMCs) obtained from a subgroup of 30 obese subjects and 30 normal-weight healthy controls of the same age and sex. PBMCs were collected before and 8-9 weeks after HAV vaccination for further analysis. These cells were stimulated with a recombinant antigen derived from HAV-VP3, and the immune response was evaluated using the IFN-γ enzyme-linked immunospot (ELISpot) assay. Results The between-group analysis indicated that the T-cell response of obese participants was comparable to that of normal-weight controls both before and after vaccination. The change in IFN-γ production from before to after vaccination in the obese group was not significantly different from that of the control group. Additionally, in the obese group, no correlation was found between IFN-γ production and clinical characteristics such as sex, body mass index, waist circumference, and acanthosis nigricans. Conclusion Testing for cellular immune response provides a comprehensive understanding of the overall immune response to vaccination. This study, the first to explore this significant aspect, suggests that obesity does not affect the short-term cellular immune response to live attenuated HAV vaccination.
Collapse
Affiliation(s)
- Tanatchabhorn Soponkanabhorn
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Narissara Suratannon
- Center of Excellence for Allergy and Clinical Immunology, Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Supranee Buranapraditkun
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chomchanat Tubjareon
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Sittichoke Prachuapthunyachart
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Sutha Eiamkulbutr
- Excellence Center of Organ Transplantation, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Voranush Chongsrisawat
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
13
|
Elmorsi R, Camacho L, Krijgh DD, Tilney GS, Lyu H, Traweek RS, Witt RG, Roubaud MS, Roland CL, Mericli AF. Sarcoma Size and Limb Dimensions Predict Complications, Recurrence, and Death in Patients with Soft Tissue Sarcoma in the Thigh: A Multidimensional Analysis. Ann Surg Oncol 2024; 31:5421-5430. [PMID: 38520583 DOI: 10.1245/s10434-024-15183-2] [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: 12/27/2023] [Accepted: 03/03/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Limb-sparing resections of thigh soft tissue sarcomas (STSs) can result in adverse outcomes. Identifying preoperative predictors for wound healing complications, tumor recurrence, and mortality is crucial for informed reconstructive decision-making. We hypothesized that preoperative measurements of thigh and tumor dimensions could serve as reliable indicators for postoperative complications, recurrence, and death. PATIENTS AND METHODS In this retrospective cohort study conducted from March 2016 to December 2021, we analyzed patients undergoing thigh STS excisions followed by reconstruction. Preoperative magnetic resonance imaging or computed tomography scans provided necessary thigh and tumor dimensions. Univariate and multivariate regression assessed relationships between these dimensions and postoperative outcomes, including complications, recurrence, and death. RESULTS Upon the analysis of 123 thighs, we found thigh width to be highly predictive of postoperative complications, even surpassing body mass index (BMI) and retaining significance in multivariate regression [odds ratio (OR) 1.19; 95% CI 1.03-1.39; p = 0.03]. Sarcoma-to-thigh width and thickness ratios predicted STS recurrence, with the thickness ratio retaining significance in multivariate regression (OR 1.03; 95% CI 1.001-1.05; p = 0.041). Notably, greater thigh thickness was independently protective against mortality in multivariate analysis (OR 0.80; 95% CI 0.65-0.98; p = 0.030). CONCLUSIONS Thigh width outperformed BMI in association with postoperative complications. This may create an opportunity for intervention, where weight loss can play a role during the neoadjuvant therapy period to potentially reduce complications. Sarcoma-to-thigh width and thickness ratios, particularly the latter, hold substantial predictive value in terms of STS recurrence. Moreover, thigh thickness is an independent predictor of survival.
Collapse
Affiliation(s)
- Rami Elmorsi
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luis Camacho
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David D Krijgh
- Department of Plastic, Reconstructive, and Hand Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gordon S Tilney
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Lyu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raymond S Traweek
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Russell G Witt
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Margaret S Roubaud
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina L Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander F Mericli
- Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Mahbub NU, Islam MM, Hong ST, Chung HJ. Dysbiosis of the gut microbiota and its effect on α-synuclein and prion protein misfolding: consequences for neurodegeneration. Front Cell Infect Microbiol 2024; 14:1348279. [PMID: 38435303 PMCID: PMC10904658 DOI: 10.3389/fcimb.2024.1348279] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/24/2024] [Indexed: 03/05/2024] Open
Abstract
Abnormal behavior of α-synuclein and prion proteins is the hallmark of Parkinson's disease (PD) and prion illnesses, respectively, being complex neurological disorders. A primary cause of protein aggregation, brain injury, and cognitive loss in prion illnesses is the misfolding of normal cellular prion proteins (PrPC) into an infectious form (PrPSc). Aggregation of α-synuclein causes disruptions in cellular processes in Parkinson's disease (PD), leading to loss of dopamine-producing neurons and motor symptoms. Alteration in the composition or activity of gut microbes may weaken the intestinal barrier and make it possible for prions to go from the gut to the brain. The gut-brain axis is linked to neuroinflammation; the metabolites produced by the gut microbiota affect the aggregation of α-synuclein, regulate inflammation and immunological responses, and may influence the course of the disease and neurotoxicity of proteins, even if their primary targets are distinct proteins. This thorough analysis explores the complex interactions that exist between the gut microbiota and neurodegenerative illnesses, particularly Parkinson's disease (PD) and prion disorders. The involvement of the gut microbiota, a complex collection of bacteria, archaea, fungi, viruses etc., in various neurological illnesses is becoming increasingly recognized. The gut microbiome influences neuroinflammation, neurotransmitter synthesis, mitochondrial function, and intestinal barrier integrity through the gut-brain axis, which contributes to the development and progression of disease. The review delves into the molecular mechanisms that underlie these relationships, emphasizing the effects of microbial metabolites such as bacterial lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs) in regulating brain functioning. Additionally, it looks at how environmental influences and dietary decisions affect the gut microbiome and whether they could be risk factors for neurodegenerative illnesses. This study concludes by highlighting the critical role that the gut microbiota plays in the development of Parkinson's disease (PD) and prion disease. It also provides a promising direction for future research and possible treatment approaches. People afflicted by these difficult ailments may find hope in new preventive and therapeutic approaches if the role of the gut microbiota in these diseases is better understood.
Collapse
Affiliation(s)
- Nasir Uddin Mahbub
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Md Minarul Islam
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Hea-Jong Chung
- Gwangju Center, Korea Basic Science Institute, Gwangju, Republic of Korea
| |
Collapse
|
15
|
Chumphoochai K, Manohong P, Niamnont N, Tamtin M, Sobhon P, Meemon K. Anti-Obesity Effects of Marine Macroalgae Extract Caulerpa lentillifera in a Caenorhabditis elegans Model. Mar Drugs 2023; 21:577. [PMID: 37999401 PMCID: PMC10672060 DOI: 10.3390/md21110577] [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: 09/28/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/25/2023] Open
Abstract
Obesity is a multifactorial disease characterized by an excessive accumulation of fat, which in turn poses a significant risk to health. Bioactive compounds obtained from macroalgae have demonstrated their efficacy in combating obesity in various animal models. The green macroalgae Caulerpa lentillifera (CL) contains numerous active constituents. Hence, in the present study, we aimed to elucidate the beneficial anti-obesity effects of extracts derived from C. lentillifera using a Caenorhabditis elegans obesity model. The ethanol (CLET) and ethyl acetate (CLEA) extracts caused a significant decrease in fat consumption, reaching up to approximately 50-60%. Triglyceride levels in 50 mM glucose-fed worms were significantly reduced by approximately 200%. The GFP-labeled dhs-3, a marker for lipid droplets, exhibited a significant reduction in its level to approximately 30%. Furthermore, the level of intracellular ROS displayed a significant decrease of 18.26 to 23.91% in high-glucose-fed worms treated with CL extracts, while their lifespan remained unchanged. Additionally, the mRNA expression of genes associated with lipogenesis, such as sbp-1, showed a significant down-regulation following treatment with CL extracts. This finding was supported by a significant decrease (at 16.22-18.29%) in GFP-labeled sbp-1 gene expression. These results suggest that C. lentillifera extracts may facilitate a reduction in total fat accumulation induced by glucose through sbp-1 pathways. In summary, this study highlights the anti-obesity potential of compounds derived from C. lentillifera extracts in a C. elegans model of obesity, mediated by the suppression of lipogenesis pathways.
Collapse
Affiliation(s)
- Kawita Chumphoochai
- Department of Anatomy, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand; (K.C.); (P.S.)
| | - Preeyanuch Manohong
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bang Mod, Bangkok 10140, Thailand; (P.M.); (N.N.)
| | - Nakorn Niamnont
- Department of Chemistry, Faculty of Science, King Mongkut’s University of Technology Thonburi, Bang Mod, Bangkok 10140, Thailand; (P.M.); (N.N.)
| | - Montakan Tamtin
- Kung Krabaen Bay Royal Development of Fisheries, Khlong Khut Sub-District, Tha Mai, Chantaburi 22000, Thailand;
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand; (K.C.); (P.S.)
| | - Krai Meemon
- Department of Anatomy, Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand; (K.C.); (P.S.)
| |
Collapse
|