1
|
Liu Y, Yu Z, Wang X, Yuan MQ, Lu MJ, Gong MR, Li Q, Xia YB, Yang GH, Xu B, Litscher G, Xu TC. Neurophysiological mechanisms of electroacupuncture in regulating pancreatic function and adipose tissue expansion. World J Diabetes 2025; 16:101354. [DOI: 10.4239/wjd.v16.i5.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/12/2025] [Accepted: 03/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Electroacupuncture (EA) has been recognized for its beneficial effects on glucolipid metabolism, potentially through the regulation of sensory nerve coordination. The expandability of peripancreatic adipose tissue (PAT) is implicated in the transition from obesity to type 2 diabetes mellitus (T2DM). However, the specific pancreatic responses to EA require further elucidation.
AIM To investigate the influence of EA on pancreatic glucolipid reduction level in a high-fat diet (HFD) rat model.
METHODS To delineate the precise pathway through which EA mediates interactions between PAT and islets, we assessed the expression levels of NGF, TRPV1, insulin, as well as other proteins in the pancreas and PAT. This approach enabled us to identify the acupoints that are most conducive to optimizing glycolipid metabolism.
RESULTS The ST25, LI11 and ST37 groups attenuated HFD-induced obesity and insulin resistance (IR) to distinct degrees, with ST25 group having the greatest effect. EA at ST25 was found to modify the local regulatory influence of PAT on the pancreatic intrinsic nervous system. Specifically, EA at ST25 obviously activated the TRPV1-CGRP-islet beta cell pathway, contributing to the relief of glucolipid metabolic stress. The beneficial effects were abrogated following the chemical silencing of TRPV1 sensory afferents, confirming their indispensable role in EA-mediated regulation of islet and PAT function. Furthermore, in TRPV1 knockout mice, a reduction in PAT inflammation was observed, along with the recovery of islet beta cell function. EA at LI11 and ST37 demonstrated anti-inflammatory properties and helped ameliorate IR.
CONCLUSION The PAT ecological niche influenced the progression from obesity to T2DM through various immunometabolic pathways. EA at ST25 could regulate glucolipid metabolism via the TRPV1-CGRP-islet beta cell pathway.
Collapse
Affiliation(s)
- Yun Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Xuan Wang
- College of Traditional Chinese Medicine, Jiangsu Vocational College of Medicine, Yancheng 224000, Jiangsu Province, China
| | - Ming-Qian Yuan
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Meng-Jiang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Mei-Rong Gong
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - You-Bing Xia
- Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Guan-Hu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH 45701, United States
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Gerhard Litscher
- High-Tech Acupuncture and Digital Chinese Medicine, Swiss University of Traditional Chinese Medicine, Bad Zurzach 5530, Switzerland
| | - Tian-Cheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
2
|
Shoyhet H, Bachinsky YH, Bekerman M, Debbi L, Ram GG, Safina D, Karnieli E, Levenberg S. Injectable skeletal muscle constructs overexpressing GLUT4 for type 2 diabetes intervention. Acta Biomater 2025; 197:216-225. [PMID: 40097128 DOI: 10.1016/j.actbio.2025.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
Skeletal muscle tissue engineering aims to repair tissue defects caused by injury, cancer, metabolic or neuromuscular disease. The need for invasive implantation techniques often limits the implantation of large tissue constructs or repeated treatments. Recent studies have reported on the development of injectable scaffolds for tissue engineering; however, fabrication of skeletal muscle tissue is particularly challenging due to the large size of human myotubes and the required mechanical properties. This work developed a collagen-based shape-memory scaffold supportive of skeletal muscle tissue growth and differentiation in vitro and maintained shape post-injection in vivo. The injectable engineered muscle construct was intramuscularly delivered via a syringe needle and integrated successfully with the native muscle tissue. We demonstrated the system's potential on a Type 2 diabetes mouse model. A prominent early sign of type 2 diabetes is the reduction in GLUT4 expression and translocation in skeletal muscle; therefore, based on a previous work published by our group, we created injectable GLUT4-overexpressing muscle constructs. Following injection, GLUT4 overexpressing skeletal muscle tissue retained its shape-memory properties and viability and improved glucose homeostasis in the diabetic mice. This work demonstrated successful minimally invasive delivery of engineered muscle tissue and potential treatment for chronic muscle-related conditions. STATEMENT OF SIGNIFICANCE: Type 2 diabetes is a widespread metabolic disorder characterized by insulin resistance and impaired glucose regulation. This study offers a minimally invasive approach to treatment through the development of an injectable skeletal muscle construct overexpressing GLUT4 to improve glucose homeostasis. Unlike traditional surgical methods, this minimally invasive system employs a collagen-based scaffold with shape-memory properties, enabling effective tissue delivery and integration. Existing therapies are limited in addressing chronic metabolic disorders that require repeated interventions. Our work fills that gap by enhancing muscle function and glucose regulation. The scaffold's unique ability to retain its structure post-injection and support muscle differentiation presents a significant advancement with broad implications for treating metabolic diseases and advancing regenerative medicine.
Collapse
Affiliation(s)
- Hagit Shoyhet
- Faculty of Biomedical Engineering, Technion, Israel; The Norman Seiden Multidisciplinary Graduate program in Nanotechnology & Nanoscience, Technion, Israel
| | | | | | - Lior Debbi
- Faculty of Biomedical Engineering, Technion, Israel
| | | | - Dina Safina
- Faculty of Biomedical Engineering, Technion, Israel
| | | | | |
Collapse
|
3
|
Mack KL, Landino NP, Tertyshnaia M, Longo TC, Vera SA, Crew LA, McDonald K, Phifer-Rixey M. Gene-by-environment Interactions and Adaptive Body Size Variation in Mice From the Americas. Mol Biol Evol 2025; 42:msaf078. [PMID: 40172935 PMCID: PMC12015161 DOI: 10.1093/molbev/msaf078] [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: 10/04/2024] [Revised: 02/14/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025] Open
Abstract
The relationship between genotype and phenotype is often mediated by the environment. Moreover, gene-by-environment (GxE) interactions can contribute to variation in phenotypes and fitness. In the last 500 yr, house mice have invaded the Americas. Despite their short residence time, there is evidence of rapid climate adaptation, including shifts in body size and aspects of metabolism with latitude. Previous selection scans have identified candidate genes for metabolic adaptation. However, environmental variation in diet as well as GxE interactions likely impact body mass variation in wild populations. Here, we investigated the role of the environment and GxE interactions in shaping adaptive phenotypic variation. Using new locally adapted inbred strains from North and South America, we evaluated response to a high-fat diet, finding that sex, strain, diet, and the interaction between strain and diet contributed significantly to variation in body size. We also found that the transcriptional response to diet is largely strain-specific, indicating that GxE interactions affecting gene expression are pervasive. Next, we used crosses between strains from contrasting climates to characterize gene expression regulatory divergence on a standard diet and on a high-fat diet. We found that gene regulatory divergence is often condition-specific, particularly for trans-acting changes. Finally, we found evidence for lineage-specific selection on cis-regulatory variation involved in diverse processes, including lipid metabolism. Overlap with scans for selection identified candidate genes for environmental adaptation with diet-specific effects. Together, our results underscore the importance of environmental variation and GxE interactions in shaping adaptive variation in complex traits.
Collapse
Affiliation(s)
- Katya L Mack
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, USA
| | - Nico P Landino
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | | | - Tiffany C Longo
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Sebastian A Vera
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Lilia A Crew
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Kristi McDonald
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Megan Phifer-Rixey
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
- Department of Biology, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
4
|
Panzeri I, Madaj Z, Fagnocchi L, Apostle S, Tompkins M, Hostetter G, Pospisilik JA. Chronic obesity does not alter cancer incidence in Trp53 R270H/+ mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.14.618190. [PMID: 39463991 PMCID: PMC11507782 DOI: 10.1101/2024.10.14.618190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Obesity is a complex chronic disease characterized by excessive adiposity and associations with numerous co-morbidities, including cancer. Despite extensive research, we have limited understanding of the mechanisms coupling obesity to cancer risk, and separately, of the contexts where obesity does or does not exacerbate disease. Here, we show that chronic high-fat diet induced obesity has no significant effect on the Trp53R270H/+ mouse, a model of human Li-Fraumeni multi-cancer syndrome. Surprisingly, despite inducing rapid and highly penetrant obesity and long-term differences in adiposity, greater than one year of HFD had no significant effect on survival or tumor burden. These findings were replicated in two separate cohorts totaling 359 mice and thus provide important negative data for the field. Given strong publication bias against negative data in the literature, this large murine cohort study represents a clear case where chronic diet-induced obesity does not accelerate or aggravate cancer outcomes. The data thus carry high impact for researchers, funders, and policymakers alike.
Collapse
Affiliation(s)
- Ilaria Panzeri
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Zachary Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Luca Fagnocchi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Stefanos Apostle
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Megan Tompkins
- Vivarium and Transgenics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, Van Andel Institute, Grand Rapids, MI, USA
| | | |
Collapse
|
5
|
Clarke GS, Li H, Heshmati E, Nicholas LM, Gatford KL, Page AJ. Pregnancy and a high-fat, high-sugar diet each attenuate mechanosensitivity of murine gastric vagal afferents, with no additive effects. J Physiol 2025; 603:1461-1481. [PMID: 40023799 PMCID: PMC11908482 DOI: 10.1113/jp286115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/03/2025] [Indexed: 03/04/2025] Open
Abstract
Gastric vagal afferents (GVAs) sense food-related mechanical stimuli and signal to the CNS to initiate meal termination. Pregnancy and diet-induced obesity are independently associated with dampened GVA mechanosensitivity and increased food intake. Whether a high-fat, high-sugar diet (HFHSD) impacts pregnancy-related adaptations in GVA signalling is unknown and was investigated in this study. Three-week-old female Glu Venus-expressing mice, on a C57BL/6 background, were fed standard laboratory diet (SLD) or HFHSD for 12 weeks, and then half of each group were mated to generate late pregnant (Day 17.5; P-SLD N = 12, P-HFHSD N = 14) or non-pregnant (NP-SLD N = 12, NP-HFHSD N = 16) groups. Body weight and food intake were monitored in Promethion metabolic cages from before mating until Day 17.5 of pregnancy or equivalent ages in non-pregnant mice, prior to tissue collection at 07.00 h for in vitro single fibre GVA recording and gene expression analysis. Pregnant mice gained more weight than non-pregnant mice but weight gain was unaffected by diet. By mid-pregnancy, light-phase food intake (kJ and g) was higher in pregnant than in non-pregnant mice (each P < 0.001) due to larger meals (kJ and g, each P < 0.001), irrespective of diet. Pregnancy and HFHSD-feeding reduced tension-sensitive GVA mechanosensitivity (each P < 0.01), but pregnancy did not further downregulate GVA stretch responses within HFHSD mice (P = 0.652). Nodose ganglia growth hormone receptor mRNA abundance was upregulated in pregnancy, possibly contributing to lower GVA mechanosensitivity during pregnancy in SLD mice. Larger light-phase meals in pregnant compared to non-pregnant HFHSD mice may therefore reflect the downregulation of other satiety pathways. KEY POINTS: Gastric vagal afferents (GVAs) regulate food intake by sensing the arrival and quantity of food and communicating this information to the brain. In standard laboratory diet (SLD) mice, gastric tension-sensitive vagal afferent mechanosensitivity was attenuated in pregnant compared to non-pregnant mice, which is concurrent with increases in total food intake and meal size. Nodose ganglia growth hormone receptor mRNA abundance was increased in pregnancy, possibly accounting for attenuated GVA mechanosensitivity in pregnant SLD mice. In non-pregnant mice, tension-sensitive GVA mechanosensitivity was selectively attenuated in high-fat, high-sugar diet (HFHSD) compared to SLD mice. Despite this, HFHSD mice ate less food and smaller meals compared to the SLD mice, suggesting other satiety mechanisms are limiting food intake. Despite higher food intake, there was no further reduction in mechanosensitivity in pregnant HFHSD mice compared to non-pregnant HFHSD mice and further studies are required to increase understanding of food intake regulation across pregnancy.
Collapse
Affiliation(s)
- Georgia S. Clarke
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Robinson Research InstituteThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research InstituteSAHMRIAdelaideSouth AustraliaAustralia
| | - Hui Li
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research InstituteSAHMRIAdelaideSouth AustraliaAustralia
| | - Elaheh Heshmati
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research InstituteSAHMRIAdelaideSouth AustraliaAustralia
| | - Lisa M. Nicholas
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Robinson Research InstituteThe University of AdelaideAdelaideSouth AustraliaAustralia
- Adelaide Centre for EpigeneticsThe University of AdelaideAdelaideSouth AustraliaAustralia
| | - Kathryn L. Gatford
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Robinson Research InstituteThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research InstituteSAHMRIAdelaideSouth AustraliaAustralia
| | - Amanda J. Page
- School of BiomedicineThe University of AdelaideAdelaideSouth AustraliaAustralia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research InstituteSAHMRIAdelaideSouth AustraliaAustralia
| |
Collapse
|
6
|
Seguella L, Corpetti C, Lu J, Pesce M, Franzin SB, Palenca I, Zilli A, Vincenzi M, Caprioli D, Paytuví-Gallart A, Sanseverino W, Rurgo S, Sarnelli G, Esposito G. Oleoylethanolamide-producing Lactobacillus paracasei F19 improves metabolic and behavioral disorders by restoring intestinal permeability and microbiota-gut-brain axis in high-fat diet-induced obese male mice. Brain Behav Immun 2025:S0889-1591(25)00053-4. [PMID: 39988008 DOI: 10.1016/j.bbi.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Metabolic and mood disorders elicited by chronic exposure of high-fat diet (HFD) are often associated with intestinal dysbiosis and persistent low-grade inflammation in the small intestine. This leads to remodeling of the epithelial barrier with disruption of the neuroepithelial circuits that control energy homeostasis by the gut-brain axis. Therefore, therapies that restoreintestinal microbial niche and barrier function are promising candidates to counter peripheral metabolic challenges that affect behaviors controlled by the brain. The endogenous oleoylethanolamine (OEA) was found to shape the intestinal microbiota profile towards a "lean-like phenotype", ameliorating pathological profiles of metabolic diseases. Further, OEA displays beneficial effects in several cognitive paradigms and preserves the epithelial barrier integrity, acting as an intestinal "gate-keeper". Here, we developed an "intestinal OEA factory" for the in-situ and controlled release of OEA by using a probiotic-based delivery system. We engineered the Lactobacillus paracasei F19 (LP) to express the human N-acylphosphatidylethanolamine-preferring phospholipase D (NAPEpld) gene and to produce OEA in response to dietary ultra-low oleate supply. We treated 12-week HFD male mice with oleate-probiotic formulations and assessed their impact on metabolic and behavioral dysfunctions, and microbiota-gut-brain signaling after 8 weeks of treatment. NAPE-expressing LP (pNAPE-LP) led to significant reduced weight loss and improved metabolic dysfunction in HFD-treated mice. Further, a parallel improvement in depressive- and anxiety-like phenotypes was associated with the duodenal barrier function retrieval, the restoration of the Firmicutes/Bacteroidetes ratio, and an increase in beneficial bacteria, such as Lactobacillus, Prevotella, and Parabacteroides. The HFD-driven changes both in the enteric and central nervous system were prevented by pNAPE-LP/oleate treatment. Collectively, our data suggest that these effects were mediated by the oleate-dependent release of OEA by pNAPE-LP since no significant effects were observed in HFD mice treated with the native probiotic alone (pLP). This oleate-regulated delivery system of OEA is a safe and efficient probiotic-based strategy for the treatment of metabolic syndrome and related behavioral disorders.
Collapse
Affiliation(s)
- Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Chiara Corpetti
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Jie Lu
- Department of Anatomy and Cell Biology, China Medical University, N0.77 Puhe Road, Shenbei New District, Shenyang City, Liaoning Province, PR China.
| | - Marcella Pesce
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Martina Vincenzi
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | - Daniele Caprioli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | | | - Walter Sanseverino
- Sequentia Biotech SL, Carrer del Dr. Trueta, 179, 08005 Barcelona, Spain.
| | - Sara Rurgo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| | - Giuseppe Esposito
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
7
|
Lee M, Son S, Oh S, Shin E, Shin H, Kwon O, Hwang S, Song H, Lim HJ. Diet-Induced Obesity Alters Granulosa Cell Transcriptome and Ovarian Immune Environment in Mice. Life (Basel) 2025; 15:330. [PMID: 40141675 PMCID: PMC11943477 DOI: 10.3390/life15030330] [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: 01/16/2025] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
Obesity affects female reproductive performance by impairing the ovarian and uterine environments. Using a diet-induced obesity mouse model, we examined whether a high-fat diet (HFD) regimen affects the gene expression profile in ovarian granulosa cells (GCs) and whether short-term HFD has similar effects on gene expression as long-term HFD. C57BL/6J mice were fed a HFD or normal diet (ND) for 16-18 weeks (long-term group) or 4 weeks (short-term group). GCs were collected from each group of mice for RNA-sequencing. RT-PCR and immunofluorescence staining were performed to validate the results. RNA-sequencing analyses of the GCs revealed that several immediate early genes, including early growth response 1 (Egr1), an important mediator of ovulation, were significantly downregulated in HFD GCs. Protein tyrosine phosphatase receptor type C (Ptprc) and hematopoietic type prostaglandin D synthase (Hpgds), both of which are associated with increased inflammation, were significantly upregulated in HFD GCs. Downregulation of Egr1 was also confirmed in the GCs of short-term HFD mice, suggesting that it constitutes an early change in response to a HFD. Increased expression of several transcription factors in HFD GCs suggests that a HFD may affect the overall transcriptional landscape. The results may indicate possible modulation of the immune environment in HFD ovaries. These results provide novel insights into the molecular changes in GCs in obese environments.
Collapse
Affiliation(s)
- Minseo Lee
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Sujin Son
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Surim Oh
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Eunbin Shin
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyejin Shin
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Ohrim Kwon
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Sohyun Hwang
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
- Department of Pathology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam-si 13520, Gyeonggi-do, Republic of Korea
| | - Haengseok Song
- Department of Life Science, Graduate School, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Hyunjung Jade Lim
- Department of Veterinary Medicine, School of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
- Department of Biomedical Science and Technology, Institute of Biomedical Science & Technology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
8
|
Fletcher E, Miserlis D, Papoutsi E, Steiner JL, Gordon B, Haynatzki G, Pacher P, Koutakis P. Chronic alcohol consumption exacerbates ischemia-associated skeletal muscle mitochondrial dysfunction in a murine model of peripheral artery disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167584. [PMID: 39581559 PMCID: PMC11931404 DOI: 10.1016/j.bbadis.2024.167584] [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: 08/22/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE Peripheral artery disease (PAD) causes ischemic mitochondriopathy-associated muscle damage, amplifying patient disability and mortality. Although alcohol and a high-fat diet enhance PAD predisposition and severity, their impact on PAD myopathy is unclear. Using our murine model of PAD, we investigated the combined effect of chronic alcohol and fat consumption on intramuscular oxidative stress and mitochondrial content, function, and quality control. The potential relationship between intramuscular aldehyde dehydrogenase 2 (ALDH2) content, oxidative stress and mitochondriopathy was also explored. METHODS Twenty-four male, 24 female, 8-month-old C57BL/6 J mice received high-fat-sucrose (HFS) or low-fat-sucrose (LFS) diets for 16-weeks, followed by either 20 % ethanol (EtOH) supplemented in the drinking water or continued water access for another 12-weeks (n = 12 mice/4 groups). The left femoral artery was ligated to induce hindlimb ischemia (HLI), and mice 4-weeks post-ligation were euthanized. RESULTS Chronic HLI was associated with an ischemic muscle mitochondriopathy, which was exacerbated by concurrent HFS and EtOH feeding. Intramuscular ALDH2 was also reduced in mice consuming HFS + EtOH, particularly in the ischemic limb, but increased in their LFS + EtOH-consuming counterparts. Moreover, reduced ALDH2 was strongly correlated with markers of oxidative stress and mitochondrial dysfunction. CONCLUSIONS ALDH2 could be a promising therapeutic target to optimize intramuscular mitochondrial function in PAD patients, particularly those who habitually consume a diet high in fat and alcohol.
Collapse
Affiliation(s)
- Emma Fletcher
- Department of Public Health, Usha Kundu MD College of Health, University of West Florida, Pensacola, FL, USA
| | - Dimitrios Miserlis
- Department of Surgery, University of Texas at Austin Dell Medical School, Austin, TX, USA
| | - Evlampia Papoutsi
- Department of Public Health, Usha Kundu MD College of Health, University of West Florida, Pensacola, FL, USA
| | - Jennifer L Steiner
- Department of Health, Nutrition and Food Sciences, Florida State University, 600 W. College Avenue, Tallahassee, FL 32306, USA; Institute of Sports Sciences and Medicine, Florida State University, 600 W. College Avenue, Tallahassee, FL 32306, USA
| | - Bradley Gordon
- Department of Health, Nutrition and Food Sciences, Florida State University, 600 W. College Avenue, Tallahassee, FL 32306, USA; Institute of Sports Sciences and Medicine, Florida State University, 600 W. College Avenue, Tallahassee, FL 32306, USA
| | - Gleb Haynatzki
- Department of Biostatistics, University of Nebraska Medical Center College of Public Health, 984375 Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Panagiotis Koutakis
- Department of Public Health, Usha Kundu MD College of Health, University of West Florida, Pensacola, FL, USA; Department of Biology, Baylor University, Waco, TX, USA.
| |
Collapse
|
9
|
Orellano MS, Scelza-Figueredo A, Lameroli Mauriz L, Sétula C, Argañarás M, Atorrasagasti C, Perone MJ, Andreone L. A dissociated glucocorticoid receptor modulator mitigates glucolipotoxicity in the endocrine pancreas and peripheral tissues: Preclinical data from a mouse model of diet-induced type 2 diabetes. Life Sci 2025; 362:123363. [PMID: 39761744 DOI: 10.1016/j.lfs.2024.123363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
AIMS Type 2 diabetes (T2D) is a prevalent metabolic disease linked to obesity and metabolic syndrome (MS). The glucolipotoxic environment (GLT) impacts tissues causing low-grade inflammation, insulin resistance and the gradual loss of pancreatic β-cell function, leading to hyperglycemia. We have previously shown that Compound A (CpdA), a plant-derived dissociative glucocorticoid receptor-modulator with inflammation-suppressive activity, displays protective effects on β-cells in type 1 diabetes murine models. This study aimed to evaluate whether the administration of CpdA can attenuate GLT effects and improve pathophysiological parameters in a murine model of T2D/MS. MAIN METHODS Eight-week-old male C57BL/6NCrl mice were fed either a standard chow diet or a high-fat/high-sucrose diet (HFHS) for 15 weeks. From week 5 of feeding, each group received i.p. injections of CpdA (2.5 μg/g) or vehicle three times a week. We also examined CpdA in vitro effect against GLT using the insulinoma cell line INS-1E and naïve isolated mouse islets. KEY FINDINGS CpdA administration in HFHS fed mice improved glucose homeostasis and insulin sensitivity with no apparent side effects. CpdA treatment also preserved pancreatic islet architecture and insulin expression, while reducing hepatic steatosis and visceral adipose tissue inflammation induced by HFHS diet. In vitro assays in INS-1E cells and naïve isolated mouse islets demonstrated that CpdA counteracted GLT-induced inhibition of glucose-stimulated insulin secretion and supported the expression of key β-cell identity genes under GLT conditions. SIGNIFICANCE These findings highlight the potential protective effect of CpdA in preserving β-cell functionality and peripheral tissue physiology in the context of T2D/MS.
Collapse
Affiliation(s)
- Miranda Sol Orellano
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina; Facultad de Ciencias Biomédicas, , Universidad Austral, Pilar, Argentina
| | - Andrea Scelza-Figueredo
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina; Facultad de Ciencias Biomédicas, , Universidad Austral, Pilar, Argentina
| | - Lucía Lameroli Mauriz
- Facultad de Ciencias Biomédicas, , Universidad Austral, Pilar, Argentina; Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina
| | - Carolina Sétula
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina; Facultad de Ciencias Biomédicas, , Universidad Austral, Pilar, Argentina
| | - Milagros Argañarás
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina
| | - Catalina Atorrasagasti
- Facultad de Ciencias Biomédicas, , Universidad Austral, Pilar, Argentina; Experimental Hepatology and Gene Therapy Program, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina
| | - Marcelo Javier Perone
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina; Facultad de Ciencias Biomédicas, , Universidad Austral, Pilar, Argentina
| | - Luz Andreone
- Immuno-Endocrinology, Diabetes & Metabolism Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET - Universidad Austral, Pilar, Argentina; Facultad de Ciencias Biomédicas, , Universidad Austral, Pilar, Argentina.
| |
Collapse
|
10
|
Peppi PF, Lira GA, Campos LRS, Santos CR, Lima EMM, Barreto-Vianna ARC. Liver under attack: impacts of high-fat diet on murine model. BRAZ J BIOL 2025; 84:e284045. [PMID: 39907329 DOI: 10.1590/1519-6984.284045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/06/2024] [Indexed: 02/06/2025] Open
Abstract
At present, non-alcoholic fatty liver disease (NAFLD) is the most prevalent liver disease worldwide, with obesity recognized as a global epidemic and type 2 diabetes a worldwide disease. In this study, 10 C57BL/6 mice were divided into two groups: the control group (SC) and the high-fat diet (HF) group. Both groups were fed their respective diets for 8 weeks. The animals were analyzed for body weight, glucose/insulin resistance, hepatic steatosis, and fibrosis to diagnose NAFLD. Results showed that the HF group animals had significantly higher body weight (P<0.0001), glucose resistance (P=0.0002), insulin resistance (P=0.0009), and blood glucose levels (P<0.05) compared to the SC group. The HF group exhibited increased hepatic steatosis (P<0.0001) and fibrosis (P<0.0001) compared to the SC group. These findings led to the conclusion that the animals in the HF group had grade and stage 2 NAFLD. Furthermore, the HF group animals were classified as obese, indicating a higher risk for developing insulin resistance and, subsequently, type 2 diabetes mellitus (T2DM). Understanding the risk factors and complications associated with NAFLD, obesity, and T2DM is crucial for preventing and treating metabolic alterations linked to a high-fat diet.
Collapse
Affiliation(s)
- P F Peppi
- Universidade Federal do Paraná - UFPR, Departamento de Biociências, Palotina, PR, Brasil
| | - G A Lira
- Universidade de Brasília - UnB, Programa de Pós-Graduação em Ciências Animais, Brasília, DF, Brasil
| | - L R S Campos
- Universidade de Brasília - UnB, Programa de Pós-Graduação em Ciências Animais, Brasília, DF, Brasil
| | - C R Santos
- Universidade de Brasília - UnB, Programa de Pós-Graduação em Ciências Animais, Brasília, DF, Brasil
| | - E M M Lima
- Universidade de Brasília - UnB, Programa de Pós-Graduação em Ciências Animais, Brasília, DF, Brasil
| | - A R C Barreto-Vianna
- Universidade Federal do Paraná - UFPR, Departamento de Biociências, Palotina, PR, Brasil
| |
Collapse
|
11
|
Kim EJ, Lee SH, Kim TH, Lee J, Choi CH, Lee SJ. Insect chitosan derived from Hermetia illucens larvae suppresses adipogenic signaling and promotes the restoration of gut microbiome balance. Int J Biol Macromol 2025; 284:138168. [PMID: 39613084 DOI: 10.1016/j.ijbiomac.2024.138168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
Chitosan, the deacetylated form of chitin, is considered a valuable source of compounds in the feed and food industries. However, the impact of Hermetia illucens larvae chitosan (HCS) with specific physicochemical characteristics on obesity mediated by lipid accumulation and microbiome dysbiosis has not been fully elucidated. We purified HCS with a low molecular weight (84 kDa), low crystallinity, and a high deacetylation rate, characterizing it through several analytical techniques, including gel permeation chromatography, FT-IR, 1H NMR, FE-SEM, and XRD analysis. HCS effectively inhibited the differentiation of 3T3-L1 preadipocytes by suppressing the production of reactive oxygen species. The adipogenic signaling of preadipocytes, mediated by the phosphorylation of mTOR and PPARγ, which are essential for the expression of fatty acid synthase, was attenuated by HCS. In mouse models fed high-fat diets, the oral administration of HCS prevented changes in white adipose tissue and liver weight and reduced plasma levels of total cholesterol. Additionally, the analysis of the microbiota using 16S rRNA revealed that HCS improved dysbiosis by modulating the composition and abundance of specific bacterial genera, including F. rodentium, L. gasseri, L. reuteri, and L. murinus. These findings highlight the potential of HCS as a candidate for the treatment of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Eun-Ju Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Seok-Hui Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Tae Hoon Kim
- FoodyWorm, Co. Ltd., 76 Geumwangtekeuno 3-gil, Geumwang-eup, Eumseong-gun, Chungcheongbuk-do 27680, Republic of Korea
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Chang-Hyung Choi
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea.
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
12
|
Banerjee M, Pandey VP. Diet-induced Obesity: Pathophysiology, Consequences and Target Specific Therapeutic Strategies. Curr Protein Pept Sci 2025; 26:113-124. [PMID: 39225225 DOI: 10.2174/0113892037329528240827180820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/09/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024]
Abstract
Diet has emerged as a pivotal factor in the current time for diet-induced obesity (DIO). A diet overloaded with fats and carbohydrates and unhealthy dietary habits contribute to the development of DIO through several mechanisms. The prominent ones include the transition of normal gut microbiota to obese microbiota, under-expression of AMPK, and abnormally high levels of adipogenesis. DIO is the root of many diseases. The present review deals with various aspects of DIO and its target proteins that can be specifically used for its treatment. Also, the currently available treatment strategies have been explored. It was found that the expression of five proteins, namely, PPARγ, FTO, CDK4, 14-3-3 ζ protein, and Galectin-1, is upregulated in DIO. They can be used as potential targets for drug-designing studies. Thus, with these targets, the treatment strategy for DIO using natural bioactive compounds can be a safer alternative to medications and bariatric surgeries.
Collapse
Affiliation(s)
- Munmun Banerjee
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| | - Veda P Pandey
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
- Institute of Food Processing and Technology, ONGC Centre for Advanced Studies, University of Lucknow, Lucknow 226007, India (Pesent Address)
| |
Collapse
|
13
|
Ormiston K, Melink Z, Andridge R, Lustberg M, Courtney DeVries A, Murphy K, Emmers K, Ziouzenkova O, Belury MA, Orchard TS. Dietary EPA and DHA enrichment of a high fat diet during doxorubicin-based chemotherapy attenuated neuroinflammatory gene expression in the brain of C57bl/6 ovariectomized mice. Brain Behav Immun 2025; 123:370-382. [PMID: 39313165 DOI: 10.1016/j.bbi.2024.09.021] [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: 05/20/2024] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
Chemotherapy agents in breast cancer are associated with chemotherapy-related cognitive impairments (CRCI). Mechanisms are not fully clear, but alterations of glucose and lipid metabolism, neuroinflammation and neurodegeneration may contribute to CRCI. The aim of this study was to investigate the combined effects of a high fat (HF) diet combined with doxorubicin-based chemotherapy on glucose and lipid metabolism, neuroinflammation, and neurodegeneration in mice. Additionally, we examined the therapeutic potential of dietary eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) to attenuate these effects. Female C57Bl/6 mice (n = 42) were fed HF, HFn-3 (2 % kcals as EPA + DHA) or Low Fat (LF) diets for seven weeks, with and without chemotherapy. In this study, two chemotherapy injections led to weight and body fat loss associated with a decrease in insulin resistance measured by HOMA-IR. HOMA-IR was significantly greater in HF versus LF groups; but HOMA-IR in HFn-3 group did not significantly differ from either HF or LF groups. Chemotherapy resulted in higher brain concentrations of the inflammatory chemokine KC/GRO. Compared to LF diet plus chemotherapy, HF diet plus chemotherapy upregulated multiple genes involved in neuroinflammation and neurodegeneration pathways. HFn-3 diet plus chemotherapy attenuated gene expression by downregulating multiple genes involved in neuroinflammation and blood brain barrier regulation, including Mapkapk2, Aqp4, and s100b, and upregulating Kcnb1 and Atxn3, genes involved in reduction of oxidative stress and anxiety, respectively. Overall, a HF diet combined with chemotherapy is associated with neuroinflammatory and neurodegenerative gene expression changes in this mouse model; dietary enrichment of EPA and DHA attenuated these effects. Further studies are needed to understand how diet impacts behavioral outcomes of CRCI.
Collapse
Affiliation(s)
- Kate Ormiston
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Zihan Melink
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Rebecca Andridge
- Division of Biostatistics, College of Public Health, The Ohio State University, United States
| | | | | | - Kelly Murphy
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Katie Emmers
- Department of Veterinary Medicine, The Ohio State University, United States
| | - Ouliana Ziouzenkova
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States
| | - Martha A Belury
- Department of Food Science and Technology, The Ohio State University, United States
| | - Tonya S Orchard
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, United States.
| |
Collapse
|
14
|
Nomura M, Murad NF, Madhavan SS, Mu WC, Eap B, Garcia TY, Aguirre CG, Verdin E, Ellerby L, Furman D, Newman JC. Ketogenic Diet Reduces Age-Induced Chronic Neuroinflammation in Mice. AGING BIOLOGY 2024; 2:20240038. [PMID: 39697898 PMCID: PMC11654834 DOI: 10.59368/agingbio.20240038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
The ketone body beta-hydroxybutyrate (BHB) is an acidic energy metabolite that is synthesized during periods of fasting or exercise. Our previous study demonstrated that an every other week cyclic ketogenic diet (Cyclic KD), which induces blood BHB levels similar to those observed during fasting, reduces midlife mortality and improves memory in aging mice. In addition to its canonical role as an energy metabolite, BHB regulates gene expression and inflammatory activation through non-energetic signaling pathways. The precise mechanisms by which BHB or KD affects brain function during aging remain incompletely understood. Using bulk RNA-sequencing (RNA-Seq), we examined whole brain gene expression of 12-month-old C57BL/6JN male mice fed KD for either one week or 14 months. While one-week KD increases some inflammatory gene expression, the 14-month Cyclic KD largely reduces age-induced neuroinflammatory gene expression. Next, a gene expression analysis of human primary brain cells (microglia, astrocytes, and neurons) using RNA-Seq revealed that BHB alone induces a mild level of inflammation in all three cell types. However, BHB inhibits the more pronounced inflammatory gene expression induced by lipopolysaccharide (LPS) in microglia. BHB exhibits a comparable inhibitory effect on LPS-induced inflammation in mouse primary microglia, which we used as an in vitro model to test and exclude known mechanisms by which BHB regulates inflammation and gene expression as responsible for this modulation of LPS-induced inflammatory gene expression. An acidic milieu resulting from BHB may be required for or contribute to the effect. Overall, we observe that BHB has the potential to attenuate the microglial response to inflammatory stimuli, such as LPS. This may contribute to an observed reduction in chronic inflammation in the brain following long-term Cyclic KD treatment in aging mice.
Collapse
Affiliation(s)
| | | | - Sidharth S. Madhavan
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Wei-Chieh Mu
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Brenda Eap
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | - Carlos Galicia Aguirre
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Lisa Ellerby
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA
- Instituto de Investigaciones en Medicina Traslacional, Universidad Austral, Consejo Nacional de Investigaciones Científicas y Técnicas, 1629, Pilar, Argentina
| | - John C. Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Division of Geriatrics, University of California, San Francisco, CA, USA
| |
Collapse
|
15
|
Hewady S, Manuel CR, Pasquali C, Koya J, Reznik SE. OM-85 attenuates high-fat diet-induced obesity, insulin resistance, gut dysbiosis and nonalcoholic steatohepatitis in a murine model. Biomed Pharmacother 2024; 181:117710. [PMID: 39626377 DOI: 10.1016/j.biopha.2024.117710] [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: 08/25/2024] [Revised: 11/04/2024] [Accepted: 11/23/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Obesity is a global epidemic that is tied to a wide range of human disorders. Chronic consumption of a high-fat diet is linked to disruption of the intestinal microbiome, which drives obesity-related pathophysiology. Broncho-Vaxom® (OM-85), a bacterial lysate used for prophylaxis of recurrent respiratory tract infections, has both immunostimulatory and immunomodulatory functions. METHODS Male C57Bl/6 mice were maintained on normal control vs. high-fat diets for 8 weeks and treated or untreated with OM-85 or with the probiotic Lactobacillus plantarum, as a positive control. Mice were evaluated for weight gain, glucose tolerance, insulin tolerance, gut microbiome composition and non-alcoholic steatohepatitis (NASH). RESULTS High-fat diet mice developed obesity, insulin resistance, NASH and gut dysbiosis with a shift from the Bacteroidetes phylum, such as Bacteroidales order and Muribaculaceae family organisms to Firmicutes groups, such as the Clostridium and Blautia genuses. Treatment with OM-85 led to 1) prevention of obesity, 2) prevention of insulin resistance, 3) attenuation of NASH and 4) attenuation of gut dysbiosis, with decreased levels of the organisms mentioned above and increases in Verrucomicrobiae phylum organisms such as Akkermansia family microbes as well as Muribaculaceae organisms. These shifts in the gut microbiome predict favorable effects on the short chain fatty acid profile in the gut and increased integrity of the intestinal barrier. Pathway analysis showed that OM-85 decreases rates of carbohydrate metabolism, providing an additional mechanism whereby OM-85 prevents obesity. CONCLUSION Immune modulators such as OM-85 should be investigated for their potential therapeutic effects on metabolism.
Collapse
Affiliation(s)
- Sarah Hewady
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA
| | | | | | | | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY, USA; Departments of Pathology and Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
16
|
Huwart SJP, Fayt C, Gangarossa G, Luquet S, Cani PD, Everard A. TLR4-dependent neuroinflammation mediates LPS-driven food-reward alterations during high-fat exposure. J Neuroinflammation 2024; 21:305. [PMID: 39580436 PMCID: PMC11585241 DOI: 10.1186/s12974-024-03297-z] [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: 08/20/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Obesity has become a global pandemic, marked by significant shifts in both the homeostatic and hedonic/reward aspects of food consumption. While the precise causes are still under investigation, recent studies have identified the role of gut microbes in dysregulating the reward system within the context of obesity. Unravelling these gut-brain connections is crucial for developing effective interventions against eating and metabolic disorders, particularly in the context of obesity. This study explores the causal role of LPS, as a key relay of microbiota component-induced neuroinflammation in the dysregulation of the reward system following exposure to high-fat diet (HFD). METHODS Through a series of behavioural paradigms related to food-reward events and the use of pharmacological agents targeting the dopamine circuit, we investigated the mechanisms associated with the development of reward dysregulation during HFD-feeding in male mice. A Toll-like receptor 4 (TLR4) full knockout model and intraventricular lipopolysaccharide (LPS) diffusion at low doses, which mimics the obesity-associated neuroinflammatory phenotype, were used to investigate the causal roles of gut microbiota-derived components in neuroinflammation and reward dysregulation. RESULTS Our study revealed that short term exposure to HFD (24 h) tended to affect food-seeking behaviour, and this effect became significant after 1 week of HFD. Moreover, we found that deletion of TLR4 induced a partial protection against HFD-induced neuroinflammation and reward dysregulation. Finally, chronic brain diffusion of LPS recapitulated, at least in part, HFD-induced molecular and behavioural dysfunctions within the reward system. CONCLUSIONS These findings highlight a link between the neuroinflammatory processes triggered by the gut microbiota components LPS and the dysregulation of the reward system during HFD-induced obesity through the TLR4 pathway, thus paving the way for future therapeutic approaches.
Collapse
Affiliation(s)
- Sabrina J P Huwart
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium
| | - Clémence Fayt
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium
| | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, F-75013, France
- Institut Universitaire de France (IUF), Paris, France
| | - Serge Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris, F-75013, France
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Av. E. Mounier, 73 Box B1.73.11, Brussels, B-1200, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium.
| |
Collapse
|
17
|
Laoung-on J, Anuduang A, Saenjum C, Srichairatanakool S, Boonyapranai K, Ounjaijean S. Pharmacological Activity of Cha-Miang ( Camellia sinensis var. assamica) in High Fat Diet-Induced Insulin-Resistant Rats. Life (Basel) 2024; 14:1515. [PMID: 39598313 PMCID: PMC11595819 DOI: 10.3390/life14111515] [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: 10/15/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
Obesity, a major global health concern, is represented by an accumulation of adipose tissue and body mass, leading to a higher incidence of non-communicable diseases (NCDs). Camellia sinensis var. assamica, known as cha-miang, contains catechin and its derivatives, which have been reported to affect several health-related concerns such as anti-cancer, anti-inflammatory, anti-hyperlipidemia, and against cardiovascular disease. The research aimed to examine the anti-obesity and insulin resistance effects of fresh cha-miang extract (CME) and fermented cha-miang extract (FCME), and to investigate the anti-obesity and anti-diabetic effects of CME and FCME in obese rats generated by a high-fat diet. The extracts demonstrated significant antioxidant potential, with CME demonstrating greater DPPH radical scavenging activity, whereas FCME excelled in ABTS radical scavenging. In the animal model, CME and FCME significantly reduced body weight, plasma insulin levels, insulin resistance, and accumulation of fat compared to the obese control group. Moreover, plasma biochemical analysis indicated that both extracts enhanced lipid profiles by reducing cholesterol, triglycerides, and LDL-cholesterol levels, while elevating HDL-cholesterol. Histological analysis revealed decreased hepatic fat accumulation, especially when extracts were concurrently treated with metformin. The research indicates that CME and FCME, especially in conjunction with metformin, have potential anti-obesity and anti-insulin resistance benefits attributed to their abundant polyphenolic content and antioxidant characteristics. This indicates that cha-miang may serve as an effective option for the management of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Jiraporn Laoung-on
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand;
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (A.A.); (K.B.)
| | - Artorn Anuduang
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (A.A.); (K.B.)
| | - Chalermpong Saenjum
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Somdet Srichairatanakool
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Kongsak Boonyapranai
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (A.A.); (K.B.)
| | - Sakaewan Ounjaijean
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (A.A.); (K.B.)
| |
Collapse
|
18
|
Barranco MM, Zecchinati F, Perdomo VG, Habib MJ, Rico MJ, Rozados VR, Salazar M, Fusini ME, Scharovsky OG, Villanueva SSM, Mainetti LE, García F. Intestinal ABC transporters: Influence on the metronomic cyclophosphamide-induced toxic effect in an obese mouse mammary cancer model. Toxicol Appl Pharmacol 2024; 492:117130. [PMID: 39426530 DOI: 10.1016/j.taap.2024.117130] [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: 01/18/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Metronomic chemotherapy (MCT) is a cancer therapeutic approach characterized by low dose drug chronic administration and limited or null toxicity. Obesity-induced metabolic alterations worsen cancer prognosis and influence the intestinal biochemical barrier, altering the Multidrug resistance-associated protein 2 (Mrp2) and Multidrug resistance protein-1 (Mdr-1), efflux pumps that transport chemotherapeutic drugs. Obesity and cancer are frequent co-morbidities; thus, our aim was to evaluate the effectiveness and toxicity of MCT with cyclophosphamide (Cy) in obese mice with metabolic alterations bearing a mammary adenocarcinoma. Simultaneously, the expression and activities of intestinal Mrp2 and Mdr-1 were assessed. CBi male mice, were fed with chow diet (C) or diet with 40 % of fat (HFD). After 16 weeks, metabolic alterations were confirmed by biochemical and morphological parameters. At that time-point, HFD group showed decreased expressions of Mrp2 mRNA (53 %) as well as Mdr-1a and Mdr-1b (42 % and 59 %, respectively), compared to C (P < 0.05). This result correlated with decreased intestinal Mrp2 and Mdr-1 efflux activities (64 % and 45 %, respectively), compared to C (P < 0.05). Ultimately, mice were challenged with M-406 mammary adenocarcinoma; when the tumor was palpable, mice were distributed into 4 groups. The % inhibition of tumor growth with Cy (30 mg/kg/day) in C + Cy was higher than that of HFD + Cy (P = 0.052). Besides, it was observed a 21 % diminution in body weight and leukopenia in the HFD + Cy group. Conclusion: Obesity-induced metabolic alterations impair intestinal Mrp2 and Mdr-1 functions, bringing about increments in Cy absorption, leading to toxicity; in addition, the antitumor effectiveness of MCT decreased in obese animals.
Collapse
MESH Headings
- Animals
- Cyclophosphamide/toxicity
- Mice
- Obesity/metabolism
- Male
- Female
- Administration, Metronomic
- Multidrug Resistance-Associated Protein 2
- Antineoplastic Agents, Alkylating/toxicity
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/chemically induced
- Mice, Obese
- Multidrug Resistance-Associated Proteins/metabolism
- Multidrug Resistance-Associated Proteins/genetics
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Adenocarcinoma/pathology
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP-Binding Cassette Transporters/metabolism
- ATP-Binding Cassette Transporters/genetics
- Diet, High-Fat
Collapse
Affiliation(s)
- María Manuela Barranco
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina
| | - Felipe Zecchinati
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Fisiología Experimental-CONICET. Rosario, Santa Fe, Argentina
| | - Virginia Gabriela Perdomo
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Martín José Habib
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - María José Rico
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Viviana Rosa Rozados
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Mario Salazar
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Laboratorio de Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Matías Ezequiel Fusini
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Olga Graciela Scharovsky
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CIC-UNR, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | | | - Leandro Ernesto Mainetti
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Fabiana García
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina..
| |
Collapse
|
19
|
Mitra A, Das A, Ghosh S, Sarkar S, Bandyopadhyay D, Gangopadhyay S, Chattopadhyay S. Metformin instigates cellular autophagy to ameliorate high-fat diet-induced pancreatic inflammation and fibrosis/EMT in mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167313. [PMID: 38901652 DOI: 10.1016/j.bbadis.2024.167313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Chronic pancreatic dysfunction is frequently observed as a consequence of prolonged high-fat diet consumption and is a serious public health concern. This pro-diabetic insult aggravates inflammation-influenced fibrotic lesions and is associated with deregulated autophagy. Metformin, a conventional anti-hyperglycemic drug, might be beneficial for pancreatic health, but the complex molecular regulations are not clarified. Considering the worldwide prevalence of chronic pancreatic dysfunction in obese individuals, we aimed to unwind the molecular intricacies explaining the involvement of oxidative stress, inflammation and fibrosis and to approbate metformin as a plausible intervention in this crossroad. MAIN METHODS Age-matched Swiss Albino mice were exposed to high-fat diet (60 kcal%) against control diet (10 kcal%) to establish diet-induced stress model. Metformin treatment was introduced after 4 weeks to metformin-control and HFD-exposed metformin groups. After 8 weeks, metabolic and molecular outcomes were assessed to establish the impact of metformin on chronic consequences of HFD-mediated injury. KEY FINDINGS High-fat diet administration to healthy mice primes oxidative stress-mediated chronic inflammation through Nrf2/Keap1/NF-κB interplay. Besides, pro-inflammatory cytokine bias leading to fibrotic (increased TGF-β, α-SMA, and MMP9) and pro-EMT (Twist1, Slug, Vimentin, E-cadherin) repercussions in pancreatic lobules were evident. Metformin distinctly rescues high-fat diet-induced remodeling of pancreatic pro-diabetic alterations and cellular survival/death switch. Further, metformin abrogates the p62-Twist1 crosstalk in an autophagy-dependent manner (elevated beclin1, LC3-II/I, Lamp2) to restore pancreatic homeostasis. CONCLUSION Our research validates the therapeutic potential of metformin in the inflammation-fibrosis nexus to ameliorate high-fat diet-induced pancreatic dysfunction and related metabolic alterations.
Collapse
Affiliation(s)
- Ankan Mitra
- Department of Physiology, University College of Science, Technology and Agriculture, University of Calcutta, 92, APC Road, Kolkata 700009, West Bengal, India
| | - Ankur Das
- Department of Physiology, University College of Science, Technology and Agriculture, University of Calcutta, 92, APC Road, Kolkata 700009, West Bengal, India
| | - Sourav Ghosh
- Department of Physiology, University College of Science, Technology and Agriculture, University of Calcutta, 92, APC Road, Kolkata 700009, West Bengal, India
| | - Swaimanti Sarkar
- Department of Physiology, University College of Science, Technology and Agriculture, University of Calcutta, 92, APC Road, Kolkata 700009, West Bengal, India
| | - Debasish Bandyopadhyay
- Department of Physiology, University College of Science, Technology and Agriculture, University of Calcutta, 92, APC Road, Kolkata 700009, West Bengal, India
| | - Somnath Gangopadhyay
- Department of Physiology, University College of Science, Technology and Agriculture, University of Calcutta, 92, APC Road, Kolkata 700009, West Bengal, India
| | - Sreya Chattopadhyay
- Department of Physiology, University College of Science, Technology and Agriculture, University of Calcutta, 92, APC Road, Kolkata 700009, West Bengal, India; Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Kolkata 700098, West Bengal, India.
| |
Collapse
|
20
|
Mackey-Alfonso SE, Butler MJ, Taylor AM, Williams-Medina AR, Muscat SM, Fu H, Barrientos RM. Short-term high fat diet impairs memory, exacerbates the neuroimmune response, and evokes synaptic degradation via a complement-dependent mechanism in a mouse model of Alzheimer's disease. Brain Behav Immun 2024; 121:56-69. [PMID: 39043341 DOI: 10.1016/j.bbi.2024.07.021] [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: 04/01/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disease characterized by profound memory impairments, synaptic loss, neuroinflammation, and hallmark pathological markers. High-fat diet (HFD) consumption increases the risk of developing AD even after controlling for metabolic syndrome, pointing to a role of the diet itself in increasing risk. In AD, the complement system, an arm of the immune system which normally tags redundant or damaged synapses for pruning, becomes pathologically overactivated leading to tagging of healthy synapses. While the unhealthy diet to AD link is strong, the underlying mechanisms are not well understood in part due to confounding variables associated with long-term HFD which can independently influence the brain. Therefore, we experimented with a short-term diet regimen to isolate the diet's impact on brain function without causing obesity. This project investigated the effect of short-term HFD on 1) memory, 2) neuroinflammation including complement, 3) AD pathology markers, 4) synaptic markers, and 5) in vitro microglial synaptic phagocytosis in the 3xTg-AD mouse model. Following the consumption of either standard chow or HFD, 3xTg-AD and non-Tg mice were tested for memory impairments. In a separate cohort of mice, levels of hippocampal inflammatory markers, complement proteins, AD pathology markers, and synaptic markers were measured. For the last set of experiments, BV2 microglial phagocytosis of synapses was evaluated. Synaptoneurosomes isolated from the hippocampus of 3xTg-AD mice fed chow or HFD were incubated with equal numbers of BV2 microglia. The number of BV2 microglia that phagocytosed synaptoneurosomes was tracked over time with a live-cell imaging assay. Finally, we incubated BV2 microglia with a complement receptor inhibitor (NIF) and repeated the assay. Behavioral analysis showed 3xTg-AD mice had significantly impaired long-term contextual and cued fear memory compared to non-Tg mice that was further impaired by HFD. HFD significantly increased inflammatory markers and complement expression while decreasing synaptic marker expression only in 3xTg-AD mice, without altering AD pathology markers. Synaptoneurosomes from HFD-fed 3xTg-AD mice were phagocytosed at a significantly higher rate than those from chow-fed mice, suggesting the synapses were altered by HFD. The complement receptor inhibitor blocked this effect in a dose-dependent manner, demonstrating the HFD-mediated increase in phagocytosis was complement dependent. This study indicates HFD consumption increases neuroinflammation and over-activates the complement cascade in 3xTg-AD mice, resulting in poorer memory. The in vitro data point to complement as a potential mechanistic culprit and therapeutic target underlying HFD's influence in increasing cognitive vulnerability to AD.
Collapse
Affiliation(s)
- Sabrina E Mackey-Alfonso
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Michael J Butler
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA
| | - Ashton M Taylor
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | | | - Stephanie M Muscat
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Ruth M Barrientos
- Institute for Behavioral Medicine Research, Ohio State University, Columbus, OH, USA; Department of Psychiatry and Behavioral Health, Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
21
|
Meyers AM, Gnazzo FG, Barrera ED, Nabatian T, Chan L, Beeler JA. DIETARY REGULATION OF SILENT SYNAPSES IN THE DORSOLATERAL STRIATUM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.24.586457. [PMID: 38585967 PMCID: PMC10996560 DOI: 10.1101/2024.03.24.586457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Obesity and drugs of abuse share overlapping neural circuits and behaviors. Silent synapses are transient synapses that are important for remodeling brain circuits. They are prevalent during early development but largely disappear by adulthood. Drugs of abuse increase silent synapses during adulthood and may facilitate reorganizing brain circuits around drug-related experience, facilitating addiction and contributing to relapse during treatment and abstinence. Whether obesity causes alterations in the expression of silent synapses in a manner similar to drugs of abuse has not been examined. Using a dietary-induced obesity paradigm, mice that chronically consumed high fat diet (HFD) exhibited increased silent synapses in both direct and indirect pathway medium spiny neurons in the dorsolateral striatum. Both the time of onset of increased silent synapses and their normalization upon discontinuation of HFD occurs on an extended time scale compared to drugs of abuse. These data demonstrate that chronic consumption of HFD, like drugs of abuse, can alter mechanisms of circuit plasticity likely facilitating neural reorganization analogous to drugs of abuse.
Collapse
Affiliation(s)
- Allison M Meyers
- Psychology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Federico G Gnazzo
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Eddy D Barrera
- Biology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Tikva Nabatian
- Cognitive Neuroscience MS program, Graduate Center, City University of New York, New York, NY, USA
| | - Larry Chan
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
| | - Jeff A Beeler
- Department of Psychology, Queens College, City University of New York, Flushing, NY, USA
- Psychology Program, The Graduate Center, City University of New York, New York, NY, USA
- Biology Program, The Graduate Center, City University of New York, New York, NY, USA
- Cognitive Neuroscience MS program, Graduate Center, City University of New York, New York, NY, USA
| |
Collapse
|
22
|
Chen X, Bushman T, Lin TY, Fu Q, Zhang S. Serum Extracellular Vesicles Reveal Metabolic Responses to Time-Restricted Feeding in High Fat Diet-Induced Obesity in Male Mice. RESEARCH SQUARE 2024:rs.3.rs-4745029. [PMID: 39399666 PMCID: PMC11469403 DOI: 10.21203/rs.3.rs-4745029/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Objective Extracellular vesicle (EV) secretion and cargo composition are dysregulated in metabolic diseases. This study aimed to identify changes in the EV size profile and protein cargoes in diet-induced obesity following time-restricted feeding (TRF) and to establish the role of EVs in obesity-related metabolic responses. Methods Mice were fed a high-fat diet (HFD) for 18 weeks prior to being placed either ad libitum or a time-restricted feeding for an additional 10 weeks. Mice on a normal chow ad libitum served as the control. The TRF group had food available for 10 hours and fasted for 14 hours per day. Results The serum EV size profile and amount displayed sex- and age-dependent changes in HFD-induced obesity, with age reducing EV amounts. HFD decreased small EV populations and increased larger EV populations, while TRF reversed these changes. Quantitative proteomic analysis showed that the abundance and composition of EV proteins changed in response to both acute stimulation with lipopolysaccharides (LPS) and HFD. Gene ontology analysis identified specific sets of EV proteins and their involved biological processes, reflecting the effect of LPS and HFD, as well as the reversal effect of TRF on metabolic and inflammatory pathways. EV proteins altered by HFD and those reversed by TRF had low protein overlap but significant functional overlap in biological processes. TRF activated the PPAR signaling pathway and the AKT-mTOR signaling pathway. The most significant impacts of HFD and TRF were observed on lipoprotein and carbohydrate metabolism, complement system, and neutrophil degranulation. The reversal effect of TRF on the complement system was pathway-specific, significantly activating the lectin complement pathway and restoring neutrophil degranulation. Conclusion Our data indicate that EVs are involved in diet-induced metabolic and inflammatory responses. Different EV populations may carry distinct sets of proteins involved in specific biological processes, thereby regulating diverse metabolic pathways efficiently.
Collapse
|
23
|
Gargiulo S, Barone V, Bonente D, Tamborrino T, Inzalaco G, Gherardini L, Bertelli E, Chiariello M. Integrated Ultrasound Characterization of the Diet-Induced Obesity (DIO) Model in Young Adult c57bl/6j Mice: Assessment of Cardiovascular, Renal and Hepatic Changes. J Imaging 2024; 10:217. [PMID: 39330437 PMCID: PMC11433005 DOI: 10.3390/jimaging10090217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Consuming an unbalanced diet and being overweight represent a global health problem in young people and adults of both sexes, and may lead to metabolic syndrome. The diet-induced obesity (DIO) model in the C57BL/6J mouse substrain that mimics the gradual weight gain in humans consuming a "Western-type" (WD) diet is of great interest. This study aims to characterize this animal model, using high-frequency ultrasound imaging (HFUS) as a complementary tool to longitudinally monitor changes in the liver, heart and kidney. Long-term WD feeding increased mice body weight (BW), liver/BW ratio and body condition score (BCS), transaminases, glucose and insulin, and caused dyslipidemia and insulin resistance. Echocardiography revealed subtle cardiac remodeling in WD-fed mice, highlighting a significant age-diet interaction for some left ventricular morphofunctional parameters. Qualitative and parametric HFUS analyses of the liver in WD-fed mice showed a progressive increase in echogenicity and echotexture heterogeneity, and equal or higher brightness of the renal cortex. Furthermore, renal circulation was impaired in WD-fed female mice. The ultrasound and histopathological findings were concordant. Overall, HFUS can improve the translational value of preclinical DIO models through an integrated approach with conventional methods, enabling a comprehensive identification of early stages of diseases in vivo and non-invasively, according to the 3Rs.
Collapse
Affiliation(s)
- Sara Gargiulo
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| | - Virginia Barone
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Denise Bonente
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | | | - Giovanni Inzalaco
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| | - Lisa Gherardini
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| | - Eugenio Bertelli
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Mario Chiariello
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy
- Core Research Laboratory (CRL), Istituto per lo Studio la Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| |
Collapse
|
24
|
Conner SJ, Borges HB, Guarin JR, Gerton TJ, Yui A, Salhany KJ, Mensah DN, Hamilton GA, Le GH, Lew KC, Zhang C, Oudin MJ. Obesity Induces Temporally Regulated Alterations in the Extracellular Matrix That Drive Breast Tumor Invasion and Metastasis. Cancer Res 2024; 84:2761-2775. [PMID: 38900938 PMCID: PMC11908800 DOI: 10.1158/0008-5472.can-23-2526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 04/16/2024] [Accepted: 06/14/2024] [Indexed: 06/22/2024]
Abstract
Obesity is associated with increased incidence and metastasis of triple-negative breast cancer, an aggressive breast cancer subtype. The extracellular matrix (ECM) is a major component of the tumor microenvironment that drives metastasis. To characterize the temporal effects of age and high-fat diet (HFD)-driven weight gain on the ECM, we injected allograft tumor cells at 4-week intervals into mammary fat pads of mice fed a control or HFD, assessing tumor growth and metastasis and evaluating the ECM composition of the mammary fat pads, lungs, and livers. Tumor growth was increased in obese mice after 12 weeks on HFD. Liver metastasis increased in obese mice only at 4 weeks, and elevated body weight correlated with increased metastasis to the lungs but not the liver. Whole decellularized ECM coupled with proteomics indicated that early stages of obesity were sufficient to induce changes in the ECM composition. Obesity led to an increased abundance of the proinvasive ECM proteins collagen IV and collagen VI in the mammary glands and enhanced the invasive capacity of cancer cells. Cells of stromal vascular fraction and adipose stem and progenitor cells were primarily responsible for secreting collagen IV and collagen VI, not adipocytes. Longer exposure to HFD increased the invasive potential of ECM isolated from the lungs and liver, with significant changes in ECM composition found in the liver with short-term HFD exposure. Together, these data suggest that changes in the breast, lungs, and liver ECM underlie some of the effects of obesity on triple-negative breast cancer incidence and metastasis. Significance: Organ-specific extracellular matrix changes in the primary tumor and metastatic microenvironment are mechanisms by which obesity contributes to breast cancer progression.
Collapse
Affiliation(s)
- Sydney J Conner
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Hannah B Borges
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Justinne R Guarin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Thomas J Gerton
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Anna Yui
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Kenneth J Salhany
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Diamond N Mensah
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Grace A Hamilton
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Giang H Le
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Katherine C Lew
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Crystal Zhang
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
25
|
Galindev U, Erdenebold U, Batnasan G, Ganzorig O, Batdorj B. Anti-obesity effects of potential probiotic Lactobacillus strains isolated from Mongolian fermented dairy products in high-fat diet-induced obese rodent model. Braz J Microbiol 2024; 55:2501-2509. [PMID: 38789906 PMCID: PMC11405555 DOI: 10.1007/s42770-024-01372-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
This study aims to investigate the anti-obesity properties of lactic acid bacteria (LAB) isolated from fermented dairy products such as "Airag" and "Khoormog" in Mongolia. These traditional dairy products are widely used in Mongolia and believe in having potential probiotic, anti-diabetes, anti-cancer, and anti-tuberculosis properties and are made from unheated two-humped camel milk and mare milk, respectively. We chose three LAB strains based on their probiotic characteristics, including tolerance of gastric and bile acids. Then we checked the anti-obesity activity of probiotic strains in vivo. An animal model was evaluated in twenty male C57BL/6J mice by inducing obesity with a high-fat diet (HFD), which was divided into five groups: regular diet group (Negative control), HFD group (Positive control), HFD with Lacticaseibacillus paracasei X-1 (X-1), Lacticaseibacillus paracasei X-17 (X-17), and Limosilactobacillus fermentum BM-325 (BM-325). For six weeks, 5 × 109 colony-forming units (CFU) of bacteria were given orally to the LAB-fed groups. Fasting blood glucose (FBG), lipid profiles, organ index, and organ morphology were all measured. The probiotic strains suppressed growth in adipose cell volume, stabilized FBG, reduced liver cell degeneration, and slowed HFD-induced body weight gain. The results suggest that some strains increase general metabolism while lowering body weight.
Collapse
Affiliation(s)
- Unurjargal Galindev
- School of Arts and Sciences, National University of Mongolia, Ulaanbaatar, Mongolia
| | | | - Galindev Batnasan
- Institute of Biomedical Sciences, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Oyundelger Ganzorig
- School of Arts and Sciences, National University of Mongolia, Ulaanbaatar, Mongolia
| | - Batjargal Batdorj
- School of Arts and Sciences, National University of Mongolia, Ulaanbaatar, Mongolia.
| |
Collapse
|
26
|
Cait J, Winder CB, Mason GJ. How much 'enrichment' is enough for laboratory rodents? A systematic review and meta-analysis re-assessing the impact of well-resourced cages on morbidity and mortality. Appl Anim Behav Sci 2024; 278:106361. [DOI: 10.1016/j.applanim.2024.106361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
27
|
Nathan K, E M, Jabaris S SL, Visweswaraiah NK. Effect of abrupt and gradual calorie restriction regimens on biochemical and behavioral markers in obese mice model. Nutr Health 2024; 30:499-511. [PMID: 36650659 DOI: 10.1177/02601060221131726] [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: 01/19/2023]
Abstract
Background: Although Calorie Restriction (CR) is primarily considered in obesity management, behavioral studies of CR and its methodology of implementation are not well-defined. Objective: The study aimed to determine the efficacy of the extensively researched method of CR-abrupt calorie restriction (ACR) and a newly proposed gradual calorie restriction (GCR) in terms of body weight, behavioral and biochemical parameters in the obese animal model-C57BL/6J mice. Design: Male obese mice were subjected to GCR regimen for 14 weeks which was compared and evaluated for anxiety-like behavior and stress levels with ACR. Plasma corticosterone was measured before initiation of CR and every three weeks following GCR and ACR, whereas plasma insulin was measured twice-after obesity induction and post 14 weeks of CR. The behavioral assessments were conducted before inducing CR and every three weeks following ACR and GCR. Results: A significant anxiety-like behavior and an increase in plasma corticosterone were observed in mice on GCR during the critical initial six weeks of CR (p < 0.05). Both groups showed a decrease in plasma glucose levels; however, the GCR group showed a significant reduction (p < 0.01). There was a significant decrease in social interaction in both groups with an increase in the latency period and a decrease in time spent with the stranger animal during the social interaction test (p < 0.05). Conclusion: The mice on GCR regimen demonstrated lesser anxiety-like behavior and low plasma corticosterone levels compared to those on ACR. This gives us a new avenue in CR research to evaluate the methodologies of implementing CR.
Collapse
Affiliation(s)
- Kousalya Nathan
- Department of Biochemistry, Chettinad Academy of Research and Education, Kanchipuram District, Tamil Nadu, India
| | - Malligai E
- Department of Biochemistry, Chettinad Academy of Research and Education, Kanchipuram District, Tamil Nadu, India
| | - Sugin Lal Jabaris S
- Department of Pharmacology, Siddha Central Research Institute, Central Council for Research in Siddha, Anna Govt. Hospital Campus, Chennai, India
| | - Naveen K Visweswaraiah
- Australian Research Council for Complementary and Integrative Medicine (ARCCIM), Faculty of Health, University of Technology, Sydney, Australia
| |
Collapse
|
28
|
Abdulwahid-Kurdi SJ. The Impact of Clinacanthus nutans (Burm. F.) Leaf Extract on Sperm Quality and Antioxidant Activity in Male Mice Induced with Streptozotocin. JBRA Assist Reprod 2024; 28:382-391. [PMID: 38712836 PMCID: PMC11349258 DOI: 10.5935/1518-0557.20240010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 01/27/2024] [Indexed: 05/08/2024] Open
Abstract
OBJECTIVE Clinacanthus nutans (C. nutans) is a medicinal herb that most people with diabetes have historically taken. It's a diet high in antioxidants, which are supposed to help people live longer and be healthier. It is the first study to suggest using C. nutans to enhance the quality of sperm in male mice given a streptozotocin (STZ) injection. METHODS Sixty mice were divided into two groups at the age of four weeks: group one was fed a regular diet (n=10), while group two was administered a high-fat diet (n=50) for eight weeks to develop obesity. Obese mice were given 100mg/kg of STZ to produce hyperglycemia with a 20% mortality rate. Then, 40 hyperglycemic mice were separated into two groups: STZ (n=10) and sample (n=30). The treatment groups were administered a methanolic extract of C. nutans leaves by gavage at doses of 150, 300, and 500mg/kg of body weight (n=10) for 4 weeks. RESULTS In contrast to the STZ group, there was a substantial (p=0.001) drop in serum blood glucose and total sperm abnormalities in mice at varying doses. Catalase, glutathione s-transferase (GST), and total antioxidant capacity significantly (p=0.001) increased in the STZ mice group at varying doses, but malondialdehyde was reduced. In comparison to STZ mice, testosterone and luteinizing hormone (LH) levels improved in mice treated with extracts of C. nutans at various doses. For all of the following dependent variables, extraction of the leaf at higher concentrations of 500 milligrams/kilogram has better efficacy than 300 and 150 mg/kg after 4 weeks of treatment. CONCLUSIONS The research and development of new natural agents to combat oxidative stress-related diseases have sparked a lot of interest. As a result, the potential leaf extract of C. nutans contains anti-hyperglycemic compounds and improves the quality of sperm in male mice.
Collapse
|
29
|
Ribeiro FM, Petriz B, Anderson M, Assis V, dos Santos Rosa T, de Luca Correa H, Cavichiolli de Oliveira N, Passos L, Fonseca A, Brito LA, Silva O, Castro A, Franco OL. Discontinuation of HIIT restores diabesity while retraining increases gut microbiota diversity. iScience 2024; 27:110365. [PMID: 39175767 PMCID: PMC11338995 DOI: 10.1016/j.isci.2024.110365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/20/2024] [Accepted: 06/21/2024] [Indexed: 08/24/2024] Open
Abstract
Investigations involving high-intensity interval training (HIIT) have proven to be efficient in controlling diabesity. This study aimed to assess the impact of discontinuing HIIT and retraining within the context of diabesity. 75 C57BL6 mice went through 5 stages: baseline, induction of diabesity with Western diet, training, detraining, and retraining (6 weeks each period). Detraining led to elevated adiposity, exacerbated metabolic parameters and intestinal health, and altered gut microbiota composition. Retraining restored blood glucose regulation and enhanced intestinal health yet did not induce fat reduction. While both training and retraining exerted an effect on the composition of the gut microbiota, the impact of diet demonstrates a more substantial potency compared to that of exercise concerning intestinal health and microbiome. These findings may contribute to a broader understanding of diabesity management and introduce perspectives for the use of specific physical training to enhance patient outcomes and intestine health.
Collapse
Affiliation(s)
- Filipe Moura Ribeiro
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Postgraduate Program in Physical Education, Catholic University of Brasília, Brasília, DF, Brazil
- Laboratory of Molecular Exercise Physiology, University Center, Brasília, DF, Brazil
| | - Bernardo Petriz
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Laboratory of Molecular Exercise Physiology, University Center, Brasília, DF, Brazil
- Postgraduate Program in Biotechnology, S-Inova Biotech, Dom Bosco Catholic University, Campo Grande, MS, Brazil
| | - Maycon Anderson
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Laboratory of Molecular Exercise Physiology, University Center, Brasília, DF, Brazil
| | - Victoria Assis
- Laboratory of Molecular Exercise Physiology, University Center, Brasília, DF, Brazil
- Laboratory of Molecular Analysis, Postgraduate Program of Sciences and Technology of Health, University of Brasilia, Brasília, DF, Brazil
| | - Thiago dos Santos Rosa
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Postgraduate Program in Physical Education, Catholic University of Brasília, Brasília, DF, Brazil
| | - Hugo de Luca Correa
- Postgraduate Program in Physical Education, Catholic University of Brasília, Brasília, DF, Brazil
| | | | - Lana Passos
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Laboratory of Molecular Exercise Physiology, University Center, Brasília, DF, Brazil
| | - Amanda Fonseca
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Laboratory of Molecular Exercise Physiology, University Center, Brasília, DF, Brazil
| | - Luiz Arnaldo Brito
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Laboratory of Molecular Exercise Physiology, University Center, Brasília, DF, Brazil
| | - Osmar Silva
- Postgraduate Program in Pharmaceutical Sciences, Universidade Evangélica de Goiás, UniEVANGÉLICA, Goiás, GO, Brazil
| | - Alinne Castro
- Postgraduate Program in Biotechnology, S-Inova Biotech, Dom Bosco Catholic University, Campo Grande, MS, Brazil
| | - Octavio Luiz Franco
- Center for Proteomics and Biochemical Analysis, Postgraduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasília, Brasília, DF, Brazil
- Postgraduate Program in Physical Education, Catholic University of Brasília, Brasília, DF, Brazil
- Postgraduate Program in Biotechnology, S-Inova Biotech, Dom Bosco Catholic University, Campo Grande, MS, Brazil
| |
Collapse
|
30
|
Ponce-Balbuena D, Tyrrell DJ, Cruz-Cortés C, Guerrero-Serna G, Da Rocha AM, Herron TJ, Song J, Raza DS, Anumonwo J, Goldstein DR, Espinoza-Fonseca LM. Paradoxical SERCA dysregulation contributes to atrial fibrillation in a model of diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606385. [PMID: 39149279 PMCID: PMC11326153 DOI: 10.1101/2024.08.02.606385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Obesity is a major risk factor for atrial fibrillation (AF) the most common serious cardiac arrhythmia, but the molecular mechanisms underlying diet-induced AF remain unclear. In this study, we subjected mice to a chronic high-fat diet and acute sympathetic activation ('two-hit' model) to study the mechanisms by which diet-induced obesity promotes AF. Surface electrocardiography revealed that diet-induced obesity and sympathetic activation synergize during intracardiac tachypacing to induce AF. At the cellular level, diet-induced obesity and acute adrenergic stimulation facilitate the formation of delayed afterdepolarizations in atrial myocytes, implicating altered Ca2+ dynamics as the underlying cause of AF. We found that diet-induced obesity does not alter the expression of major Ca2+-handling proteins in atria, including the sarcoplasmic reticulum Ca2+-ATPase (SERCA), a major component of beat-to-beat Ca2+ cycling in the heart. Paradoxically, obesity reduces phospholamban phosphorylation, suggesting decreased SERCA activity, yet atrial myocytes from obese mice showed a significantly increased Ca2+ transient amplitude and SERCA-mediated Ca2+ uptake. Adrenergic stimulation further increases the Ca2+ transient amplitude but does not affect Ca2+ reuptake in atrial myocytes from obese mice. Transcriptomics analysis showed that a high-fat diet prompts upregulation of neuronatin, a protein that has been implicated in obesity and is known to stimulate SERCA activity. We propose a mechanism in which obesity primes SERCA for paradoxical activation, and adrenergic stimulation facilitates AF conversion through a Ca2+-induced Ca2+ release gain in atrial myocytes. Overall, this study links obesity, altered Ca2+ signaling, and AF, and targeting this mechanism may prove effective for treating obesity-induced AF.
Collapse
Affiliation(s)
- Daniela Ponce-Balbuena
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Daniel J. Tyrrell
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carlos Cruz-Cortés
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Guadalupe Guerrero-Serna
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Andre Monteiro Da Rocha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Todd J. Herron
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Jianrui Song
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Danyal S. Raza
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Justus Anumonwo
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| | - Daniel R. Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Microbiology and Immunology, University of Michigan, MI 48109, USA
| | - L. Michel Espinoza-Fonseca
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Center for Arrhythmia Research, University of Michigan, MI 48109, USA
| |
Collapse
|
31
|
Bah TM, Davis CM, Allen EM, Borkar RN, Perez R, Grafe MR, Raber J, Pike MM, Alkayed NJ. Soluble epoxide hydrolase inhibition reverses cognitive dysfunction in a mouse model of metabolic syndrome by modulating inflammation. Prostaglandins Other Lipid Mediat 2024; 173:106850. [PMID: 38735559 PMCID: PMC11218661 DOI: 10.1016/j.prostaglandins.2024.106850] [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: 03/01/2024] [Revised: 04/26/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Midlife metabolic syndrome (MetS) is associated with cognitive impairment in late life. The mechanism of delayed MetS-related cognitive dysfunction (MetSCD) is not clear, but it has been linked to systemic inflammation and chronic cerebral microangiopathy. Currently there is no treatment for late life MetSCD other than early risk factor modification. We investigated the effect of soluble epoxide hydrolase (sEH) inhibitor 4-[[trans-4-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]cyclohexyl]oxy]-benzoic acid (t-AUCB) on cognitive performance, cerebral blood flow (CBF), and central and peripheral inflammation in the high-fat diet (HFD) model of MetS in mice. At 6 weeks of age, male mice were randomly assigned to receive either HFD or standard chow (STD) for 6 months. Mice received either t-AUCB or vehicle for 4 weeks. Cognitive performance was evaluated, followed by CBF measurement using magnetic resonance imaging (MRI). At the end of the study, blood was collected for measurement of eicosanoids and inflammatory cytokines. The brains were then analyzed by immunohistochemistry for glial activation markers. The HFD caused a significant impairment in novel object recognition. Treatment with t-AUCB increased plasma levels of 14,15-EET, prevented this cognitive impairment and modified hippocampal glial activation and plasma cytokine levels, without affecting CBF in mice on HFD. In conclusion, sEH inhibition for four weeks prevents cognitive deficits in mice on chronic HFD by modulating inflammatory processes without affecting CBF.
Collapse
Affiliation(s)
- Thierno M Bah
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Catherine M Davis
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Elyse M Allen
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Rohan N Borkar
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ruby Perez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Marjorie R Grafe
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA
| | - Martin M Pike
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Nabil J Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
32
|
Chen YL, Wu JM, Chen KY, Wu MH, Yang PJ, Lee PC, Chen PD, Kuo TC, Yeh SL, Lin MT. Intravenous calcitriol administration improves the liver redox status and attenuates ferroptosis in mice with high-fat diet-induced obesity complicated with sepsis. Biomed Pharmacother 2024; 177:116926. [PMID: 38906016 DOI: 10.1016/j.biopha.2024.116926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024] Open
Abstract
Obesity aggravates ferroptosis, and vitamin D (VD) may inhibit ferroptosis. We hypothesized that weight reduction and/or calcitriol administration have benefits against the sepsis-induced liver redox imbalance and ferroptosis in obese mice. Mice were fed a high-fat diet for 11 weeks, then half of the mice continued to consume the diet, while the other half were transferred to a low-energy diet for 5 weeks. After feeding the respective diets for 16 weeks, sepsis was induced by cecal ligation and puncture (CLP). Septic mice were divided into four experimental groups: OS group, obese mice injected with saline; OD group, obese mice with calcitriol; WS group, weight-reduction mice with saline; and WD group, weight-reduction mice with calcitriol. Mice in the respective groups were euthanized at 12 or 24 h after CLP. Results showed that the OS group had the highest inflammatory mediators and lipid peroxide levels in the liver. Calcitriol treatment reduced iron content, enhanced the reduced glutathione/oxidized glutathione ratio, upregulated nuclear factor erythroid 2-related factor 2, ferroptosis-suppressing protein 1, and solute carrier family 7 member 11 expression levels. Also, mitochondrion-associated nicotinamide adenine dinucleotide phosphate oxidase 1, peroxisome proliferator-activated receptor-γ coactivator 1, hypoxia-inducible factor-1α, and heme oxidase-1 expression levels increased in the late phase of sepsis. These results were not noted in the WS group. These findings suggest that calcitriol treatment elicits a more-balanced glutathione redox status, alleviates liver ferroptosis, and enhances mitochondrial biogenesis-associated gene expressions. Weight reduction alone had minimal influences on liver ferroptosis and mitochondrial biogenesis in obese mice with sepsis.
Collapse
Affiliation(s)
- Ya-Ling Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Jin-Ming Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Kuen-Yuan Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Hsun Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Jen Yang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Po-Da Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ting-Chun Kuo
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Sung-Ling Yeh
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ming-Tsan Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| |
Collapse
|
33
|
Dole NS, Betancourt-Torres A, Kaya S, Obata Y, Schurman CA, Yoon J, Yee CS, Khanal V, Luna CA, Carroll M, Salinas JJ, Miclau E, Acevedo C, Alliston T. High-fat and high-carbohydrate diets increase bone fragility through TGF-β-dependent control of osteocyte function. JCI Insight 2024; 9:e175103. [PMID: 39171528 PMCID: PMC11343608 DOI: 10.1172/jci.insight.175103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Obesity can increase the risk of bone fragility, even when bone mass is intact. This fragility stems from poor bone quality, potentially caused by deficiencies in bone matrix material properties. However, cellular and molecular mechanisms leading to obesity-related bone fragility are not fully understood. Using male mouse models of obesity, we discovered TGF-β signaling plays a critical role in mediating the effects of obesity on bone. High-carbohydrate and high-fat diets increase TGF-β signaling in osteocytes, which impairs their mitochondrial function, increases cellular senescence, and compromises perilacunar/canalicular remodeling and bone quality. By specifically inhibiting TGF-β signaling in mouse osteocytes, some of the negative effects of high-fat and high-carbohydrate diets on bones, including the lacunocanalicular network, perilacunar/canalicular remodeling, senescence, and mechanical properties such as yield stress, were mitigated. DMP1-Cre-mediated deletion of TGF-β receptor II also blunted adverse effects of high-fat and high-carbohydrate diets on energy balance and metabolism. These findings suggest osteocytes are key in controlling bone quality in response to high-fat and high-carbohydrate diets. Calibrating osteocyte function could mitigate bone fragility associated with metabolic diseases while reestablishing energy balance.
Collapse
Affiliation(s)
- Neha S. Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock Arkansas, USA
| | - Andrés Betancourt-Torres
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Yoshihiro Obata
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Charles A. Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| | - Jihee Yoon
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Cristal S. Yee
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Vivek Khanal
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock Arkansas, USA
| | - Clarissa Aguirre Luna
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Madeline Carroll
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock Arkansas, USA
| | - Jennifer J. Salinas
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Elizabeth Miclau
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Claire Acevedo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, California, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| |
Collapse
|
34
|
Marain NF, Jonckheere AC, Dilissen E, Cremer J, Roskams T, Colemont M, Bullens DM, Dupont LJ, Vanoirbeek JA. Combined Exercise and Diet Induce Airway Hyperreactivity While Reducing Liver Steatosis in Mice with Diet-Induced Obesity. Nutrients 2024; 16:2129. [PMID: 38999877 PMCID: PMC11243263 DOI: 10.3390/nu16132129] [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: 06/03/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Obesity is a multi-organ system disease, which is associated with, e.g., a higher prevalence of non-alcoholic fatty liver disease (NAFLD) and asthma. Little is known regarding the effect of obesity-related parameters (including liver integrity) and the respiratory phenotype after a combination of physical activity and diet. METHODS Thirty-two C57BL/6 mice were, after 27 weeks of a high fat diet (HFD), randomly assigned to two dietary interventions for three weeks: a HFD or a normal chow diet (NCD). In both dietary groups, half of the animals were subjected to a sub-maximal exercise protocol. Lung function, lung inflammation, liver histology, and metabolic profile were determined. RESULTS Mice with obesity did not show airway hyperreactivity after methacholine provocation. Sub-maximal exercise with diet (NCD/E) induced a significant reduction in forced expiratory volume in 0.1 s after methacholine provocation. NCD/E had significantly more neutrophils and inflammation (IFN-γ, TNF-α, IL-4, and IL-17F) in bronchoalveolar lavage compared to non-exercising mice on a HFD (HFD/NE). However, more epithelial injury (serum surfactant protein D and IL-33) was seen in HFD/NE. Additionally, hepatic steatosis and fibrosis were reduced by combined diet and sub-maximal exercise. CONCLUSIONS Combining sub-maximal exercise with diet induced airway hyperreactivity and pulmonary inflammation, while body weight, hepatic steatosis, and fibrosis improved.
Collapse
Affiliation(s)
- Nora F Marain
- KU Leuven, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery, 3000 Leuven, Belgium
| | - Anne-Charlotte Jonckheere
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, 3000 Leuven, Belgium
| | - Ellen Dilissen
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, 3000 Leuven, Belgium
| | - Jonathan Cremer
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, 3000 Leuven, Belgium
| | - Tania Roskams
- KU Leuven, Department of Imaging & Pathology, Translational Cell & Tissue Research, 3000 Leuven, Belgium
| | - Marieke Colemont
- KU Leuven, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery, 3000 Leuven, Belgium
| | - Dominique M Bullens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, 3000 Leuven, Belgium
- Clinical Division of Paediatrics, UZ Leuven, 3000 Leuven, Belgium
| | - Lieven J Dupont
- KU Leuven, Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery, 3000 Leuven, Belgium
- Clinical Division of Respiratory Medicine, UZ Leuven, 3000 Leuven, Belgium
| | - Jeroen A Vanoirbeek
- KU Leuven, Department of Public Health and Primary Care, 3000 Leuven, Belgium
| |
Collapse
|
35
|
Kwon J, Sa M, Kim H, Seong Y, Lee CJ. Egocentric 3D Skeleton Learning in a Deep Neural Network Encodes Obese-like Motion Representations. Exp Neurobiol 2024; 33:119-128. [PMID: 38993079 PMCID: PMC11247279 DOI: 10.5607/en24008] [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: 04/16/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024] Open
Abstract
Obesity is a growing health concern, mainly caused by poor dietary habits. Yet, accurately tracking the diet and food intake of individuals with obesity is challenging. Although 3D motion capture technology is becoming increasingly important in healthcare, its potential for detecting early signs of obesity has not been fully explored. In this research, we used a deep LSTM network trained with individual identity (identity-trained deep LSTM network) to analyze 3D time-series skeleton data from mouse models with diet-induced obesity. First, we analyzed the data from two different viewpoints: allocentric and egocentric. Second, we trained various deep recurrent networks (e.g., RNN, GRU, LSTM) to predict the identity. Lastly, we tested whether these models effectively encode obese-like motion representations by training a support vector classifier with the latent features from the last layer. Our experimental results indicate that the optimal performance is achieved when utilizing an identity-trained deep LSTM network in conjunction with an egocentric viewpoint. This approach suggests a new way to use deep learning to spot health risks in mouse models of obesity and should be useful for detecting early signs of obesity in humans.
Collapse
Affiliation(s)
- Jea Kwon
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - Hyewon Kim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Department of Pre-Medicine, Eulji University School of Medicine, Daejeon 34824, Korea
| | - Yejin Seong
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - C. Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34126, Korea
| |
Collapse
|
36
|
Ding M, Nielsen K. Inbred Mouse Models in Cryptococcus neoformans Research. J Fungi (Basel) 2024; 10:426. [PMID: 38921412 PMCID: PMC11204852 DOI: 10.3390/jof10060426] [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: 04/25/2024] [Revised: 06/01/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Animal models are frequently used as surrogates to understand human disease. In the fungal pathogen Cryptococcus species complex, several variations of a mouse model of disease were developed that recapitulate different aspects of human disease. These mouse models have been implemented using various inbred and outbred mouse backgrounds, many of which have genetic differences that can influence host response and disease outcome. In this review, we will discuss the most commonly used inbred mouse backgrounds in C. neoformans infection models.
Collapse
Affiliation(s)
| | - Kirsten Nielsen
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
37
|
Zabolotneva AA, Kolesnikova IM, Vasiliev IY, Grigoryeva TV, Roumiantsev SA, Shestopalov AV. The Obesogenic Gut Microbiota as a Crucial Factor Defining the Depletion of Predicted Enzyme Abundance for Vitamin B12 Synthesis in the Mouse Intestine. Biomedicines 2024; 12:1280. [PMID: 38927487 PMCID: PMC11201498 DOI: 10.3390/biomedicines12061280] [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: 03/29/2024] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Currently, obesity is a critical global public health burden. Numerous studies have demonstrated the regulation of the pathogenesis of obesity and metabolic abnormalities by the gut microbiota and microbial factors; however, their involvement in the various degrees of obesity is not yet well understood. Previously, obesity has been shown to be associated with decreased levels of vitamin B12. Considering exclusive microbial production of vitamin B12, we hypothesized that a decrease in cobalamin levels in obese individuals may be at least partially caused by its depleted production in the intestinal tract by the commensal microbiota. In the present study, our aim was to estimate the abundance of enzymes and metabolic pathways for vitamin B12 synthesis in the gut microbiota of mouse models of alimentary and genetically determined obesity, to evaluate the contribution of the obesogenic microbiome to vitamin B12 synthesis in the gut. We have defined a significantly lower predicted abundance of enzymes and metabolic pathways for vitamin B12 biosynthesis in obese mice compared to non-obese mice, wherein enzyme depletion was more pronounced in lepr(-/-) (db/db) mice, which developed severe obesity. The predicted abundance of enzymes involved in cobalamin synthesis is strongly correlated with the representation of several microbes in high-fat diet-fed mice, while there were almost no correlations in db/db mice. Therefore, the degree of obesity and the composition of the correspondent microbiota are the main contributors to the representation of genes and pathways for cobalamin biosynthesis in the mouse gut.
Collapse
Affiliation(s)
- Anastasia A. Zabolotneva
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, N. I. Pirogov Russian National Research Medical University, 1 Ostrovitianov Str., Moscow 117997, Russia; (I.M.K.); (S.A.R.); (A.V.S.)
- Laboratory of Biochemistry of Signaling Pathways, Endocrinology Research Center, 11 Dm. Ulyanova Str., Moscow 117036, Russia; (I.Y.V.); (T.V.G.)
| | - Irina M. Kolesnikova
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, N. I. Pirogov Russian National Research Medical University, 1 Ostrovitianov Str., Moscow 117997, Russia; (I.M.K.); (S.A.R.); (A.V.S.)
- Laboratory of Biochemistry of Signaling Pathways, Endocrinology Research Center, 11 Dm. Ulyanova Str., Moscow 117036, Russia; (I.Y.V.); (T.V.G.)
| | - Ilya Yu. Vasiliev
- Laboratory of Biochemistry of Signaling Pathways, Endocrinology Research Center, 11 Dm. Ulyanova Str., Moscow 117036, Russia; (I.Y.V.); (T.V.G.)
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Str., Kazan 420008, Russia
| | - Tatiana V. Grigoryeva
- Laboratory of Biochemistry of Signaling Pathways, Endocrinology Research Center, 11 Dm. Ulyanova Str., Moscow 117036, Russia; (I.Y.V.); (T.V.G.)
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Str., Kazan 420008, Russia
| | - Sergei A. Roumiantsev
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, N. I. Pirogov Russian National Research Medical University, 1 Ostrovitianov Str., Moscow 117997, Russia; (I.M.K.); (S.A.R.); (A.V.S.)
- Laboratory of Biochemistry of Signaling Pathways, Endocrinology Research Center, 11 Dm. Ulyanova Str., Moscow 117036, Russia; (I.Y.V.); (T.V.G.)
| | - Aleksandr V. Shestopalov
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, N. I. Pirogov Russian National Research Medical University, 1 Ostrovitianov Str., Moscow 117997, Russia; (I.M.K.); (S.A.R.); (A.V.S.)
- Laboratory of Biochemistry of Signaling Pathways, Endocrinology Research Center, 11 Dm. Ulyanova Str., Moscow 117036, Russia; (I.Y.V.); (T.V.G.)
| |
Collapse
|
38
|
Conlon JM, Owolabi BO, Flatt PR, Abdel-Wahab YHA. Amphibian host-defense peptides with potential for Type 2 diabetes therapy - an updated review. Peptides 2024; 175:171180. [PMID: 38401671 DOI: 10.1016/j.peptides.2024.171180] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Investigations conducted since 2018 have identified several host-defense peptides present in frog skin secretions whose properties suggest the possibility of their development into a new class of agent for Type 2 diabetes (T2D) therapy. Studies in vitro have described peptides that (a) stimulate insulin release from BRIN-BD11 clonal β-cells and isolated mouse islets, (b) display β-cell proliferative activity and protect against cytokine-mediated apoptosis and (c) stimulate production of the anti-inflammatory cytokine IL-10 and inhibit production of the pro-inflammatory cytokines TNF-α and IL-1β. Rhinophrynin-27, phylloseptin-3.2TR and temporin F are peptides with therapeutic potential. Studies in vivo carried out in db/db and high fat-fed mice have shown that twice-daily administration of [S4K]CPF-AM1 and [A14K]PGLa-AM1, analogs of peptides first isolated from the octoploid frog Xenopus amieti, over 28 days lowers circulating glucose and HbA1c concentrations, increases insulin sensitivity and improves glucose tolerance and lipid profile. Peptide treatment produced potentially beneficial changes in the expression of skeletal muscle genes involved in insulin signaling and islet genes involved in insulin secretion in these murine models of T2D. Lead compounds uncovered by the study of frog HDPs may provide a basis for the design of new types of agents that can be used, alone or in combination with existing therapies, for the treatment of T2D.
Collapse
Affiliation(s)
- J Michael Conlon
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK.
| | - Bosede O Owolabi
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| | - Yasser H A Abdel-Wahab
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland BT52 1SA, UK
| |
Collapse
|
39
|
Siouti E, Salagianni M, Manioudaki M, Pavlos E, Klinakis A, Galani IE, Andreakos E. Notch signaling in adipose tissue macrophages prevents diet-induced inflammation and metabolic dysregulation. Eur J Immunol 2024; 54:e2350669. [PMID: 38339772 DOI: 10.1002/eji.202350669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
The importance of macrophages in adipose tissue (AT) homeostasis and inflammation is well established. However, the potential cues that regulate their function remain incompletely understood. To bridge this important gap, we sought to characterize novel pathways involved using a mouse model of diet-induced obesity. By performing transcriptomics analysis of AT macrophages (ATMs), we found that late-stage ATMs from high-fat diet mice presented with perturbed Notch signaling accompanied by robust proinflammatory and metabolic changes. To explore the hypothesis that the deregulated Notch pathway contributes to the development of AT inflammation and diet-induced obesity, we employed a genetic approach to abrogate myeloid Notch1 and Notch2 receptors. Our results revealed that the combined loss of Notch1 and Notch2 worsened obesity-related metabolic dysregulation. Body and AT weight gain was higher, blood glucose levels increased and metabolic parameters were substantially worsened in deficient mice fed high-fat diet. Moreover, serum insulin and leptin were elevated as were triglycerides. Molecular analysis of ATMs showed that deletion of Notch receptors escalated inflammation through the induction of an M1-like pro-inflammatory phenotype. Our findings thus support a protective role of myeloid Notch signaling in adipose tissue inflammation and metabolic dysregulation.
Collapse
Affiliation(s)
- Eleni Siouti
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria Salagianni
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria Manioudaki
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Eleftherios Pavlos
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Apostolos Klinakis
- Center for Basic Research, Biomedical Research Foundation Academy of Athens, Athens, 11527, Greece
| | - Ioanna-Evdokia Galani
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
40
|
Bankole T, Ma T, Arora I, Lei Z, Raju M, Li Z, Li Y. The Effect of Broccoli Glucoraphanin Supplementation on Ameliorating High-Fat-Diet-Induced Obesity through the Gut Microbiome and Metabolome Interface. Mol Nutr Food Res 2024; 68:e2300856. [PMID: 38676466 DOI: 10.1002/mnfr.202300856] [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: 12/19/2023] [Revised: 03/22/2024] [Indexed: 04/29/2024]
Abstract
SCOPE Obesity and its metabolic comorbidities pose a major global challenge for public health. Glucoraphanin (GRN) is a natural bioactive compound enriched in broccoli that is known to have potential health benefits against various human chronic diseases. METHODS AND RESULTS This study investigats the effects of broccoli GRN supplementation on body weight, metabolic parameters, gut microbiome and metabolome associated with obesity. The study is conducted on an obese-related C57BL/6J mouse model through the treatment of normal control diet, high-fat diet (HFD)and GRN-supplemented HFD (HFD-GRN) to determine the metabolic protection of GRN. The results shows that GRN treatment alleviates obesity-related traits leading to improved glucose metabolism in HFD-fed animals. Mechanically, the study noticed that GRN significantly shifts the gut microbial diversity and composition to an eubiosis status. GRN supplement also significantly alters plasma metabolite profiles. Further integrated analysis reveal a complex interaction between the gut microbes and host metabolism that may contribute to GRN-induced beneficial effects against HFD. CONCLUSION These results indicate that beneficial effects of broccoli GRN on reversing HFD-induced adverse metabolic parameters may be attributed to its impacts on reprogramming microbial community and metabolites. Identification of the mechanistic functions of GRN further warrants it as a dietary candidate for obesity prevention.
Collapse
Affiliation(s)
- Taiwo Bankole
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, University of Maryland, College Park, MD, 20742, USA
| | - Itika Arora
- Department of Microbiology and Immunology, University of Miami, Miami, FL, 33136, USA
| | - Zhentian Lei
- Metabolomics Center, University of Missouri at Columbia, Columbia, MO, 65211, USA
| | - Murugesan Raju
- Bioinformatics and Analytics Core, University of Missouri at Columbia, Columbia, MO, 65211, USA
| | - Zhenhai Li
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Yuanyuan Li
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| |
Collapse
|
41
|
Fofana M, Li Z, Li H, Li W, Wu L, Lu L, Liu Q. Decreased Ubiquitination and Acetylation of Histones 3 and 4 Are Associated with Obesity-Induced Disorders of Spermatogenesis in Mice. TOXICS 2024; 12:296. [PMID: 38668519 PMCID: PMC11055147 DOI: 10.3390/toxics12040296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Obesity, a chronic metabolic disorder, is related to cardiovascular diseases, diabetes, cancer, and reproductive disorders. The relationship between obesity and male infertility is now well recognized, but the mechanisms involved are unclear. We aimed to observe the effect of obesity on spermatogenesis and to investigate the role of histone ubiquitination and acetylation modifications in obesity-induced spermatogenesis disorders. METHODS Thirty male C57BL/6J mice were randomly divided into two groups. The control group was fed with a general maintenance diet (12% fat), while a high-fat diet (HFD) group was fed with 40% fat for 10 weeks; then, they were mated with normal females. The fertility of male mice was calculated, testicular and sperm morphology were observed, and the expression levels of key genes and the levels of histone acetylation and ubiquitination modification during spermatogenesis were detected. RESULTS The number of sperm was decreased, as well as the sperm motility, while the number of sperm with malformations was increased. In the testes, the mRNA and protein expression levels of gonadotropin-regulated testicular RNA helicase (GRTH/DDX25), chromosome region maintenance-1 protein (CRM1), high-mobility group B2 (HMGB2), phosphoglycerate kinase 2 (PGK2), and testicular angiotensin-converting enzyme (tACE) were decreased. Furthermore, obesity led to a decrease in ubiquitinated H2A (ubH2A) and reduced levels of histone H3 acetylation K18 (H3AcK18) and histone H4 acetylation K5, K8, K12, and K16 (H4tetraAck), which disrupted protamine 1 (Prm1) deposition in testis tissue. CONCLUSION These results suggest that low levels of histone ubiquitination and acetylation are linked with obesity-induced disorders during spermatogenesis, contributing to a better understanding of obesity-induced damage to male reproduction.
Collapse
Affiliation(s)
- Mahamadou Fofana
- Center for Global Health, China International Cooperation Center for Environment and Human Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (M.F.); (Z.L.); (H.L.); (W.L.)
| | - Zhenyang Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (M.F.); (Z.L.); (H.L.); (W.L.)
| | - Han Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (M.F.); (Z.L.); (H.L.); (W.L.)
| | - Wenqi Li
- Center for Global Health, China International Cooperation Center for Environment and Human Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (M.F.); (Z.L.); (H.L.); (W.L.)
| | - Lu Wu
- Suzhou Center for Disease Control and Prevention, Suzhou Institute for Advanced Study of Public Health, Suzhou School, Nanjing Medical University, Suzhou 215004, China;
| | - Lu Lu
- Animal Core Facility, The Key Laboratory of Model Animal, Jiangsu Animal Experimental Center for Medical and Pharmaceutical Research, Nanjing Medical University, Nanjing 211166, China
| | - Qizhan Liu
- Center for Global Health, China International Cooperation Center for Environment and Human Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (M.F.); (Z.L.); (H.L.); (W.L.)
- Suzhou Center for Disease Control and Prevention, Suzhou Institute for Advanced Study of Public Health, Suzhou School, Nanjing Medical University, Suzhou 215004, China;
| |
Collapse
|
42
|
Lobato S, Castillo-Granada AL, Bucio-Pacheco M, Salomón-Soto VM, Álvarez-Valenzuela R, Meza-Inostroza PM, Villegas-Vizcaíno R. PM 2.5, component cause of severe metabolically abnormal obesity: An in silico, observational and analytical study. Heliyon 2024; 10:e28936. [PMID: 38601536 PMCID: PMC11004224 DOI: 10.1016/j.heliyon.2024.e28936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Obesity is currently one of the most alarming pathological conditions due to the progressive increase in its prevalence. In the last decade, it has been associated with fine particulate matter suspended in the air (PM2.5). The purpose of this study was to explore the mechanistic interaction of PM2.5 with a high-fat diet (HFD) through the differential regulation of transcriptional signatures, aiming to identify the association of these particles with metabolically abnormal obesity. The research design was observational, using bioinformatic methods and an explanatory approach based on Rothman's causal model. We propose three new transcriptional signatures in murine adipose tissue. The sum of transcriptional differences between the group exposed to an HFD and PM2.5, compared to the control group, were 0.851, 0.265, and -0.047 (p > 0.05). The HFD group increased body mass by 20% with two positive biomarkers of metabolic impact. The group exposed to PM2.5 maintained a similar weight to the control group but exhibited three positive biomarkers. Enriched biological pathways (p < 0.05) included PPAR signaling, small molecule transport, adipogenesis genes, cytokine-cytokine receptor interaction, and HIF-1 signaling. Transcriptional regulation predictions revealed CpG islands and common transcription factors. We propose three new transcriptional signatures: FAT-PM2.5-CEJUS, FAT-PM2.5-UP, and FAT-PM2.5-DN, whose transcriptional regulation profile in adipocytes was statistically similar by dietary intake and HFD and exposure to PM2.5 in mice; suggesting a mechanistic interaction between both factors. However, HFD-exposed murines developed moderate metabolically abnormal obesity, and PM2.5-exposed murines developed severe abnormal metabolism without obesity. Therefore, in Rothman's terms, it is concluded that HFD is a sufficient cause of the development of obesity, and PM2.5 is a component cause of severe abnormal metabolism of obesity. These signatures would be integrated into a systemic biological process that would induce transcriptional regulation in trans, activating obesogenic biological pathways, restricting lipid mobilization pathways, decreasing adaptive thermogenesis and angiogenesis, and altering vascular tone thus inducing a severe metabolically abnormal obesity.
Collapse
Affiliation(s)
- Sagrario Lobato
- Departamento de Investigación en Salud, Servicios de Salud del Estado de Puebla, 15 South Street 302, Puebla, Mexico
- Promoción y Educación para la Salud, Universidad Abierta y a Distancia de México. Universidad Avenue 1200, 1st Floor, quadrant 1-2, Xoco, Benito Juarez, 03330, Mexico City, Mexico
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato, Mexico
| | - A. Lourdes Castillo-Granada
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato, Mexico
- Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Guelatao Avenue 66, Ejército de Oriente Indeco II ISSSTE, Iztapalapa, 09230, Mexico City, Mexico
| | - Marcos Bucio-Pacheco
- Educación Superior, Centro de Estudios, “Justo Sierra”, Surutato, Badiraguato, Mexico
- Facultad de Biología, Universidad Autónoma de Sinaloa, Americas Avenue, Universitarios Blvd., University City, 80040, Culiacán Rosales, Mexico
| | | | | | | | | |
Collapse
|
43
|
Xhonneux I, Marei WFA, Meulders B, Andries S, Leroy JLMR. The interplay of maternal and offspring obesogenic diets: the impact on offspring metabolism and muscle mitochondria in an outbred mouse model. Front Physiol 2024; 15:1354327. [PMID: 38585221 PMCID: PMC10995298 DOI: 10.3389/fphys.2024.1354327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/01/2024] [Indexed: 04/09/2024] Open
Abstract
Consumption of obesogenic (OB) diets increases the prevalence of maternal obesity worldwide, causing major psychological and social burdens in women. Obesity not only impacts the mother's health and fertility but also elevates the risk of obesity and metabolic disorders in the offspring. Family lifestyle is mostly persistent through generations, possibly contributing to the growing prevalence of obesity. We hypothesized that offspring metabolic health is dependent on both maternal and offspring diet and their interaction. We also hypothesized that the sensitivity of the offspring to the diet may be influenced by the match or mismatch between offspring and maternal diets. To test these hypotheses, outbred Swiss mice were fed a control (C, 10% fat, 7% sugar, and n = 14) or OB diet (60% fat, 20% sugar, and n = 15) for 7 weeks and then mated with the same control males. Mice were maintained on the same corresponding diet during pregnancy and lactation, and the offspring were kept with their mothers until weaning. The study focused only on female offspring, which were equally distributed at weaning and fed C or OB diets for 7 weeks, resulting in four treatment groups: C-born offspring fed C or OB diets (C » C and C » OB) and OB-born offspring fed C or OB diets (OB » C and OB » OB). Adult offspring's systemic blood profile (lipid and glucose metabolism) and muscle mitochondrial features were assessed. We confirmed that the offspring's OB diet majorly impacted the offspring's health by impairing the offspring's serum glucose and lipid profiles, which are associated with abnormal muscle mitochondrial ultrastructure. Contrarily, maternal OB diet was associated with increased expression of mitochondrial complex markers and mitochondrial morphology in offspring muscle, but no additive effects of (increased sensitivity to) an offspring OB diet were observed in pups born to obese mothers. In contrast, their metabolic profile appeared to be healthier compared to those born to lean mothers and fed an OB diet. These results are in line with the thrifty phenotype hypothesis, suggesting that OB-born offspring are better adapted to an environment with high energy availability later in life. Thus, using a murine outbred model, we could not confirm that maternal obesogenic diets contribute to female familial obesity in the following generations.
Collapse
Affiliation(s)
- Inne Xhonneux
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Waleed F. A. Marei
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Ben Meulders
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Silke Andries
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| | - Jo L. M. R. Leroy
- Department of Veterinary Sciences, Laboratory of Veterinary Physiology and Biochemistry, Gamete Research Centre, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
44
|
Md Shahrulnizam NAN, Mohd Efendy Goon MD, Ab Rahim S, Lew SW, Sheikh Abdul Kadir SH, Ibrahim E. Palm-based tocotrienol-rich fraction (TRF) supplementation modulates cardiac sod1 expression, fxr target gene expression, and tauro-conjugated bile acid levels in aleptinemic mice fed a high-fat diet. GENES & NUTRITION 2024; 19:3. [PMID: 38413846 PMCID: PMC10898183 DOI: 10.1186/s12263-024-00742-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/10/2024] [Indexed: 02/29/2024]
Abstract
Tocotrienol-rich fraction (TRF) has been reported to protect the heart from oxidative stress-induced inflammation. It is, however, unclear whether the protective effects of TRF against oxidative stress involve the activation of farnesoid X receptor (fxr), a bile acid receptor, and the regulation of bile acid metabolites. In the current study, we investigated the effects of TRF supplementation on antioxidant activities, expression of fxr and its target genes in cardiac tissue, and serum untargeted metabolomics of high-fat diet-fed mice. Mice were divided into high-fat diet (HFD) with or without TRF supplementation (control) for 6 weeks. At the end of the intervention, body weight (BW), waist circumference (WC), and random blood glucose were measured. Heart tissues were collected, and the gene expression of sod1, sod2, gpx, and fxr and its target genes shp and stat3 was determined. Serum was subjected to untargeted metabolomic analysis using UHPLC-Orbitrap. In comparison to the control, the WC of the TRF-treated group was higher (p >0.05) than that of the HFD-only group, in addition there was no significant difference in weight or random blood glucose level. Downregulation of sod1, sod2, and gpx expression was observed in TRF-treated mice; however, only sod1 was significant when compared to the HFD only group. The expression of cardiac shp (fxr target gene) was significantly upregulated, but stat3 was significantly downregulated in the TRF-treated group compared to the HFD-only group. Biochemical pathways found to be influenced by TRF supplementation include bile acid secretion, primary bile acid biosynthesis, and biotin and cholesterol metabolism. In conclusion, TRF supplementation in HFD-fed mice affects antioxidant activities, and more interestingly, TRF also acts as a signaling molecule that is possibly involved in several bile acid-related biochemical pathways accompanied by an increase in cardiac fxr shp expression. This study provides new insight into TRF in deregulating bile acid receptors and metabolites in high-fat diet-fed mice.
Collapse
Affiliation(s)
- Nur Aliah Natasha Md Shahrulnizam
- Institute of Medical Molecular Biotechnology (IMMB), Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia
| | - Mohd Danial Mohd Efendy Goon
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerFoRM), Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia
| | - Sharaniza Ab Rahim
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia
| | - Sook Weih Lew
- Department of Pediatrics, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia
| | - Siti Hamimah Sheikh Abdul Kadir
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerFoRM), Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia.
| | - Effendi Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, 47000 Sungai Buloh, Selangor, Malaysia.
| |
Collapse
|
45
|
Bahramzadeh A, Samavarchi Tehrani S, Goodarzi G, Seyyedebrahimi S, Meshkani R. Combination therapy of metformin and morin attenuates insulin resistance, inflammation, and oxidative stress in skeletal muscle of high-fat diet-fed mice. Phytother Res 2024; 38:912-924. [PMID: 38091524 DOI: 10.1002/ptr.8086] [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: 06/21/2023] [Revised: 10/25/2023] [Accepted: 11/26/2023] [Indexed: 02/15/2024]
Abstract
Lipid accumulation, inflammation, and oxidative stress are the most important causes of muscle insulin resistance. The aim of this study was to investigate the single and combined treatment effects of metformin (MET) and morin (MOR) on lipid accumulation, inflammation, and oxidative stress in the skeletal muscle of mice fed a high-fat diet. The mice were supplemented with MET (230 mg/kg diet), MOR (100 mg/kg diet), and MET + MOR for 9 weeks. Our results revealed that single treatment with MET or MOR, and with a stronger effect of MET + MOR combined treatment, reduced body weight gain, improved glucose intolerance and enhanced Akt phosphorylation in the muscle tissue. In addition, plasma and muscle triglyceride levels were decreased after treatment with MET and MOR. The expression of genes involved in macrophage infiltration and polarization and pro-inflammatory cytokines showed that MET + MOR combined treatment, significantly reduced inflammation in the muscle. Furthermore, combined treatment of MET + MOR with greater efficacy than the single treatment improved several oxidative stress markers in the muscle. Importantly, combined treatment of MET and MOR could increase the expression of nuclear factor erythroid 2-related factor 2, the master regulator of the antioxidant response. These findings suggest that combination of MET with MOR might ameliorate insulin resistance, inflammation, and oxidative stress in the skeletal muscle of mice fed high-fat diet.
Collapse
Affiliation(s)
- Arash Bahramzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadra Samavarchi Tehrani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathobiology and Laboratory Science, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - ShadiSadat Seyyedebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Lee H, Kim SY, Lim Y. Solanum melongena extract supplementation protected skeletal muscle and brain damage by regulation of BDNF/PGC1α/irisin pathway via brain function-related myokines in high-fat diet induced obese mice. J Nutr Biochem 2024; 124:109537. [PMID: 38030047 DOI: 10.1016/j.jnutbio.2023.109537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
In this study, we investigated the protective effects of SM on skeletal muscle and brain damage by regulation of BDNF/PGC1α/irisin pathway via brain function related myokines in high-fat diet-induced OB mice. OB was induced by high-fat diet for 6 weeks. SM extract (SME) was administered with 200 mg/kg BW (LSM) and 500 mg/kg BW (HSM) by oral gavage every day for 12 weeks. Behavior tests such as grip strength, Y-maze, and passive avoidance test were conducted to analyze muscle and cognitive function. Histopathological changes in skeletal muscle and brain were examined by hematoxylin and eosin staining and the protein levels of biomarkers related to oxidative stress, inflammation, protein degradation, neuro-plasticity, and cell cycling were measured by western blot. SME regulated morphological changes (muscle cross-sectional area: 1.23%, 1.40%; density of neurons in hippocampus:1.74%, 1.73%) in T2DM mice. Importantly, SME supplementation significantly increased several muscle-derived myokines which might influence the expression of neuronal markers in OB mice (FGF21: 1.27%, 1.34%; PGC1α: 1.0%, 1.32%; IRISIN: 1.9%, 1.08%; BDNF: 1.35%, 1.23%). Accordingly, SME activated hippocampal neurotrophic factors including BDNF (1.0%, 1.2%) and its associated PGC1α/irisin pathway (PGC1α :1.1%, 1.1%; IRISIN:1.1%, 0.9%) significantly. This study demonstrated the possibliy that protective myokines increased by SME supplementation may contribute to neuro-protection in OB mice. Taken together, the current study suggests that SME can be used to prevent skeletal muscle and brain damage in OB by protecting against oxidative stress and inflammatin via modulation of the BDNF/PGC1α/irisin pathway in the therapeutic approach of obese patients.
Collapse
Affiliation(s)
- Heaji Lee
- Department of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea
| | - Sun Yeou Kim
- Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-gu, Incheon, Republic of Korea
| | - Yunsook Lim
- Department of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Paul S, Bhardwaj J, Binukumar BK. Cdk5-mediated oligodendrocyte myelin breakdown and neuroinflammation: Implications for the link between Type 2 Diabetes and Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166986. [PMID: 38092158 DOI: 10.1016/j.bbadis.2023.166986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/07/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Oligodendrocytes, crucial myelinating glia in the central nervous system, play a vital role in maintaining axonal integrity and facilitating efficient nerve impulse conduction. The degradation of myelin in oligodendrocytes has been implicated in Alzheimer's disease (AD) and cognitive dysfunction. Interestingly, individuals with Type 2 Diabetes (T2D) have a significantly higher likelihood of developing cognitive impairment, possibly due to insulin resistance and glucose toxicity within the central nervous system (CNS). However, the precise relationship between these two disorders remains elusive. Our study proposes a potential link between T2D and AD, involving Cdk5-mediated breakdown of oligodendrocyte myelin and neuroinflammation. In the context of T2D, glucose toxicity in oligodendrocytes leads to heightened Cdk5 kinase activity and cPLA2 hyperactivation, resulting in chronic inflammation and myelin deterioration. This myelin breakdown in oligodendrocytes is thought to contribute to the development of AD and cognitive dysfunction. Notably, the administration of a Cdk5 inhibitor (TFP5) effectively alleviates neuroinflammation and myelin degradation. Moreover, our findings demonstrate heightened activity of Cdk5, cPLA2, and phospho-cPLA2 levels in the brain of a mouse model with Type 2 Diabetes (T2D). Hence, our findings suggest that targeting Cdk5 could be a promising therapeutic strategy to counteract AD pathogenesis in T2D-related conditions.
Collapse
Affiliation(s)
- Sangita Paul
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Juhi Bhardwaj
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - B K Binukumar
- CSIR Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
48
|
Aggarwal H, Gautam J, Kumari D, Gupta SK, Bajpai S, Chaturvedi K, Kumar Y, Dikshit M. Comparative profiling of gut microbiota and metabolome in diet-induced obese and insulin-resistant C57BL/6J mice. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119643. [PMID: 37996062 DOI: 10.1016/j.bbamcr.2023.119643] [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: 06/19/2023] [Revised: 10/28/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023]
Abstract
Diet-based models are commonly used to investigate obesity and related disorders. We conducted a comparative profiling of three obesogenic diets HFD, high fat diet; HFHF, high fat high fructose diet; and HFCD, high fat choline deficient diet to assess their impact on the gut microbiome and metabolome. After 20 weeks, we analyzed the gut microbiota and metabolomes of liver, plasma, cecal, and fecal samples. Fecal and plasma bile acids (BAs) and fecal short-chain fatty acids (SCFAs) were also examined. Significant changes were observed in fecal and cecal metabolites, with increased Firmicutes and decreased Bacteroidetes in the HFD, HFHF, and HFCD-fed mice compared to chow and LFD (low fat diet)-fed mice. Most BAs were reduced in plasma and fecal samples of obese groups, except taurocholic acid, which increased in HFCD mice's plasma. SCFAs like acetate and butyrate significantly decreased in obesogenic diet groups, while propionic acid specifically decreased in the HFCD group. Pathway analysis revealed significant alterations in amino acid, carbohydrate metabolism, and nucleic acid biosynthesis pathways in obese mice. Surprisingly, even LFD-fed mice showed distinct changes in microbiome and metabolite profiles compared to the chow group. This study provides insights into gut microbiome dysbiosis and metabolite alterations induced by obesogenic and LFD diets in various tissues. These findings aid in selecting suitable diet models to study the role of the gut microbiome and metabolites in obesity and associated disorders, with potential implications for understanding similar pathologies in humans.
Collapse
Affiliation(s)
- Hobby Aggarwal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Sneh Bajpai
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Kartikey Chaturvedi
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| | - Madhu Dikshit
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
49
|
Meda Venkata SP, Li H, Xu L, Wang JM. The Impact of Obesity on Diabetes Onset and Neovascularization in Mouse Models of Metabolic Stress. Int J Mol Sci 2024; 25:1214. [PMID: 38279212 PMCID: PMC10816627 DOI: 10.3390/ijms25021214] [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: 11/14/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Animal models of metabolic disorders are essential to studying pathogenic mechanisms and developing therapies for diabetes, but the induction protocols vary, and sexual dimorphism often exists. In a chronic diabetic model of diet-induced obesity (DIO) and low-dose streptozotocin (STZ)-induced hyperglycemia, blood glucose and lipid profiles were measured. The high-fat (HF) diet damaged insulin sensitivity and increased triglycerides, total cholesterol, LDL-cholesterol, HDL-cholesterol, and liver lipid deposition. STZ increased blood glucose and liver fibrosis with less effects on blood lipids or liver lipid deposition. The combination of DIO and STZ treatments led to significant liver lipid deposition and fibrosis. Female mice showed delayed body weight gain on HF diet and resisted STZ-induced hyperglycemia. However, once they developed DIO, which occurs around 26 weeks of HF diet, the female mice were prone to STZ-induced hyperglycemia. In hindlimb ischemia, male mice in the DIO-STZ group showed significantly worse neovascularization compared with DIO or STZ groups. The DIO-STZ females showed significantly worse recovery than the DIO-STZ males. Our observations suggest that DIO-STZ is a plausible model for studying metabolic and cardiovascular disorders in obesity and diabetes. Moreover, the findings in female animals stress the need to assess sexual dimorphism and investigate the underlying mechanisms that contribute to the worse vasculopathy manifestations in females in metabolic models.
Collapse
Affiliation(s)
- Sai Pranathi Meda Venkata
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
| | - Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
| | - Liping Xu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, 3122 Applebaum Building, Detroit, MI 48201, USA; (S.P.M.V.); (L.X.)
- Centers for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Karmanos Cancer Institute, Detroit, MI 48201, USA
| |
Collapse
|
50
|
Lamichhane G, Liu J, Lee SJ, Lee DY, Zhang G, Kim Y. Curcumin Mitigates the High-Fat High-Sugar Diet-Induced Impairment of Spatial Memory, Hepatic Metabolism, and the Alteration of the Gut Microbiome in Alzheimer's Disease-Induced (3xTg-AD) Mice. Nutrients 2024; 16:240. [PMID: 38257133 PMCID: PMC10818691 DOI: 10.3390/nu16020240] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
The escalating prevalence of metabolic diseases and an aging demographic has been correlated with a concerning rise in Alzheimer's disease (AD) incidence. This study aimed to access the protective effects of curcumin, a bioactive flavonoid from turmeric, on spatial memory, metabolic functions, and the regulation of the gut microbiome in AD-induced (3xTg-AD) mice fed with either a normal chow diet (NCD) or a high-fat high-sugar diet (HFHSD). Our findings revealed an augmented susceptibility of the HFHSD-fed 3xTg-AD mice for weight gain and memory impairment, while curcumin supplementation demonstrated a protective effect against these changes. This was evidenced by significantly reduced body weight gain and improved behavioral and cognitive function in the curcumin-treated group. These improvements were substantiated by diminished fatty acid synthesis, altered cholesterol metabolism, and suppressed adipogenesis-related pathways in the liver, along with modified synaptic plasticity-related pathways in the brain. Moreover, curcumin enriched beneficial gut microbiota, including Oscillospiraceae and Rikenellaceae at the family level, and Oscillibacter, Alistipes, Pseudoflavonifractor, Duncaniella, and Flintibacter at the genus level. The observed alteration in these gut microbiota profiles suggests a potential crosswalk in the liver and brain for regulating metabolic and cognitive functions, particularly in the context of obesity-associated cognitive disfunction, notably AD.
Collapse
Affiliation(s)
- Gopal Lamichhane
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Jing Liu
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (J.L.); (G.Z.)
| | - Su-Jeong Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Da-Yeon Lee
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (J.L.); (G.Z.)
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA; (G.L.); (S.-J.L.); (D.-Y.L.)
| |
Collapse
|