1
|
Nalepa IF, Nielsen V, Wolf TE, Touma C, Grupe M, Asuni AA, Ratner C. Sex differences in the murine HPA axis after acute and repeated restraint stress. Stress 2025; 28:2447079. [PMID: 39819340 DOI: 10.1080/10253890.2024.2447079] [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: 07/04/2024] [Accepted: 12/08/2024] [Indexed: 01/19/2025] Open
Abstract
Chronic stress and stress-related mental illnesses such as major depressive disorder (MDD) constitute some of the leading causes of disability worldwide with a higher prevalence in women compared to men. However, preclinical research into stress and MDD is heavily biased toward using male animals only. Aberrant activity of the hypothalamic-pituitary-adrenal (HPA) axis has been linked to the development of MDD and several animal models of MDD have been established based on HPA axis dysregulation. In the present study, we compared stress biomarkers and behavior of male and female mice after acute and chronic restraint stress to investigate potential effects of sex differences in the stress response. Further, the validity of the interrupted repeated restraint stress (IRRS) model as an animal model for the HPA axis disturbances seen in MDD was assessed. After acute stress, female mice showed increased corticosterone secretion and changes in molecular markers suggesting increased HPA axis feedback sensitivity. Acute stress-induced signs of anxiety-like behavior were observed in male mice only suggesting that female mice may be more resilient to the anxiogenic effects of acute stress. Males and females responded similarly to IRRS with no sustained perturbations in HPA axis biomarkers. The IRRS model did not adequately translate to the changes reported in MDD with HPA axis overactivity and more severe perturbation models are likely needed. However, in alignment with previous studies, these data support that there are important sex differences in the HPA axis and that these may contribute to the etiology of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Isabella Flor Nalepa
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| | - Vibeke Nielsen
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| | | | - Chadi Touma
- Osnabrück University, Behavioural Biology, Osnabrück, Germany
| | - Morten Grupe
- Department of Symptom Biology, H. Lundbeck A/S, Valby, Denmark
| | - Ayodeji A Asuni
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| | - Cecilia Ratner
- Department of Preclinical Fluid Biomarkers & Occupancy, H. Lundbeck A/S, Valby, Denmark
| |
Collapse
|
2
|
Zong J, Wu X, Huang X, Yuan L, Yuan K, Zhang Z, Jiang M, Ping Z, Cheong LY, Xu A, Hoo RLC. Adipocyte-derived shed Syndecan-4 suppresses lipolysis contributing to impaired adipose tissue browning and adaptive thermogenesis. Mol Metab 2025; 96:102133. [PMID: 40180176 PMCID: PMC12004711 DOI: 10.1016/j.molmet.2025.102133] [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] [Received: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
Lipolysis in white adipose tissue (WAT) provides fatty acids as energy substrates for thermogenesis to increase energy expenditure. Syndecan-4 (Sdc4) is a transmembrane proteoglycan bearing heparan sulfate chains. Although single nucleotide polymorphisms (SNPs) of the Sdc4 gene have been identified linking to metabolic syndromes, its specific function in adipose tissue remains obscure. Here, we show that Sdc4 serves as a regulator of lipid metabolism and adaptive thermogenesis. Sdc4 expression and shedding are elevated in the white adipose tissue (WAT) of diet-induced obese mice. Adipocyte-specific deletion of Sdc4 promotes lipolysis and WAT browning, thereby raising whole-body energy expenditure to protect against diet-induced obesity. Mechanistically, fibroblast growth factor 2 (FGF2) is a paracrine factor that maintains energy homeostasis. Elevated shed Sdc4 concentrates and delivers FGF2 to fibroblast growth factor receptor 1 (FGFR1) on adipocytes, which in turn suppresses lipolysis by reducing hormone-sensitive lipase (HSL) activity, thus exaggerating adipose tissue dysfunction upon high-fat diet induction. Sdc4-deficient adipocytes show higher lipolytic and thermogenic capacity by enhancing HSL phosphorylation and UCP1 expression. Overall, our study reveals that adipocyte-derived shed Sdc4 is a novel suppressor of lipolysis, contributing to decreased energy expenditure, thus exaggerating obesity. Targeting shed Sdc4 is a potential therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Jiuyu Zong
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaowen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Lufengzi Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kai Yuan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Mengxue Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Zhihui Ping
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ruby Lai Chong Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
3
|
Mj VB, Eh TM, Jm ZM, Em BC, U OM, Ej UG, Td SG, DA OA, Mj VJ. Sex-specific differences in NAFLD development: effect of a high-sucrose diet on biochemical, histological, and genetic markers in C57bl/6N mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025; 35:1237-1250. [PMID: 39107980 DOI: 10.1080/09603123.2024.2386113] [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: 03/22/2024] [Accepted: 07/25/2024] [Indexed: 01/03/2025]
Abstract
Sucrose intake is a potential risk factor for non-alcoholic fatty liver disease (NAFLD). Individual characteristics such as sex, play arole in the biological variation of the disease, potentially related to genetic regulation. This research evaluated sex differences in biochemical, histopathological, and gene expression responses associated with NAFLD in C57bl/6N mice on a high sucrose diet. Female and male mice were assigned to control or high sucrose diets (50% sucrose solution) for 20 weeks. After sacrifice, blood and hepatic tissue were collected for analysis. Female mice revealed moderate-to-high NAFLD, whereas male mice showed mild-to-moderate NAFLD. Sex-specific variations were observed in Cd36 gene expression, an upregulation in females compared with the male group, and Adipor1 gene expression showed significant downregulation in the female group in response to high sucrose diet compared with the control group. These findings highlight the importance of considering gender disparities in the treatment and management of NAFLD.
Collapse
Affiliation(s)
- Vega Burgueño Mj
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Torres Montoya Eh
- Facultad de Biología, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Zazueta-Moreno Jm
- Facultad de Biología, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Barron-Cabrera Em
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Osuna-Martínez U
- Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Urías-García Ej
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Salinas-Garza Td
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Ochoa-Acosta DA
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| | - Vergara-Jiménez Mj
- Facultad de Ciencias de la Nutrición y Gastronomía, Universidad Autónoma de Sinaloa, Culiacán de Rosales, México
| |
Collapse
|
4
|
Schell LD, Carmody RN. An energetic framework for gut microbiome-mediated obesity induced by early-life exposure to antibiotics. Cell Host Microbe 2025; 33:470-483. [PMID: 40209676 DOI: 10.1016/j.chom.2025.03.009] [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: 11/21/2024] [Revised: 01/15/2025] [Accepted: 03/11/2025] [Indexed: 04/12/2025]
Abstract
Early-life antibiotic (ELA) exposure has garnered attention for its potential role in modulating obesity risk, although outcomes from mouse experiments and human epidemiological studies often vary based on dosage and sex. Low-dose (subtherapeutic) antibiotics can enhance energy availability through moderate alterations in gut microbiome profile, while high-dose (therapeutic) antibiotics substantially deplete the gut microbiota, thereby contributing to short-term negative energy balance. In this perspective, we propose a framework to understand how these distinct impacts of antibiotics on the gut microbiome during critical developmental windows shape long-term obesity risk through their influence on host energy balance. Using this framework, we then propose several hypotheses to explain variation in ELA-induced obesity outcomes across males and females. We conclude by discussing the evolutionary implications of ELAs, positing that the response of the gut microbiome to ELAs may signal energy availability and environmental volatility, influencing metabolic programming and adaptive traits across generations.
Collapse
Affiliation(s)
- Laura D Schell
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
5
|
Khatiz A, Tomlinson C, Ruzhytska B, Croft EK, Amrani A, Dunn S, Mendrek A, Gris D. Real-time behavioral monitoring of C57BL/6J mice during reproductive cycle. Front Neurosci 2025; 19:1509822. [PMID: 40098987 PMCID: PMC11911481 DOI: 10.3389/fnins.2025.1509822] [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: 10/11/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction The present study aims to identify differences in behavioral profiles in post-pubertal C57BL/6J males and female mice across distinct phases of the reproductive cycle in a home cage environment. Methods To reduce human bias, we used an automated behavioral analysis system HomeCageScan from CleverSys Inc. Mice were monitored continuously, and resulting data were summarized across 24-h, light, and dark cycles. Behavioral activities of each period were analyzed using hierarchical clustering, factor analysis, and principal component analysis. Results Females exhibited higher levels of physically demanding activities, including ambulatory and exploratory movements, particularly during estrus and metestrus, with estrus showing up to 30% more activity than males. In contrast, males consistently engaged in more sleep-related behaviors across all phases, with significantly higher engagement during the light cycle compared to females in proestrus and estrus (p < 0.0001); the extent of this sex difference was greater during proestrus and estrus than in metestrus and diestrus (p < 0.01). Notably, distinct patterns of sleep fragmentation were observed, with females experiencing greater disruptions during the light cycle, while males showed similar disruptions during the dark cycle. Feeding and resourcing behaviors were highest in males, showing up to 20% increase compared to cycling females, as well as significantly engaging in habituation-related behaviors such as feeding and digging. Interphase differences were observed within females, such as a significant increase of habituation-related activities during estrus compared to proestrus and diestrus (p < 0.05), while during the dark cycle, these activities peaked during the diestrus phase (p < 0.05). Female mice in the metestrus phase exhibited more sleep-related behaviors than those in proestrus. Discussion Our study has revealed prevalent behavioral differences due to sex, and inter-phase variations by employing a continuous monitoring approach designed to reduce bias. This methodology ensures a comprehensive understanding of natural behavioral patterns and strategies.
Collapse
Affiliation(s)
- Ariane Khatiz
- Program of Physiology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Cassidy Tomlinson
- Program of Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Bohdana Ruzhytska
- Program of Translational Medical Bioengineering, National Technical University of Ukraine, Kyiv, Ukraine
| | - Erika Kathe Croft
- Department of Pediatrics, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Abdelaziz Amrani
- Department of Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Shannon Dunn
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Biological Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Adrianna Mendrek
- Department of Psychology, Bishop’s University, Sherbrooke, QC, Canada
| | - Denis Gris
- Department of Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
- Department of Physiology and Pharmacology, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
6
|
Moody M, Zainadine N, Doktorski T, Trivedi R, Schmidt TA, Deymier A. Potassium bicarbonate, not sodium bicarbonate, maintains acidosis-mediated bone dissolution. Bone 2025; 192:117369. [PMID: 39674389 DOI: 10.1016/j.bone.2024.117369] [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: 04/02/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Treatments for metabolic acidosis are not well studied; however, one treatment that is commonly used is sodium bicarbonate administration. Sodium bicarbonate has been shown to help reduce symptoms of metabolic acidosis, but its benefits for bone health remain uncertain. Potassium bicarbonate has become a potential new treatment due to its reduction in bone resorption markers, unlike sodium bicarbonate. However, very few studies have looked at the connection between bone functionality and potassium bicarbonate supplementation, especially under the influence of an acidic challenge. To determine the impact of potassium bicarbonate and sodium bicarbonate on the mechanical, structural, compositional, and cellular properties of bone, acidotic mice were given either potassium bicarbonate or sodium bicarbonate for seven days. Blood gas analysis was conducted to evaluate their acidotic states throughout the study. After experimentation, the mice were euthanized, and their femurs excised for further analysis. Before bicarbonate supplementation, the acidotic mice given sodium bicarbonate were in acidosis while the acidotic mice given potassium bicarbonate were in acidemia. The bicarbonate treatment somewhat rescued the blood gas parameters in both acidosis groups, but acidemia and bone dissolution continued occurring in the acidotic mice given potassium bicarbonate, as made evident by the continuous elevation in blood sodium levels compared to the control. The acidosis group given potassium bicarbonate group also had worsened composition and structure, while the acidosis group given sodium bicarbonate had no changes in bone metrics. In this study, potassium bicarbonate was not effective at reducing bone dissolution under acidotic conditions.
Collapse
Affiliation(s)
- Mikayla Moody
- Dept of Biomedical Engineering, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Nayara Zainadine
- Dept of Biomedical Engineering, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Trey Doktorski
- Dept of Biomedical Engineering, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Ruchir Trivedi
- Department of Nephrology, School of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Tannin A Schmidt
- Dept of Biomedical Engineering, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Alix Deymier
- Dept of Biomedical Engineering, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
7
|
Dileo E, Saba F, Parasiliti-Caprino M, Rosso C, Bugianesi E. Impact of Sexual Dimorphism on Therapy Response in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease: From Conventional and Nutritional Approaches to Emerging Therapies. Nutrients 2025; 17:477. [PMID: 39940335 PMCID: PMC11821005 DOI: 10.3390/nu17030477] [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: 12/21/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a spectrum of liver disease ranging from hepatic fat accumulation to steatohepatitis (metabolic dysfunction-associated steatohepatitis, MASH), fibrosis, cirrhosis, and potentially hepatocellular carcinoma in the absence of excessive alcohol consumption. MASLD is characterized by substantial inter-individual variability in terms of severity and rate of progression, with a prevalence that is generally higher in men than in women. Steroids metabolism is characterized by sexual dimorphism and may have an impact on liver disease progression; indeed, several therapeutic strategies targeting hormone receptors are under phase 2/3 development. Despite the fact that the importance of sexual dimorphism in the setting of MASLD is well recognized, the underlying molecular mechanisms that can potentially drive the disease toward progression are not clear. The aim of this review is to delve into the crosstalk between sexual dimorphism and steroid hormone perturbation under nutritional and pharmacological intervention.
Collapse
Affiliation(s)
| | | | | | - Chiara Rosso
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.D.); (F.S.); (M.P.-C.)
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.D.); (F.S.); (M.P.-C.)
| |
Collapse
|
8
|
Tavares MR, Dos Santos WO, Amaral AG, List EO, Kopchick JJ, Alves GA, Frazao R, Dos Santos JDM, Cruz AG, Camporez JP, Donato J. Growth hormone receptor in VGLUT2 or Sim1 cells regulates glycemia and insulin sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2407225121. [PMID: 39700135 DOI: 10.1073/pnas.2407225121] [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: 04/10/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
Growth hormone (GH) has several metabolic effects, including a profound impact on glucose homeostasis. For example, GH oversecretion induces insulin resistance and increases the risk of developing diabetes mellitus. Here, we show that GH receptor (GHR) ablation in vesicular glutamate transporter 2 (VGLUT2)-expressing cells, which comprise a subgroup of glutamatergic neurons, led to a slight decrease in lean body mass without inducing changes in body adiposity. VGLUT2∆GHR mice exhibited reduced glycemia and improved glucose tolerance and insulin sensitivity. Among different glutamatergic neuronal populations, we found that GHR inactivation in Sim1-expressing cells recapitulated the phenotype observed in VGLUT2∆GHR mice. Furthermore, Sim1∆GHR mice exhibited reduced endogenous glucose production and improved hepatic insulin sensitivity without alterations in whole-body or muscle glucose uptake. Sim1∆GHR mice were protected against acute but not chronic diabetogenic effects of exogenous GH administration. Pharmacological activation of ATP-sensitive potassium channels in the brain normalized blood glucose levels in Sim1∆GHR mice. In conclusion, the absence of GHR signaling in VGLUT2/Sim1-expressing cells causes a persistent reduction in glycemia and improves hepatic insulin sensitivity. Central glucose-sensing mechanisms are likely involved in the reduced glycemia exhibited by Sim1∆GHR mice. The current findings uncover a mechanism involved in the effects of GHR signaling in regulating glucose homeostasis.
Collapse
Affiliation(s)
- Mariana R Tavares
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Willian O Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Andressa G Amaral
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701
| | - Guilherme A Alves
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil
| | - Renata Frazao
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-900, Brazil
| | - Jessica D M Dos Santos
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil
| | - Alessandra G Cruz
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil
| | - João Paulo Camporez
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
9
|
Chen X, Lin E, Haghighatian MM, Shepard LW, Hattar S, Kuruvilla R, Zhao H. Light modulates glucose and lipid homeostasis via the sympathetic nervous system. SCIENCE ADVANCES 2024; 10:eadp3284. [PMID: 39661675 PMCID: PMC11633741 DOI: 10.1126/sciadv.adp3284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
Light is an important environmental factor for vision and for diverse physiological and psychological functions. Light can also modulate glucose metabolism. Here, we show that in mice, light is critical for glucose and lipid homeostasis by regulating the sympathetic nervous system, independent of circadian disruption. Light deprivation from birth elicits insulin hypersecretion, glucagon hyposecretion, lower gluconeogenesis, and reduced lipolysis by 6 to 8 weeks in male, but not female, mice. These metabolic defects are consistent with blunted sympathetic activity, and indeed, sympathetic responses to a cold stimulus are substantially attenuated in dark-reared mice. Further, long-term dark rearing leads to body weight gain, insulin resistance, and glucose intolerance. Notably, metabolic dysfunction can be partially alleviated by 5 weeks exposure to a regular light-dark cycle. These studies provide insight into circadian-independent mechanisms by which light directly influences whole-body physiology and better understanding of metabolic disorders linked to aberrant environmental light conditions.
Collapse
Affiliation(s)
- Xiangning Chen
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eugene Lin
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
10
|
Kwon SY, Park YJ. Function of NAD metabolism in white adipose tissue: lessons from mouse models. Adipocyte 2024; 13:2313297. [PMID: 38316756 PMCID: PMC10877972 DOI: 10.1080/21623945.2024.2313297] [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: 12/14/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Nicotinamide Adenine Dinucleotide (NAD) is an endogenous substance in redox reactions and regulates various functions in metabolism. NAD and its precursors are known for their anti-ageing and anti-obesity properties and are mainly active in the liver and muscle. Boosting NAD+ through supplementation with the precursors, such as nicotinamide mononucleotide (NMN) or nicotinamide riboside (NR), enhances insulin sensitivity and circadian rhythm in the liver, and improves mitochondrial function in the muscle. Recent evidence has revealed that the adipose tissue could be another direct target of NAD supplementation by attenuating inflammation and fat accumulation. Moreover, murine studies with genetically modified models demonstrated that nicotinamide phosphoribosyltransferase (NAMPT), a NAD regulatory enzyme that synthesizes NMN, played a critical role in lipogenesis and lipolysis in an adipocyte-specific manner. The tissue-specific effects of NAD+ metabolic pathways indicate a potential of the NAD precursors to control metabolic stress particularly via focusing on adipose tissue. Therefore, this narrative review raises an importance of NAD metabolism in white adipose tissue (WAT) through a variety of studies using different mouse models.
Collapse
Affiliation(s)
- So Young Kwon
- Graduate Program in System Health and Engineering, Ewha Womans University, Seoul, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Yoon Jung Park
- Graduate Program in System Health and Engineering, Ewha Womans University, Seoul, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Tavares MR, Dos Santos WO, Furigo IC, List EO, Kopchick JJ, Donato J. Growth Hormone Receptor in Lateral Hypothalamic Neurons Is Required for Increased Food-Seeking Behavior during Food Restriction in Male Mice. J Neurosci 2024; 44:e1761232024. [PMID: 39358046 PMCID: PMC11580784 DOI: 10.1523/jneurosci.1761-23.2024] [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: 09/18/2023] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024] Open
Abstract
Growth hormone (GH) action in the brain regulates neuroendocrine axes, energy and glucose homeostasis, and several neurological functions. The lateral hypothalamic area (LHA) contains numerous neurons that respond to a systemic GH injection by expressing the phosphorylated STAT5, a GH receptor (GHR) signaling marker. However, the potential role of GHR signaling in the LHA is unknown. In this study, we demonstrated that ∼70% of orexin- and leptin receptor (LepR)-expressing neurons in the LHA are responsive to GH. Male mice carrying inactivation of the Ghr gene in the LHA were generated via bilateral injections of an adeno-associated virus. In ad libitum-fed mice, GHR ablation in LHA neurons did not significantly change energy and glucose homeostasis. Subsequently, mice were subjected to 5 d of 40% food restriction. Food restriction decreased body weight, energy expenditure, and carbohydrate oxidation. These effects were similarly observed in control and LHAΔGHR mice. While food-deprived control mice progressively increased ambulatory/exploratory activity and food-seeking behavior, LHAΔGHR mice did not show hyperactivity induced by food restriction. GHR ablation in the LHA reduced the percentage of orexin neurons expressing c-Fos during food restriction. Additionally, an acute GH injection increased the expression of c-Fos in LHAORX neurons. Inactivation of Ghr in LepR-expressing cells did not prevent hyperactivity in food-deprived mice, whereas whole-brain Ghr knock-out mice showed reduced ambulatory activity during food restriction. Our findings indicate that GHR signaling in the LHA regulates the activity of orexin neurons and is necessary to increase food-seeking behavior in food-deprived male mice.
Collapse
Affiliation(s)
- Mariana R Tavares
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Sao Paulo, Brazil
| | - Willian O Dos Santos
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Sao Paulo, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Sao Paulo, Brazil
- Centre for Health and Life Sciences, Coventry University, Coventry CV1 2DS, Warwickshire, United Kingdom
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Sao Paulo, Brazil
| |
Collapse
|
12
|
Chen X, Lin E, Haghighatian MM, Shepard LW, Hattar S, Kuruvilla R, Zhao H. Light modulates glucose and lipid homeostasis via the sympathetic nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617839. [PMID: 39416062 PMCID: PMC11483057 DOI: 10.1101/2024.10.11.617839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Light is an important environmental factor for vision, and for diverse physiological and psychological functions. Light can also modulate glucose metabolism. Here, we show that in mice, light is critical for glucose and lipid homeostasis by regulating the sympathetic nervous system, independent of circadian disruption. Light deprivation from birth elicits insulin hypersecretion, glucagon hyposecretion, lower gluconeogenesis, and reduced lipolysis by 6-8 weeks, in male, but not, female mice. These metabolic defects are consistent with blunted sympathetic activity, and indeed, sympathetic responses to a cold stimulus are significantly attenuated in dark-reared mice. Further, long-term dark rearing leads to body weight gain, insulin resistance, and glucose intolerance. Notably, metabolic dysfunction can be partially alleviated by 5 weeks exposure to a regular light-dark cycle. These studies provide insight into circadian-independent mechanisms by which light directly influences whole-body physiology and inform new approaches for understanding metabolic disorders linked to aberrant environmental light conditions.
Collapse
Affiliation(s)
- Xiangning Chen
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Eugene Lin
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | | | | | - Samer Hattar
- Section on Light and Circadian Rhythms, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, 21218, USA
| |
Collapse
|
13
|
Schmidt AV, Bharathi SS, Solo KJ, Bons J, Rose JP, Schilling B, Goetzman ES. Sirt2 Regulates Liver Metabolism in a Sex-Specific Manner. Biomolecules 2024; 14:1160. [PMID: 39334926 PMCID: PMC11430619 DOI: 10.3390/biom14091160] [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: 08/16/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Sirtuin-2 (Sirt2), an NAD+-dependent lysine deacylase enzyme, has previously been implicated as a regulator of glucose metabolism, but the specific mechanisms remain poorly defined. Here, we observed that Sirt2-/- males, but not females, have decreased body fat, moderate hypoglycemia upon fasting, and perturbed glucose handling during exercise compared to wild type controls. Conversion of injected lactate, pyruvate, and glycerol boluses into glucose via gluconeogenesis was impaired, but only in males. Primary Sirt2-/- male hepatocytes exhibited reduced glycolysis and reduced mitochondrial respiration. RNAseq and proteomics were used to interrogate the mechanisms behind this liver phenotype. Loss of Sirt2 did not lead to transcriptional dysregulation, as very few genes were altered in the transcriptome. In keeping with this, there were also negligible changes to protein abundance. Site-specific quantification of the hepatic acetylome, however, showed that 13% of all detected acetylated peptides were significantly increased in Sirt2-/- male liver versus wild type, representing putative Sirt2 target sites. Strikingly, none of these putative target sites were hyperacetylated in Sirt2-/- female liver. The target sites in the male liver were distributed across mitochondria (44%), cytoplasm (32%), nucleus (8%), and other compartments (16%). Despite the high number of putative mitochondrial Sirt2 targets, Sirt2 antigen was not detected in purified wild type liver mitochondria, suggesting that Sirt2's regulation of mitochondrial function occurs from outside the organelle. We conclude that Sirt2 regulates hepatic protein acetylation and metabolism in a sex-specific manner.
Collapse
Affiliation(s)
- Alexandra V. Schmidt
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA; (A.V.S.); (S.S.B.); (K.J.S.)
| | - Sivakama S. Bharathi
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA; (A.V.S.); (S.S.B.); (K.J.S.)
| | - Keaton J. Solo
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA; (A.V.S.); (S.S.B.); (K.J.S.)
| | - Joanna Bons
- The Buck Institute for Research on Aging, Novato, CA 94945, USA; (J.B.)
| | - Jacob P. Rose
- The Buck Institute for Research on Aging, Novato, CA 94945, USA; (J.B.)
| | - Birgit Schilling
- The Buck Institute for Research on Aging, Novato, CA 94945, USA; (J.B.)
| | - Eric S. Goetzman
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA; (A.V.S.); (S.S.B.); (K.J.S.)
| |
Collapse
|
14
|
Qi G, Tang H, Gong P, Liu Y, He C, Hu J, Kang S, Chen L, Qin S. Sex-specific hypothalamic neuropathology and glucose metabolism in an amyloidosis transgenic mouse model of Alzheimer's disease. Cell Biosci 2024; 14:120. [PMID: 39272160 PMCID: PMC11395863 DOI: 10.1186/s13578-024-01295-5] [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: 05/17/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Amyloid toxicity and glucose metabolic disorders are key pathological features during the progression of Alzheimer's disease (AD). While the hypothalamus plays a crucial role in regulating systemic energy balance, the distribution of amyloid plaques in the preoptic, anterior, tuberal, and mammillary regions of the hypothalamus in AD mice, particularly across both sexes, remains largely unclear. Our ongoing research aims to explore hypothalamic neuropathology and glucose metabolic disturbances in a well-described APP/PS1 mouse model of AD. RESULTS Immunocytochemical staining revealed that Old-AD-Female mice exhibited a greater hypothalamic Amyloid β (Aβ) burden than their Old-AD-Male counterparts, with the mammillary bodies showing the most severe accumulation. Analysis of ionized calcium binding adaptor molecule 1 (IBA1) immunoreactivity and Iba1 mRNA indicated differential microgliosis based on sex, while tanycytic territory and ZO-1 tight junction protein expression remained stable in AD mice. Moreover, sex-specific peripheral glucose metabolic parameters (random and fasting blood glucose) seemed to be exacerbated by age. Old AD mice of both sexes exhibited limited hypothalamic activation (c-Fos + cells) in response to blood glucose fluctuations. Hypothalamic Glut 1 expression decreased in young but increased in old female AD mice compared with age-matched male AD mice. Pearson correlation analysis further supported a negative correlation between hypothalamic Aβ load and random blood glucose in old AD groups of both genders, shedding light on the mechanisms underlying this amyloidosis mouse model. CONCLUSION Aged APP/PS1 mice exhibit sex-specific hypothalamic neuropathology and differential glucose metabolism, highlighting distinct pathological mechanisms within each gender.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Han Tang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Pifang Gong
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yitong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenzhao He
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianian Hu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Siying Kang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Liang Chen
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
15
|
Yagoub S, Chesters RA, Ott J, Zhu J, Cantacorps L, Ritter K, Lippert RN. Acute elevated dietary fat alone is not sufficient to decrease AgRP projections in the paraventricular nucleus of the hypothalamus in mice. Sci Rep 2024; 14:20043. [PMID: 39209992 PMCID: PMC11362280 DOI: 10.1038/s41598-024-70870-0] [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: 05/02/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Within the brain, the connections between neurons are constantly changing in response to environmental stimuli. A prime environmental regulator of neuronal activity is diet, and previous work has highlighted changes in hypothalamic connections in response to diets high in dietary fat and elevated sucrose. We sought to determine if the change in hypothalamic neuronal connections was driven primarily by an elevation in dietary fat alone. Analysis was performed in both male and female animals. We measured Agouti-related peptide (AgRP) neuropeptide and Synaptophysin markers in the paraventricular nucleus of the hypothalamus (PVH) in response to an acute 48 h high fat diet challenge. Using two image analysis methods described in previous studies, an effect of a high fat diet on AgRP neuronal projections in the PVH of male or female mice was not identified. These results suggest that it may not be dietary fat alone that is responsible for the previously published alterations in hypothalamic connections. Future work should focus on deciphering the role of individual macronutrients on neuroanatomical and functional changes.
Collapse
Affiliation(s)
- Selma Yagoub
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- University of Potsdam, Potsdam, Germany
| | - Robert A Chesters
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jonathan Ott
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- University of Potsdam, Potsdam, Germany
| | - Jiajie Zhu
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- University of Potsdam, Potsdam, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lídia Cantacorps
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Katrin Ritter
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Rachel N Lippert
- German Institute for Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Mondal S, Rathor R, Singh SN, Suryakumar G. miRNA and leptin signaling in metabolic diseases and at extreme environments. Pharmacol Res Perspect 2024; 12:e1248. [PMID: 39017237 PMCID: PMC11253706 DOI: 10.1002/prp2.1248] [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: 01/01/2024] [Revised: 05/27/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
The burden of growing concern about the dysregulation of metabolic processes arises due to complex interplay between environment and nutrition that has great impact on genetics and epigenetics of an individual. Thereby, any abnormality at the level of food intake regulating hormones may contribute to the development of metabolic diseases in any age group due to malnutrition, overweight, changing lifestyle, and exposure to extreme environments such as heat stress (HS), cold stress, or high altitude (HA). Hormones such as leptin, adiponectin, ghrelin, and cholecystokinin regulate appetite and satiety to maintain energy homeostasis. Leptin, an adipokine and a pleiotropic hormone, play major role in regulating the food intake, energy gain and energy expenditure. Using in silico approach, we have identified the major genes (LEP, LEPR, JAK2, STAT3, NPY, POMC, IRS1, SOCS3) that play crucial role in leptin signaling pathway. Further, eight miRNAs (hsa-miR-204-5p, hsa-miR-211-5p, hsa-miR-30, hsa-miR-3163, hsa-miR-33a-3p, hsa-miR-548, hsa-miR-561-3p, hsa-miR-7856-5p) from TargetScan 8.0 database were screened out that commonly target these genes. The role of these miRNAs should be explored as they might play vital role in regulating the appetite, energy metabolism, metabolic diseases (obesity, type 2 diabetes, cardiovascular diseases, inflammation), and to combat extreme environments. The miRNAs regulating leptin signaling and appetite may be useful for developing novel therapeutics for metabolic diseases.
Collapse
Affiliation(s)
- Samrita Mondal
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Richa Rathor
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Som Nath Singh
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | | |
Collapse
|
17
|
Valentine Y, Nikolajczyk BS. T cells in obesity-associated inflammation: The devil is in the details. Immunol Rev 2024; 324:25-41. [PMID: 38767210 PMCID: PMC11694249 DOI: 10.1111/imr.13354] [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] [Indexed: 05/22/2024]
Abstract
Obesity presents a significant health challenge, affecting 41% of adults and 19.7% of children in the United States. One of the associated health challenges of obesity is chronic low-grade inflammation. In both mice and humans, T cells in circulation and in the adipose tissue play a pivotal role in obesity-associated inflammation. Changes in the numbers and frequency of specific CD4+ Th subsets and their contribution to inflammation through cytokine production indicate declining metabolic health, that is, insulin resistance and T2D. While some Th subset alterations are consistent between mice and humans with obesity, some changes mainly characterize male mice, whereas female mice often resist obesity and inflammation. However, protection from obesity and inflammation is not observed in human females, who can develop obesity-related T-cell inflammation akin to males. The decline in female sex hormones after menopause is also implicated in promoting obesity and inflammation. Age is a second underappreciated factor for defining and regulating obesity-associated inflammation toward translating basic science findings to the clinic. Weight loss in mice and humans, in parallel with these other factors, does not resolve obesity-associated inflammation. Instead, inflammation persists amid modest changes in CD4+ T cell frequencies, highlighting the need for further research into resolving changes in T-cell function after weight loss. How lingering inflammation after weight loss affecting the common struggle to maintain lower weight is unknown. Semaglutide, a newly popular pharmaceutical used for treating T2D and reversing obesity, holds promise for alleviating obesity-associated health complications, yet its impact on T-cell-mediated inflammation remains unexplored. Further work in this area could significantly contribute to the scientific understanding of the impacts of weight loss and sex/hormones in obesity and obesity-associated metabolic decline.
Collapse
Affiliation(s)
- Yolander Valentine
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Barbara S. Nikolajczyk
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
18
|
Cope H, Elsborg J, Demharter S, McDonald JT, Wernecke C, Parthasarathy H, Unadkat H, Chatrathi M, Claudio J, Reinsch S, Avci P, Zwart SR, Smith SM, Heer M, Muratani M, Meydan C, Overbey E, Kim J, Chin CR, Park J, Schisler JC, Mason CE, Szewczyk NJ, Willis CRG, Salam A, Beheshti A. Transcriptomics analysis reveals molecular alterations underpinning spaceflight dermatology. COMMUNICATIONS MEDICINE 2024; 4:106. [PMID: 38862781 PMCID: PMC11166967 DOI: 10.1038/s43856-024-00532-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/23/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Spaceflight poses a unique set of challenges to humans and the hostile spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. METHODS To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on space flown murine transcriptomic datasets focused on the skin, biochemical profiles of 50 NASA astronauts and human transcriptomic datasets generated from blood and hair samples of JAXA astronauts, as well as blood samples obtained from the NASA Twins Study, and skin and blood samples from the first civilian commercial mission, Inspiration4. RESULTS Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation are identified as potential drivers for skin health risks during spaceflight. Additionally, a machine learning model is utilized to determine gene pairings associated with spaceflight response in the skin. While we identified spaceflight-induced dysregulation, such as alterations in genes associated with skin barrier function and collagen formation, our results also highlight the remarkable ability for organisms to re-adapt back to Earth via post-flight re-tuning of gene expression. CONCLUSION Our findings can guide future research on developing countermeasures for mitigating spaceflight-associated skin damage.
Collapse
Affiliation(s)
- Henry Cope
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
| | - Jonas Elsborg
- Department of Energy Conversion and Storage, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Abzu, Copenhagen, 2150, Denmark
| | | | - J Tyson McDonald
- Department of Radiation Medicine, School of Medicine, Georgetown University, Washington D.C., WA, 20057, USA
| | - Chiara Wernecke
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Department of Aerospace and Geodesy, TUM School of Engineering and Design, Technical University of Munich, Munich, Germany
| | - Hari Parthasarathy
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- College of Engineering and Haas School of Business, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Hriday Unadkat
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- School of Engineering and Applied Science, Princeton University, Princeton, NJ, 08540, USA
| | - Mira Chatrathi
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- College of Letters and Science, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jennifer Claudio
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA
| | - Sigrid Reinsch
- NASA GeneLab For High Schools Program (GL4HS), Space Biology Program, NASA Ames Research Center, Moffett Field, CA, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA
| | - Pinar Avci
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 80337, Munich, Germany
| | - Sara R Zwart
- University of Texas Medical Branch, Galveston, TX, USA
| | - Scott M Smith
- Biomedical Research and Environmental Sciences Division, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, 77058, USA
| | - Martina Heer
- IU International University of Applied Sciences, Erfurt and University of Bonn, Bonn, Germany
| | - Masafumi Muratani
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, 305-8575, Japan
- Department of Genome Biology, Institute of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Cem Meydan
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Eliah Overbey
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Jangkeun Kim
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Christopher R Chin
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Jiwoon Park
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, USA
| | - Jonathan C Schisler
- McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, USA
| | - Nathaniel J Szewczyk
- School of Medicine, University of Nottingham, Derby, DE22 3DT, UK
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Craig R G Willis
- School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Amr Salam
- St John's Institute of Dermatology, King's College London, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, Great Maze Pond, London, SE1 9RT, UK
| | - Afshin Beheshti
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett field, CA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
19
|
de Souza GO, Dos Santos WO, Donato J. Ironing out obesity. Trends Endocrinol Metab 2024; 35:456-458. [PMID: 38599900 DOI: 10.1016/j.tem.2024.04.001] [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: 03/21/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024]
Abstract
Obesity is associated with dysfunctions in hypothalamic neurons that regulate metabolism, including agouti-related protein (AgRP)-expressing neurons. In a recent article, Zhang et al. demonstrated that either diet- or genetically induced obesity promoted iron accumulation specifically in AgRP neurons. Preventing iron overload in AgRP neurons mitigated diet-induced obesity and related comorbidities in male mice.
Collapse
Affiliation(s)
- Gabriel O de Souza
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Willian O Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
20
|
Cornejo MP, Fernandez G, Cabral A, Barrile F, Heredia F, García Romero G, Zubimendi Sampieri JP, Quelas JI, Cantel S, Fehrentz JA, Alonso A, Pla R, Ferran JL, Andreoli MF, De Francesco PN, Perelló M. GHSR in a Subset of GABA Neurons Controls Food Deprivation-Induced Hyperphagia in Male Mice. Endocrinology 2024; 165:bqae061. [PMID: 38815068 DOI: 10.1210/endocr/bqae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
The growth hormone secretagogue receptor (GHSR), primarily known as the receptor for the hunger hormone ghrelin, potently controls food intake, yet the specific Ghsr-expressing cells mediating the orexigenic effects of this receptor remain incompletely characterized. Since Ghsr is expressed in gamma-aminobutyric acid (GABA)-producing neurons, we sought to investigate whether the selective expression of Ghsr in a subset of GABA neurons is sufficient to mediate GHSR's effects on feeding. First, we crossed mice that express a tamoxifen-dependent Cre recombinase in the subset of GABA neurons that express glutamic acid decarboxylase 2 (Gad2) enzyme (Gad2-CreER mice) with reporter mice, and found that ghrelin mainly targets a subset of Gad2-expressing neurons located in the hypothalamic arcuate nucleus (ARH) and that is predominantly segregated from Agouti-related protein (AgRP)-expressing neurons. Analysis of various single-cell RNA-sequencing datasets further corroborated that the primary subset of cells coexpressing Gad2 and Ghsr in the mouse brain are non-AgRP ARH neurons. Next, we crossed Gad2-CreER mice with reactivable GHSR-deficient mice to generate mice expressing Ghsr only in Gad2-expressing neurons (Gad2-GHSR mice). We found that ghrelin treatment induced the expression of the marker of transcriptional activation c-Fos in the ARH of Gad2-GHSR mice, yet failed to induce food intake. In contrast, food deprivation-induced refeeding was higher in Gad2-GHSR mice than in GHSR-deficient mice and similar to wild-type mice, suggesting that ghrelin-independent roles of GHSR in a subset of GABA neurons is sufficient for eliciting full compensatory hyperphagia in mice.
Collapse
Affiliation(s)
- María Paula Cornejo
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Gimena Fernandez
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Agustina Cabral
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Franco Barrile
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Florencia Heredia
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Guadalupe García Romero
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | | | | | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Antonia Alonso
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - Ramon Pla
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - José Luis Ferran
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia 30100, Spain
- Institute of Biomedical Research of Murcia-IMIB, Virgen de la Arrixaca University Hospital, Murcia 30100, Spain
| | - María Florencia Andreoli
- Instituto de Desarrollo e Investigaciones Pediátricas (IDIP), HIAEP Sor María Ludovica de La Plata, Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala 751 24, Sweden
| | - Pablo Nicolas De Francesco
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
| | - Mario Perelló
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional la Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata 1900, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala 751 24, Sweden
| |
Collapse
|
21
|
Rodríguez-Ibarra C, Díaz-Urbina D, Zagal-Salinas AA, Medina-Reyes EI, Déciga-Alcaraz A, Hernández-Pando R, Chirino YI. Oral exposure to food grade titanium dioxide (E171) induces intestinal and behavioural alterations in adult mice but limited effects in young mice. J Trace Elem Med Biol 2024; 83:127409. [PMID: 38394968 DOI: 10.1016/j.jtemb.2024.127409] [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: 11/10/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Food-grade titanium dioxide (E171), a white colourant widely used in ultra-processed food products, has been banned in the European Union. However, its usage is still permitted in medicines, and in several other countries. The estimated intake of E171 in children is higher than in adults, which led us to hypothesise that E171 induces differential effects depending on age, with adult mice being the most susceptible due to age, despite the lower dose. AIM To evaluate the effects of oral administration of E171 on intestinal permeability, ileum, and colon histology, and how these effects impact anxious and depressive behaviour in young and adult mice of both sexes. METHODS Young and adult mice of both sexes C57BL/6 mice received 10 mg/kgbw E171/3 times per week for 3 months. E171 was administered orally in water by pipetting, while control groups only received drinking water, then intestinal permeability, histology and animal behaviour were analysed. RESULTS E171 showed an amorphous shape, primary particles sized below 1 µm and anatase crystalline structure. Oral administration of E171 disrupted the intestinal permeability in adult male and female mice, but no effects were observed in young mice of both sexes. E171 promoted ileal adenoma formation in half of the adult female population, moreover hyperplastic crypts, and hyperplastic goblet cells at histological level in adult mice of both sexes. The colon presented hyperplastic goblet cells, hyperchromatic nuclei, increased proliferation and DNA damage in adult mice of both sexes. The anxiety and depressive behaviour were only altered in adult mice treated with E171, but no changes were detected in young animals of both sexes. CONCLUSIONS Adult mice displayed higher susceptibility in all parameters analysed in this study compared to young mice of both sexes.
Collapse
Affiliation(s)
- Carolina Rodríguez-Ibarra
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico
| | - Daniel Díaz-Urbina
- Laboratorio de Neurobiología de la Alimentación. Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico; Laboratory of Neurobiology on Compulsive Behaviors, The National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institute of Health, Bethesda, MD, USA
| | - Alejandro A Zagal-Salinas
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico
| | - Estefany I Medina-Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico
| | - Alejandro Déciga-Alcaraz
- Química de Aerosoles Orgánicos Atmosféricos, Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, CP 04510 Ciudad de México, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Vasco de Quiroga 15, Belisario Domínguez Sección 16, Tlalpan, CP 14080 Ciudad de México, Mexico
| | - Yolanda I Chirino
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. de los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla de Baz, CP 54090 Estado de México, Mexico.
| |
Collapse
|
22
|
Pontifex MG, Vauzour D, Muller M. Sexual dimorphism in the context of nutrition and health. Proc Nutr Soc 2024; 83:109-119. [PMID: 37665115 DOI: 10.1017/s0029665123003610] [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] [Indexed: 09/05/2023]
Abstract
Diets and dietary constituents that we consume have a considerable impact on disease risk. Intriguingly these effects may be modulated to some extent by sex. Lack of female representation in nutritional studies as well as a lack of stratification by sex has and continues to limit our understanding of these sex × diet interactions. Here we provide an overview of the current and available literature describing how exposure to certain dietary patterns (Western-style diet, Mediterranean diet, vegetarian/vegan, ketogenic diet) and dietary constituents (dietary fibre, PUFA and plant bioactive) influences disease risk in a sex-specific manner. Interestingly, these sex differences appear to be highly disease-specific. The identification of such sex differences in response to diet stresses the importance of sex stratification in nutritional research.
Collapse
Affiliation(s)
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Michael Muller
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| |
Collapse
|
23
|
Namasivayam S, Tilves C, Ding H, Wu S, Domingue JC, Ruiz-Bedoya C, Shah A, Bohrnsen E, Schwarz B, Bacorn M, Chen Q, Levy S, Dominguez Bello MG, Jain SK, Sears CL, Mueller NT, Hourigan SK. Fecal transplant from vaginally seeded infants decreases intraabdominal adiposity in mice. Gut Microbes 2024; 16:2353394. [PMID: 38743047 PMCID: PMC11095576 DOI: 10.1080/19490976.2024.2353394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
Exposing C-section infants to the maternal vaginal microbiome, coined "vaginal seeding", partially restores microbial colonization. However, whether vaginal seeding decreases metabolic disease risk is unknown. Therefore, we assessed the effect of vaginal seeding of human infants on adiposity in a murine model. Germ-free mice were colonized with transitional stool from human infants who received vaginal seeding or control (placebo) seeding in a double-blind randomized trial. There was a reduction in intraabdominal adipose tissue (IAAT) volume in male mice that received stool from vaginally seeded infants compared to control infants. Higher levels of isoleucine and lower levels of nucleic acid metabolites were observed in controls and correlated with increased IAAT. This suggests that early changes in the gut microbiome and metabolome caused by vaginal seeding have a positive impact on metabolic health.
Collapse
Affiliation(s)
- Sivaranjani Namasivayam
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Curtis Tilves
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Hua Ding
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shaoguang Wu
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jada C Domingue
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camilo Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ankit Shah
- Inova Health System, Inova Women’s Hospital, Falls Church, VA, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases; National Institutes of Health, Hamilton, MT, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases; National Institutes of Health, Hamilton, MT, USA
| | - Mickayla Bacorn
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Qing Chen
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shira Levy
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria Gloria Dominguez Bello
- Departments of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA
- Humans and the microbiome program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, Canada
| | - Sanjay K Jain
- Center for Infection and Inflammation Imaging Research, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia L Sears
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA
| | - Suchitra K Hourigan
- Clinical Microbiome Unit (CMU), Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of Pediatric Gastroenterology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
24
|
Nucera S, Scarano F, Macrì R, Mollace R, Gliozzi M, Carresi C, Ruga S, Serra M, Tavernese A, Caminiti R, Coppoletta A, Cardamone A, Montalcini T, Pujia A, Palma E, Muscoli C, Barillà F, Musolino V, Mollace V. The Effect of an Innovative Combination of Bergamot Polyphenolic Fraction and Cynara cardunculus L. Extract on Weight Gain Reduction and Fat Browning in Obese Mice. Int J Mol Sci 2023; 25:191. [PMID: 38203362 PMCID: PMC10779365 DOI: 10.3390/ijms25010191] [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: 11/23/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Obesity is one of the world's most serious public health issues, with a high risk of developing a wide range of diseases. As a result, focusing on adipose tissue dysfunction may help to prevent the metabolic disturbances commonly associated with obesity. Nutraceutical supplementation may be a crucial strategy for improving WAT inflammation and obesity and accelerating the browning process. The aim of this study was to perform a preclinical "proof of concept" study on Bergacyn®, an innovative formulation originating from a combination of bergamot polyphenolic fraction (BPF) and Cynara cardunculus (CyC), for the treatment of adipose tissue dysfunction. In particular, Bergacyn® supplementation in WD/SW-fed mice at doses of 50 mg/kg given orally for 12 weeks, was able to reduce body weight and total fat mass in the WD/SW mice, in association with an improvement in plasma biochemical parameters, including glycemia, total cholesterol, and LDL levels. In addition, a significant reduction in serum ALT levels was highlighted. The decreased WAT levels corresponded to an increased weight of BAT tissue, which was associated with a downregulation of PPARγ as compared to the vehicle group. Bergacyn® was able to restore PPARγ levels and prevent NF-kB overexpression in the WAT of mice fed a WD/SW diet, suggesting an improved oxidative metabolism and inflammatory status. These results were associated with a significant potentiation of the total antioxidant status in WD/SW mice. Finally, our data show, for the first time, that Bergacyn® supplementation may be a valuable approach to counteract adipose tissue dysfunction and obesity-associated effects on cardiometabolic risk.
Collapse
Affiliation(s)
- Saverio Nucera
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Stefano Ruga
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Maria Serra
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Rosamaria Caminiti
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Annarita Coppoletta
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Antonio Cardamone
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Tiziana Montalcini
- Clinical Nutrition Unit, Department of Clinical and Experimental Medicine, University Magna of Græcia of Catanzaro, 88100 Catanzaro, Italy;
| | - Arturo Pujia
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Francesco Barillà
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
- Renato Dulbecco Institute, Lamezia Terme, 88046 Catanzaro, Italy
| |
Collapse
|
25
|
Méndez N, Corvalan F, Halabi D, Ehrenfeld P, Maldonado R, Vergara K, Seron-Ferre M, Torres-Farfan C. From gestational chronodisruption to noncommunicable diseases: Pathophysiological mechanisms of programming of adult diseases, and the potential therapeutic role of melatonin. J Pineal Res 2023; 75:e12908. [PMID: 37650128 DOI: 10.1111/jpi.12908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
During gestation, the developing fetus relies on precise maternal circadian signals for optimal growth and preparation for extrauterine life. These signals regulate the daily delivery of oxygen, nutrients, hormones, and other biophysical factors while synchronizing fetal rhythms with the external photoperiod. However, modern lifestyle factors such as light pollution and shift work can induce gestational chronodisruption, leading to the desynchronization of maternal and fetal circadian rhythms. Such disruptions have been associated with adverse effects on cardiovascular, neurodevelopmental, metabolic, and endocrine functions in the fetus, increasing the susceptibility to noncommunicable diseases (NCDs) in adult life. This aligns with the Developmental Origins of Health and Disease theory, suggesting that early-life exposures can significantly influence health outcomes later in life. The consequences of gestational chronodisruption also extend into adulthood. Environmental factors like diet and stress can exacerbate the adverse effects of these disruptions, underscoring the importance of maintaining a healthy circadian rhythm across the lifespan to prevent NCDs and mitigate the impact of gestational chronodisruption on aging. Research efforts are currently aimed at identifying potential interventions to prevent or mitigate the effects of gestational chronodisruption. Melatonin supplementation during pregnancy emerges as a promising intervention, although further investigation is required to fully understand the precise mechanisms involved and to develop effective strategies for promoting health and preventing NCDs in individuals affected by gestational chronodisruption.
Collapse
Affiliation(s)
- Natalia Méndez
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Fernando Corvalan
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Diego Halabi
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- School of Dentistry, Facultad de Medicina, Universidad Austral de Chile, Santiago, Chile
| | - Pamela Ehrenfeld
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- School of Dentistry, Facultad de Medicina, Universidad Austral de Chile, Santiago, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo Maldonado
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- School of Dentistry, Facultad de Medicina, Universidad Austral de Chile, Santiago, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Karina Vergara
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Maria Seron-Ferre
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- School of Dentistry, Facultad de Medicina, Universidad Austral de Chile, Santiago, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Programa de Fisiopatología, ICBM, Facultad de Medicina, Universidad de Chile, Santiago de Chile
| | - Claudia Torres-Farfan
- Laboratorio de Cronobiología del Desarrollo, Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
26
|
Yammine L, Picatoste B, Abdullah N, Leahey RA, Johnson EF, Gómez-Banoy N, Rosselot C, Wen J, Hossain T, Goncalves MD, Lo JC, Garcia-Ocaña A, McGraw TE. Spatiotemporal regulation of GIPR signaling impacts glucose homeostasis as revealed in studies of a common GIPR variant. Mol Metab 2023; 78:101831. [PMID: 37925022 PMCID: PMC10665708 DOI: 10.1016/j.molmet.2023.101831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/06/2023] Open
Abstract
OBJECTIVE Glucose-dependent insulinotropic polypeptide (GIP) has a role in controlling postprandial metabolic tone. In humans, a GIP receptor (GIPR) variant (Q354, rs1800437) is associated with a lower body mass index (BMI) and increased risk for Type 2 Diabetes. To better understand the impacts of GIPR-Q354 on metabolism, it is necessary to study it in an isogeneic background to the predominant GIPR isoform, E354. To accomplish this objective, we used CRISPR-CAS9 editing to generate mouse models of GIPR-Q354 and GIPR-E354. Here we characterize the metabolic effects of GIPR-Q354 variant in a mouse model (GIPR-Q350). METHODS We generated the GIPR-Q350 mice for in vivo studies of metabolic impact of the variant. We isolated pancreatic islets from GIPR-Q350 mice to study insulin secretion ex vivo. We used a β-cell cell line to understand the impact of the GIPR-Q354 variant on the receptor traffic. RESULTS We found that female GIPR-Q350 mice are leaner than littermate controls, and male GIPR-Q350 mice are resistant to diet-induced obesity, in line with the association of the variant with reduced BMI in humans. GIPR-Q350 mice of both sexes are more glucose tolerant and exhibit an increased sensitivity to GIP. Postprandial GIP levels are reduced in GIPR-Q350 mice, revealing feedback regulation that balances the increased sensitivity of GIP target tissues to secretion of GIP from intestinal endocrine cells. The increased GIP sensitivity is recapitulated ex vivo during glucose stimulated insulin secretion assays in islets. Generation of cAMP in islets downstream of GIPR activation is not affected by the Q354 substitution. However, post-activation traffic of GIPR-Q354 variant in β-cells is altered, characterized by enhanced intracellular dwell time and increased localization to the Trans-Golgi Network (TGN). CONCLUSIONS Our data link altered intracellular traffic of the GIPR-Q354 variant with GIP control of metabolism. We propose that this change in spatiotemporal signaling underlies the physiologic effects of GIPR-Q350/4 and GIPR-E350/4 in mice and humans. These findings contribute to a more complete understanding of the impact of GIPR-Q354 variant on glucose homeostasis that could perhaps be leveraged to enhance pharmacologic targeting of GIPR for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Lucie Yammine
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Belén Picatoste
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Nazish Abdullah
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Rosemary A Leahey
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Emma F Johnson
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Nicolás Gómez-Banoy
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Carolina Rosselot
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jennifer Wen
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Tahmina Hossain
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | | | - James C Lo
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Adolfo Garcia-Ocaña
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA; Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Cardiothoracic Surgery, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
27
|
de Souza GO, Teixeira PDS, Câmara NOS, Donato J. mTORC1 Signaling in AgRP Neurons Is Not Required to Induce Major Neuroendocrine Adaptations to Food Restriction. Cells 2023; 12:2442. [PMID: 37887286 PMCID: PMC10605346 DOI: 10.3390/cells12202442] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Hypothalamic mTORC1 signaling is involved in nutrient sensing. Neurons that express the agouti-related protein (AgRP) are activated by food restriction and integrate interoceptive and exteroceptive signals to control food intake, energy expenditure, and other metabolic responses. To determine whether mTORC1 signaling in AgRP neurons is necessary for regulating energy and glucose homeostasis, especially in situations of negative energy balance, mice carrying ablation of the Raptor gene exclusively in AgRP-expressing cells were generated. AgRPΔRaptor mice showed no differences in body weight, fat mass, food intake, or energy expenditure; however, a slight improvement in glucose homeostasis was observed compared to the control group. When subjected to 5 days of food restriction (40% basal intake), AgRPΔRaptor female mice lost less lean body mass and showed a blunted reduction in energy expenditure, whereas AgRPΔRaptor male mice maintained a higher energy expenditure compared to control mice during the food restriction and 5 days of refeeding period. AgRPΔRaptor female mice did not exhibit the food restriction-induced increase in serum corticosterone levels. Finally, although hypothalamic fasting- or refeeding-induced Fos expression showed no differences between the groups, AgRPΔRaptor mice displayed increased hyperphagia during refeeding. Thus, some metabolic and neuroendocrine responses to food restriction are disturbed in AgRPΔRaptor mice.
Collapse
Affiliation(s)
- Gabriel O. de Souza
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil; (G.O.d.S.); (P.D.S.T.)
| | - Pryscila D. S. Teixeira
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil; (G.O.d.S.); (P.D.S.T.)
| | - Niels O. S. Câmara
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil;
| | - Jose Donato
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, SP, Brazil; (G.O.d.S.); (P.D.S.T.)
| |
Collapse
|
28
|
Barrile F, Cassano D, Fernandez G, De Francesco PN, Reynaldo M, Cantel S, Fehrentz JA, Donato J, Schiöth HB, Zigman JM, Perello M. Ghrelin's orexigenic action in the lateral hypothalamic area involves indirect recruitment of orexin neurons and arcuate nucleus activation. Psychoneuroendocrinology 2023; 156:106333. [PMID: 37454647 PMCID: PMC10530520 DOI: 10.1016/j.psyneuen.2023.106333] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/30/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVE Ghrelin is a potent orexigenic hormone, and the lateral hypothalamic area (LHA) has been suggested as a putative target mediating ghrelin's effects on food intake. Here, we aimed to investigate the presence of neurons expressing ghrelin receptor (a.k.a. growth hormone secretagogue receptor, GHSR) in the mouse LHA (LHAGHSR neurons), its physiological implications and the neuronal circuit recruited by local ghrelin action. METHODS We investigated the distribution of LHAGHSR neurons using different histologic strategies, including the use of a reporter mice expressing enhanced green fluorescent protein under the control of the GHSR promoter. Also, we investigated the physiological implications of local injections of ghrelin within the LHA, and the extent to which the orexigenic effect of intra-LHA-injected ghrelin involves the arcuate nucleus (ARH) and orexin neurons of the LHA (LHAorexin neurons) RESULTS: We found that: 1) LHAGHSR neurons are homogeneously distributed throughout the entire LHA; 2) intra-LHA injections of ghrelin transiently increase food intake and locomotor activity; 3) ghrelin's orexigenic effect in the LHA involves the indirect recruitment of LHAorexin neurons and the activation of ARH neurons; and 4) LHAGHSR neurons are not targeted by plasma ghrelin. CONCLUSIONS We provide a compelling neuroanatomical and functional characterization of LHAGHSR neurons in male mice that indicates that LHAGHSR cells are part of a hypothalamic neuronal circuit that potently induces food intake.
Collapse
Affiliation(s)
- Franco Barrile
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Gimena Fernandez
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Mirta Reynaldo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - José Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, Brazil
| | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Jeffrey M Zigman
- Center for Hypothalamic Research, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires, Argentina; Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden.
| |
Collapse
|
29
|
Mansano NDS, Vieira HR, Araujo-Lopes R, Szawka RE, Donato J, Frazao R. Fasting Modulates GABAergic Synaptic Transmission to Arcuate Kisspeptin Neurons in Female Mice. Endocrinology 2023; 164:bqad150. [PMID: 37793082 DOI: 10.1210/endocr/bqad150] [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: 06/20/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/06/2023]
Abstract
It is well-established that the hypothalamic-pituitary-gonadal (HPG) axis is suppressed due to negative energy balance. However, less information is available on whether kisspeptin neuronal activity contributes to fasting-induced responses. In the present study, female and male mice were fasted for 24 hours or provided food ad libitum (fed group) to determine whether acute fasting is sufficient to modulate kisspeptin neuronal activity. In female mice, fasting attenuated luteinizing hormone (LH) and prolactin (PRL) serum levels and increased follicle-stimulating hormone levels compared with the fed group. In contrast, fasting did not affect gonadotropin or PRL secretion in male mice. By measuring genes related to LH pulse generation in micropunches obtained from the arcuate nucleus of the hypothalamus (ARH), we observed that fasting reduced Kiss1 mRNA levels in female and male mice. In contrast, Pdyn expression was upregulated only in fasted female mice, whereas no changes in the Tac2 mRNA levels were observed in both sexes. Interestingly, the frequency and amplitude of the GABAergic postsynaptic currents recorded from ARH kisspeptin neurons (ARHKisspeptin) were reduced in 24-hour fasted female mice but not in males. Additionally, neuropeptide Y induced a hyperpolarization in the resting membrane potential of ARHKisspeptin neurons of fed female mice but not in males. Thus, the response of ARHKisspeptin neurons to fasting is sexually dependent with a female bias, associated with changes in gonadotropins and PRL secretion. Our findings suggest that GABAergic transmission to ARHKisspeptin neurons modulates the activity of the HPG axis during situations of negative energy balance.
Collapse
Affiliation(s)
- Naira da Silva Mansano
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| | - Henrique Rodrigues Vieira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| | - Roberta Araujo-Lopes
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Belo Horizonte, MG 31270-901, Brazil
| | - Raphael Escorsim Szawka
- Universidade Federal de Minas Gerais, Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Belo Horizonte, MG 31270-901, Brazil
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofísica, São Paulo, SP 05508-000, Brazil
| | - Renata Frazao
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Anatomia, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
30
|
Fernandez G, De Francesco PN, Cornejo MP, Cabral A, Aguggia JP, Duque VJ, Sayar N, Cantel S, Burgos JI, Fehrentz JA, Rorato R, Atasoy D, Mecawi AS, Perello M. Ghrelin Action in the PVH of Male Mice: Accessibility, Neuronal Targets, and CRH Neurons Activation. Endocrinology 2023; 164:bqad154. [PMID: 37823477 PMCID: PMC11491828 DOI: 10.1210/endocr/bqad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/08/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023]
Abstract
The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.
Collapse
Affiliation(s)
- Gimena Fernandez
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Pablo N De Francesco
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - María P Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Agustina Cabral
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Julieta P Aguggia
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
| | - Victor J Duque
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Nilufer Sayar
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - Sonia Cantel
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Juan I Burgos
- Centro de Investigaciones Cardiovasculares “Dr. Horacio Eugenio Cingolani” (CONICET and National University of La Plata), La Plata 1900, Buenos Aires, Argentina
| | - Jean-Alain Fehrentz
- Institut des Biomolécules Max Mousseron, University of Montpellier, CNRS, ENSCM, Montpellier cedex 5 34293, France
| | - Rodrigo Rorato
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Carver College of Medicine, Iowa Neuroscience Institute and Fraternal Order of Eagles Diabetes Research Center (FOEDRC), University of Iowa, Iowa City, IA 52242, USA
| | - André S Mecawi
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, CEP: 04023-062, Brazil
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], La Plata, Buenos Aires 1900, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala 751 05, Sweden
| |
Collapse
|
31
|
Buckels EJ, Tan J, Hsu H, Zhu Y, Buchanan CM, Matthews BG, Lee KL. Preptin Deficiency Does Not Protect against High-Fat Diet-Induced Metabolic Dysfunction or Bone Loss in Mice. JBMR Plus 2023; 7:e10777. [PMID: 37614298 PMCID: PMC10443080 DOI: 10.1002/jbm4.10777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/27/2023] [Accepted: 05/18/2023] [Indexed: 08/25/2023] Open
Abstract
Preptin is derived from the cleavage of the E-peptide of pro-insulin-like growth factor (IGF)-II and is an insulin secretagogue. Observational studies have linked elevated circulating preptin to metabolic dysfunction in humans; however, a causal role for preptin in metabolic dysfunction has not been established. Additionally, preptin can promote osteoblast proliferation and differentiation, suggesting a link with skeletal health. We previously described a global preptin knockout (KO) model. In this study, we sought to uncover the impact of preptin KO in mice on the response to a moderately high-fat diet (HFD) and low-fat diet (LFD). HFD groups had higher weight and fat mass gain, lower trabecular and cortical bone volume and fracture load, and higher liver triglycerides. In males, preptin deficiency led to lower blood glucose than wild-type (WT) mice under LFD conditions. This was accompanied by differences in bone microarchitecture, including lower trabecular bone volume fraction, trabecular number, and lower cortical thickness. These differences were absent in female mice, although KO females had a HFD-driven increase in fat mass and liver triglycerides that was absent in WT mice. Female WT mice had increased glucose-stimulated insulin secretion under HFD conditions that was absent in female KO mice. Overall, preptin may have a detrimental impact on metabolism and a positive impact on bone health in male mice and may protect against liver fat storage in females while enabling islet compensation under HFD conditions. When we consider that serum preptin levels are elevated in humans of both sexes in pathological states in which insulin levels are elevated, the impact of preptin on comorbidity risk needs to be better understood. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Emma J. Buckels
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandAucklandNew Zealand
| | - Joey Tan
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Huai‐Ling Hsu
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Yuting Zhu
- Department of Engineering ScienceUniversity of AucklandAucklandNew Zealand
| | - Christina M. Buchanan
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
| | - Brya G. Matthews
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandAucklandNew Zealand
| | - Kate L. Lee
- Department of Molecular Medicine and PathologyUniversity of AucklandAucklandNew Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
32
|
Holmes-Hampton GP, Kumar VP, Valenzia K, Ghosh SP. Sex as a Factor in Murine Radiation Research: Implications for Countermeasure Development. Cytogenet Genome Res 2023; 163:187-196. [PMID: 37348469 DOI: 10.1159/000531630] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/18/2023] [Indexed: 06/24/2023] Open
Abstract
There is an increased threat of exposure to ionizing radiation; in the event of such exposure, the availability of medical countermeasures will be vital to ensure the protection of the population. Effective countermeasures should be efficacious across a varied population and most importantly amongst both males and females. Radiation research must be conducted in animal models which act as a surrogate for the human response. Here, we identify differences in survival in male and female C57BL/6 in both a total body irradiation (TBI) model using the Armed Forces Radiobiology Research Institute (AFRRI) 60Co source and a partial body irradiation (PBI) model using the AFRRI Linear Accelerator (LINAC) with 4 MV photons and 2.5% bone marrow shielding. In both models, we observed a higher degree of radioresistance in female animals and a corresponding radiosensitivity in males. One striking difference in male and female rodents is body size/weight and we investigated the role of pre-irradiation body weight on survivability for animals irradiated at the same dose of irradiation (8 Gy TBI, 14 Gy PBI). We found that weight does not influence survival in the TBI model and that heavier males but lighter females have increased survival in the PBI model. This incongruence in survival amongst the sexes should be taken into consideration in the course of developing radiation countermeasures for response to a mass casualty incident.
Collapse
Affiliation(s)
- Gregory P Holmes-Hampton
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Vidya P Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Kaylee Valenzia
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Sanchita P Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Wasinski F, Tavares MR, Gusmao DO, List EO, Kopchick JJ, Alves GA, Frazao R, Donato J. Central growth hormone action regulates neuroglial and proinflammatory markers in the hypothalamus of male mice. Neurosci Lett 2023; 806:137236. [PMID: 37030549 PMCID: PMC10133206 DOI: 10.1016/j.neulet.2023.137236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023]
Abstract
Growth hormone (GH) action in specific neuronal populations regulates neuroendocrine responses, metabolism, and behavior. However, the potential role of central GH action on glial function is less understood. The present study aims to determine how the hypothalamic expression of several neuroglial markers is affected by central GH action in male mice. The dwarf GH- and insulin-like growth factor-1 (IGF-1)-deficient Ghrhrlit/lit mice showed decreased mRNA expression of Nes (Nestin), Gfap, Iba1, Adgre1 (F4/80), and Tnf (TNFα) in the hypothalamus, compared to wild-type animals. In contrast, transgenic overexpression of GH led to high serum GH and IGF-1 levels, and increased hypothalamic expression of Nes, Gfap, Adgre1, Iba1, and Rax. Hepatocyte-specific GH receptor (GHR) knockout mice, which are characterized by high serum GH levels, but reduced IGF-1 secretion, showed increased mRNA expression of Gfap, Iba1, Tnf, and Sox10, demonstrating that the increase in GH levels alters the hypothalamic expression of glial markers associated with neuroinflammation, independently of IGF-1. Conversely, brain-specific GHR knockout mice showed reduced expression of Gfap, Adgre1, and Vim (vimentin), indicating that brain GHR signaling is necessary to mediate GH-induced changes in the expression of several neuroglial markers. In conclusion, the hypothalamic mRNA levels of several neuroglial markers associated with inflammation are directly modulated by GHR signaling in male mice.
Collapse
Affiliation(s)
- Frederick Wasinski
- Department of Neurology and Neurosurgery, Universidade Federal de Sao Paulo, Sao Paulo, SP 04039-032, Brazil
| | - Mariana R Tavares
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Daniela O Gusmao
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Guilherme A Alves
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-900, Brazil
| | - Renata Frazao
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-900, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, SP 05508-000, Brazil.
| |
Collapse
|
34
|
Korgan AC, Oliveira-Abreu K, Wei W, Martin SLA, Bridges ZJD, Leal-Cardoso JH, Kaczorowski CC, O'Connell KMS. High sucrose consumption decouples intrinsic and synaptic excitability of AgRP neurons without altering body weight. Int J Obes (Lond) 2023; 47:224-235. [PMID: 36725979 PMCID: PMC10023568 DOI: 10.1038/s41366-023-01265-w] [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: 10/04/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023]
Abstract
BACKGROUND/OBJECTIVE As the obesity epidemic continues, the understanding of macronutrient influence on central nervous system function is critical for understanding diet-induced obesity and potential therapeutics, particularly in light of the increased sugar content in processed foods. Previous research showed mixed effects of sucrose feeding on body weight gain but has yet to reveal insight into the impact of sucrose on hypothalamic functioning. Here, we explore the impact of liquid sucrose feeding for 12 weeks on body weight, body composition, caloric intake, and hypothalamic AgRP neuronal function and synaptic plasticity. METHODS Patch-clamp electrophysiology of hypothalamic AgRP neurons, metabolic phenotyping and food intake were performed on C57BL/6J mice. RESULTS While mice given sugar-sweetened water do not gain significant weight, they do show subtle differences in body composition and caloric intake. When given sugar-sweetened water, mice show similar alterations to AgRP neuronal excitability as in high-fat diet obese models. Increased sugar consumption also primes mice for increased caloric intake and weight gain when given access to a HFD. CONCLUSIONS Our results show that elevated sucrose consumption increased activity of AgRP neurons and altered synaptic excitability. This may contribute to obesity in mice and humans with access to more palatable (HFD) diets.
Collapse
Affiliation(s)
- Austin C Korgan
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | - Klausen Oliveira-Abreu
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, CE, Brazil
| | - Wei Wei
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
- Georgia State University, Atlanta, GA, USA
| | | | - Zoey J D Bridges
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
| | | | - Catherine C Kaczorowski
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA
- Neuroscience Program, Graduate School of Biomedical Science, Tufts University School of Medicine, Boston, MA, USA
| | - Kristen M S O'Connell
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, USA.
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, USA.
- Neuroscience Program, Graduate School of Biomedical Science, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
35
|
Cope H, Elsborg J, Demharter S, Mcdonald JT, Wernecke C, Parthasarathy H, Unadkat H, Chatrathi M, Claudio J, Reinsch S, Zwart S, Smith S, Heer M, Muratani M, Meydan C, Overbey E, Kim J, Park J, Schisler J, Mason C, Szewczyk N, Willis C, Salam A, Beheshti A. More than a Feeling: Dermatological Changes Impacted by Spaceflight. RESEARCH SQUARE 2023:rs.3.rs-2367727. [PMID: 36798347 PMCID: PMC9934743 DOI: 10.21203/rs.3.rs-2367727/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Spaceflight poses a unique set of challenges to humans and the hostile Spaceflight environment can induce a wide range of increased health risks, including dermatological issues. The biology driving the frequency of skin issues in astronauts is currently not well understood. To address this issue, we used a systems biology approach utilizing NASA's Open Science Data Repository (OSDR) on spaceflown murine transcriptomic datasets focused on the skin, biomedical profiles from fifty NASA astronauts, and confirmation via transcriptomic data from JAXA astronauts, the NASA Twins Study, and the first civilian commercial mission, Inspiration4. Key biological changes related to skin health, DNA damage & repair, and mitochondrial dysregulation were determined to be involved with skin health risks during Spaceflight. Additionally, a machine learning model was utilized to determine key genes driving Spaceflight response in the skin. These results can be used for determining potential countermeasures to mitigate Spaceflight damage to the skin.
Collapse
|
36
|
Nicotinamide riboside kinase 1 protects against diet and age-induced pancreatic β-cell failure. Mol Metab 2022; 66:101605. [PMID: 36165811 PMCID: PMC9557729 DOI: 10.1016/j.molmet.2022.101605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Disturbances in NAD+ metabolism have been described as a hallmark for multiple metabolic and age-related diseases, including type 2 diabetes. While alterations in pancreatic β-cell function are critical determinants of whole-body glucose homeostasis, the role of NAD+ metabolism in the endocrine pancreas remains poorly explored. Here, we aimed to evaluate the role of nicotinamide riboside (NR) metabolism in maintaining NAD+ levels and pancreatic β-cell function in pathophysiological conditions. METHODS Whole body and pancreatic β-cell-specific NRK1 knockout (KO) mice were metabolically phenotyped in situations of high-fat feeding and aging. We also analyzed pancreatic β-cell function, β-cell mass and gene expression. RESULTS We first demonstrate that NRK1, the essential enzyme for the utilization of NR, is abundantly expressed in pancreatic β-cells. While NR treatment did not alter glucose-stimulated insulin secretion in pancreatic islets from young healthy mice, NRK1 knockout mice displayed glucose intolerance and compromised β-cells response to a glucose challenge upon high-fat feeding or aging. Interestingly, β cell dysfunction stemmed from the functional failure of other organs, such as liver and kidney, and the associated changes in circulating peptides and hormones, as mice lacking NRK1 exclusively in β-cells did not show altered glucose homeostasis. CONCLUSIONS This work unveils a new physiological role for NR metabolism in the maintenance of glucose tolerance and pancreatic β-cell function in high-fat feeding or aging conditions.
Collapse
|
37
|
Long-term sulforaphane-treatment restores redox homeostasis and prevents cognitive decline in middleaged female and male rats, but cannot revert previous damage in old animals. Biogerontology 2022; 23:587-613. [PMID: 35960458 DOI: 10.1007/s10522-022-09984-9] [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: 05/22/2022] [Accepted: 08/02/2022] [Indexed: 11/02/2022]
Abstract
Aging is a complex and detrimental process, which disrupts most organs and systems within the organisms. The nervous system is morphologically and functionally affected during normal aging, and oxidative stress has been involved in age-related damage, leading to cognitive decline and neurodegenerative processes. Sulforaphane (SFN) is a hormetin that activates the antioxidant and anti-inflammatory responses. So, we aimed to evaluate if SFN long-term treatment was able to prevent age-associated cognitive decline in adult and old female and male rats. Memory was evaluated in adult (15-month-old), and old (21-month-old) female and male Wistar rats after three months of SFN treatment. Young rats (4-month-old) were used as age controls. The antioxidant response induction, the redox state (GSH/GSSG), and oxidative damage were determined in the brain cortex (Cx) and hippocampus (Hc). Our results showed that SFN restored redox homeostasis in the Cx and Hc of adult rats, thus preventing cognitive decline in both sexes; however, the redox responses were not the same in males and females. Old rats were not able to recover their redox state as adults did, but they had a mild improvement. These results suggest that SFN mainly prevents rather than reverts neural damage; though, there might also be a range of opportunities to use hormetins like SFN, to improve redox modulation in old animals.
Collapse
|
38
|
Smith A, Woodside B, Abizaid A. Ghrelin and the Control of Energy Balance in Females. Front Endocrinol (Lausanne) 2022; 13:904754. [PMID: 35909536 PMCID: PMC9334675 DOI: 10.3389/fendo.2022.904754] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Ghrelin is considered one of the most potent orexigenic peptide hormones and one that promotes homeostatic and hedonic food intake. Research on ghrelin, however, has been conducted predominantly in males and particularly in male rodents. In female mammals the control of energy metabolism is complex and it involves the interaction between ovarian hormones like estrogen and progesterone, and metabolic hormones. In females, the role that ghrelin plays in promoting feeding and how this is impacted by ovarian hormones is not well understood. Basal ghrelin levels are higher in females than in males, and ghrelin sensitivity changes across the estrus cycle. Yet, responses to ghrelin are lower in female and seem dependent on circulating levels of ovarian hormones. In this review we discuss the role that ghrelin plays in regulating homeostatic and hedonic food intake in females, and how the effects of ghrelin interact with those of ovarian hormones to regulate feeding and energy balance.
Collapse
Affiliation(s)
- Andrea Smith
- Department of Neuroscience, Carleton Unversity, Ottawa, ON, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton Unversity, Ottawa, ON, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton Unversity, Ottawa, ON, Canada
- Stress, Trauma and Relience (STAR) Work Group Carleton University, Ottawa, ON, Canada
- *Correspondence: Alfonso Abizaid,
| |
Collapse
|