1
|
Salberg S, Macowan M, Doshen A, Yamakawa GR, Sgro M, Marsland B, Henderson LA, Mychasiuk R. A high fat, high sugar diet exacerbates persistent post-surgical pain and modifies the brain-microbiota-gut axis in adolescent rats. Neuroimage 2025; 307:121057. [PMID: 39870258 DOI: 10.1016/j.neuroimage.2025.121057] [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: 08/12/2024] [Revised: 01/11/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025] Open
Abstract
Persistent post-surgical pain (PPSP) occurs in a proportion of patients following surgical interventions. Research suggests that specific microbiome components are important for brain development and function, with recent studies demonstrating that chronic pain results in changes to the microbiome. Consumption of a high fat, high sugar (HFHS) diet can drastically alter composition of the microbiome and is a modifiable risk factor for many neuroinflammatory conditions. Therefore, we investigated how daily consumption of a HFHS diet modified the development of PPSP, brain structure and function, and the microbiome. In addition, we identified significant correlations between the microbiome and brain in animals with PPSP. Male and female rats were maintained on a control or HFHS diet. Animals were further allocated to a sham or surgery on postnatal day (p) p35. The von Frey task measured mechanical nociceptive sensitivity at a chronic timepoint (p65-67). Between p68-72 rats underwent in-vivo MRI to examine brain volume and diffusivity. At p73 fecal samples were used for downstream 16 s rRNA sequencing. Spearman correlation analyses were performed between individual microbial abundance and MRI diffusivity to determine if specific bacterial species were associated with PPSP-induced brain changes. We found that consumption of a HFHS diet exacerbated PPSP in adolescents. The HFHS diet reduced overall brain volume and increased white and grey matter density. The HFHS diet interacted with the surgical intervention to modify diffusivity in numerous brain regions which were associated with specific changes to the microbiome. These findings demonstrate that premorbid characteristics can influence the development of PPSP and advance our understanding of the contribution that the microbiome has on function of the brain-microbiota-gut axis.
Collapse
Affiliation(s)
- Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia
| | - Matthew Macowan
- Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Angela Doshen
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, NSW, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia
| | - Marissa Sgro
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia
| | - Benjamin Marsland
- Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, NSW, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Gastroenterology, Immunology, Neuroscience (GIN) Discovery Program, Australia.
| |
Collapse
|
2
|
Shrivastava P, Lu Y, Su S, Kobayashi Y, Zhao Y, Lien N, Masoud AR, Lukiw WJ, Hong S. Maresin-like 1 Ameliorates Neuropathology of Alzheimer's Disease in Brains of a Transgenic Mouse Model. Biomedicines 2024; 12:2865. [PMID: 39767773 PMCID: PMC11673747 DOI: 10.3390/biomedicines12122865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
(1) Background: Impeded resolution of inflammation contributes substantially to the pathogenesis of Alzheimer's disease (AD); consequently, resolving inflammation is pivotal to the amelioration of AD pathology. This can potentially be achieved by the treatment with specialized pro-resolving lipid mediators (SPMs), which should resolve neuroinflammation in brains. (2) Methods: Here, we report the histological effects of long-term treatment with an SPM, maresin-like 1 (MarL1), on AD pathogenesis in a transgenic 5xFAD mouse model. (3) Results: MarL1 treatment reduced Aβ overload, curbed the loss of neurons in brains especially cholinergic neurons associated with cleaved-caspase-3-associated apoptotic degeneration, reduced microgliosis and the pro-inflammatory M1 polarization of microglia, curbed the AD-associated decline in anti-inflammatory Iba1+Arg-1+-M2 microglia, inhibited phenotypic switching to pro-inflammatory N1 neutrophils, promoted the blood-brain barrier-associated tight-junction protein claudin-5 and decreased neutrophil leakage in 5xFAD brains, and induced the switch of neutrophils toward the inflammation-resolving N2 phenotype. (4) Conclusions: Long-term administration of MarL1 mitigates AD-related neuropathogenesis in brains by curbing neuroinflammation and neurodegeneration, based on the histological results. These findings provide preclinical leads and mechanistic insights for the development of MarL1 into an effective modality to ameliorate AD pathogenesis.
Collapse
Affiliation(s)
- Pallavi Shrivastava
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
| | - Yan Lu
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
| | - Shanchun Su
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
| | - Yuichi Kobayashi
- Department of Bioengineering, Tokyo Institute of Technology, Box B-52, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
- Organization for the Strategic Coordination of Research and Intellectual Properties, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Yuhai Zhao
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
| | - Nathan Lien
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
| | - Abdul-Razak Masoud
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
| | - Walter J. Lukiw
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
- Department of Ophthalmology, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA
| | - Song Hong
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA (Y.Z.); (N.L.); (A.-R.M.)
- Department of Ophthalmology, School of Medicine, Louisiana State University Health New Orleans, 2020 Gravier St., New Orleans, LA 70112, USA
| |
Collapse
|
3
|
Arenas-Mosquera D, Cerny N, Cangelosi A, Geoghegan P, Malchiodi E, De Marzi M, Pinto A, Goldstein J. High-fat and high-carbohydrate diets worsen the mouse brain susceptibility to damage produced by enterohemorrhagic Escherichia coli Shiga toxin 2. Heliyon 2024; 10:e39871. [PMID: 39553573 PMCID: PMC11564992 DOI: 10.1016/j.heliyon.2024.e39871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/19/2024] Open
Abstract
Background Nutrition quality could be one of the reasons why, in the face of a Shiga toxin-producing enterohemorrhagic Escherichia coli outbreak, some patients experience more profound deleterious effects than others, including unfortunate deaths. Thus, the aim of this study was to determine whether high-fat and/or high-carbohydrate diets could negatively modulate the deleterious action of Shiga toxin 2 on ventral anterior and ventral lateral thalamic nuclei and the internal capsule, the neurological centers responsible for motor activity. Methods Mice were fed a regular, high-fat, high-carbohydrate diet or a combination of both previous to the intravenous administration of Shiga toxin 2 or vehicle. Four days after intravenous administration, mice were subjected to behavioral tests and then sacrificed for histological and immunofluorescence assays to determine alterations in the neurovascular unit at the cellular and functional levels. Statistical analysis was performed using one-way analysis of variance followed by Bonferroni post hoc test. The criterion for significance was p = 0.0001 for all experiments. Results The high-fat and the high-carbohydrate diets significantly heightened the deleterious effect of Stx2, while the combination of both diets yielded the worst results, including endothelial glycocalyx and oligodendrocyte alterations, astrocyte and microglial reactivity, neurodegeneration, and motor and sensitivity impairment. Conclusions In view of the results presented here, poor nutrition could negatively influence patients affected by Stx2 at a neurological level. Systemic effects, however, cannot be ruled out.
Collapse
Affiliation(s)
- D. Arenas-Mosquera
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica ''Houssay" (IFIBIO), Laboratorio de Neurofisiopatología, Facultad de Medicina, Paraguay 2155 Piso 7, 1121, Ciudad de Buenos Aires, Argentina
| | - N. Cerny
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología e Instituto de Estudios de La Inmunidad Humoral (IDEHU), UBA-CONICET, Junín 956 Piso 4, 1113, Ciudad de Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología e Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Paraguay 2155 Piso 12, 1121, Ciudad de Buenos Aires, Argentina
| | - A. Cangelosi
- Centro Nacional de Control de Calidad de Biológicos (CNCCB), "ANLIS, Dr. Carlos G. Malbrán", Avenida Vélez Sarsfield 563, 1282, Ciudad de Buenos Aires, Argentina
| | - P.A. Geoghegan
- Centro Nacional de Control de Calidad de Biológicos (CNCCB), "ANLIS, Dr. Carlos G. Malbrán", Avenida Vélez Sarsfield 563, 1282, Ciudad de Buenos Aires, Argentina
| | - E.L. Malchiodi
- Universidad de Buenos Aires, IDEHU-CONICET, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Junín 956, Piso 4°, 1113, Ciudad de Buenos Aires, Argentina
| | - M. De Marzi
- Universidad Nacional de Luján, Departamento de Ciencias Básicas, Ruta 5 y Avenida Constitución (6700) Luján, Buenos Aires, Argentina, Universidad Nacional de Luján, Instituto de Ecología y Desarrollo Sustentable (INEDES)-CONICET, Laboratorio de Inmunología, Ruta 5 y Avenida Constitución (6700) Luján, Buenos Aires, Argentina
| | - A. Pinto
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica ''Houssay" (IFIBIO), Laboratorio de Neurofisiopatología, Facultad de Medicina, Paraguay 2155 Piso 7, 1121, Ciudad de Buenos Aires, Argentina
| | - J. Goldstein
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Fisiología y Biofísica ''Houssay" (IFIBIO), Laboratorio de Neurofisiopatología, Facultad de Medicina, Paraguay 2155 Piso 7, 1121, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
4
|
Sweetat S, Shabat MB, Theotokis P, Suissa N, Karafoulidou E, Touloumi O, Abu-Fanne R, Abramsky O, Wolf G, Saada A, Lotan A, Grigoriadis N, Rosenmann H. Ovariectomy and High Fat-Sugar-Salt Diet Induced Alzheimer's Disease/Vascular Dementia Features in Mice. Aging Dis 2024; 15:2284-2300. [PMID: 38913044 PMCID: PMC11346392 DOI: 10.14336/ad.2024.03110] [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: 01/29/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
While the vast majority of Alzheimer's disease (AD) is non-familial, the animal models of AD that are commonly used for studying disease pathogenesis and development of therapy are mostly of a familial form. We aimed to generate a model reminiscent of the etiologies related to the common late-onset Alzheimer's disease (LOAD) sporadic disease that will recapitulate AD/dementia features. Naïve female mice underwent ovariectomy (OVX) to accelerate aging/menopause and were fed a high fat-sugar-salt diet to expose them to factors associated with increased risk of development of dementia/AD. The OVX mice fed a high fat-sugar-salt diet responded by dysregulation of glucose/insulin, lipid, and liver function homeostasis and increased body weight with slightly increased blood pressure. These mice developed AD-brain pathology (amyloid and tangle pathologies), gliosis (increased burden of astrocytes and activated microglia), impaied blood vessel density and neoangiogenesis, with cognitive impairment. Thus, OVX mice fed on a high fat-sugar-salt diet imitate a non-familial sporadic/environmental form of AD/dementia with vascular damage. This model is reminiscent of the etiologies related to the LOAD sporadic disease that represents a high portion of AD patients, with an added value of presenting concomitantly AD and vascular pathology, which is a common condition in dementia. Our model can, thereby, provide a valuable tool for studying disease pathogenesis and for the development of therapeutic approaches.
Collapse
Affiliation(s)
- Sahar Sweetat
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. Hadassah BrainLabs-National Knowledge Center for Research on Brain Diseases, Hadassah-Hebrew University Medical Center, Jerusalem Israel
| | - Moti Ben Shabat
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. Hadassah BrainLabs-National Knowledge Center for Research on Brain Diseases, Hadassah-Hebrew University Medical Center, Jerusalem Israel
| | - Paschalis Theotokis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Nir Suissa
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. Hadassah BrainLabs-National Knowledge Center for Research on Brain Diseases, Hadassah-Hebrew University Medical Center, Jerusalem Israel
| | - Eleni Karafoulidou
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Olga Touloumi
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Rami Abu-Fanne
- Department of Clinical Biochemistry, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Oded Abramsky
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Gilly Wolf
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. Hadassah BrainLabs-National Knowledge Center for Research on Brain Diseases, Hadassah-Hebrew University Medical Center, Jerusalem Israel
- Biological Psychiatry Laboratory, Hadassah Hebrew University Medical Center, Jerusalem Israel Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Psychology, School of Psychology and Social Sciences, Achva Academic College, Be'er Tuvia, Israel
| | - Ann Saada
- Department of Genetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amit Lotan
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. Hadassah BrainLabs-National Knowledge Center for Research on Brain Diseases, Hadassah-Hebrew University Medical Center, Jerusalem Israel
- Biological Psychiatry Laboratory, Hadassah Hebrew University Medical Center, Jerusalem Israel Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nikolaos Grigoriadis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Greece
| | - Hanna Rosenmann
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel. Hadassah BrainLabs-National Knowledge Center for Research on Brain Diseases, Hadassah-Hebrew University Medical Center, Jerusalem Israel
| |
Collapse
|
5
|
Merchak AR, Bolen ML, Tansey MG, Menees KB. Thinking outside the brain: Gut microbiome influence on innate immunity within neurodegenerative disease. Neurotherapeutics 2024; 21:e00476. [PMID: 39482179 PMCID: PMC11585893 DOI: 10.1016/j.neurot.2024.e00476] [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: 08/19/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 11/03/2024] Open
Abstract
The complex network of factors that contribute to neurodegeneration have hampered the discovery of effective preventative measures. While much work has focused on brain-first therapeutics, it is becoming evident that physiological changes outside of the brain are the best target for early interventions. Specifically, myeloid cells, including peripheral macrophages and microglia, are a sensitive population of cells whose activity can directly impact neuronal health. Myeloid cell activity includes cytokine production, migration, debris clearance, and phagocytosis. Environmental measures that can modulate these activities range from toxin exposure to diet. However, one of the most influential mediators of myeloid fitness is the gut microenvironment. Here, we review the current data about the role of myeloid cells in gastrointestinal disorders, Parkinson's disease, dementia, and multiple sclerosis. We then delve into the gut microbiota modulating therapies available and clinical evidence for their use in neurodegeneration. Modulating lifestyle and environmental mediators of inflammation are one of the most promising interventions for neurodegeneration and a systematic and concerted effort to examine these factors in healthy aging is the next frontier.
Collapse
Affiliation(s)
- Andrea R Merchak
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - MacKenzie L Bolen
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| | - Kelly B Menees
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
6
|
Shah J, Orosz T, Singh A, Laxma SP, Gross RE, Smith N, Vroegop S, Sudler S, Porter JT, Colon M, Jun L, Babu JR, Shim M, Broderick TL, Al-Nakkash L. Influence of Exercise and Genistein to Mitigate the Deleterious Effects of High-Fat High-Sugar Diet on Alzheimer's Disease-Related Markers in Male Mice. Int J Mol Sci 2024; 25:9019. [PMID: 39201705 PMCID: PMC11354341 DOI: 10.3390/ijms25169019] [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: 07/08/2024] [Revised: 08/09/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
The prevalence of obesity and related consequences, including insulin resistance and Alzheimer's-like neuropathology, has increased dramatically. Contributing to this prevalence is the shift in lifestyle preference away from wholesome foods and exercise to the Western-style diet and sedentarism. Despite advances in drug development, a healthy diet and regular exercise remain the most effective approaches to mitigating the unwanted sequelae of diet-induced obesity on brain health. In this study, we used the high-fat high-sugar (HFHS) mouse model of neurodegeneration to examine the effects of exercise training (HFHS+Ex), genistein treatment (HFHS+Gen), and combination treatment (HFHS+Ex+Gen) on proteins relating to neurodegeneration in the brain of male mice. After a period of 12 weeks, as expected, HFHS feeding increased body weight, adipose tissue weight, and systemic plasma inflammation (TNF-α) compared to lean mice fed a standard diet. HFHS feeding also increased protein expression of brain markers of insulin resistance (pGSK-3β, p-IR), apoptosis (caspase 3), early neurofibrillary tangles (CP13), and amyloid-beta precursor (CT20). Compared to HFHS mice, Ex decreased body weight, plasma TNF-α, and expression of pGSK-3β, caspase 3, CP13, amyloid-β precursor (22c11), and ADAM10. Treatment with Gen was equally protective on these markers and decreased the expression of p-IR. Combination treatment with Ex and Gen afforded the greatest overall benefits, and this group exhibited the greatest reduction in body and adipose tissue weight and all brain markers, except for 22c11 and ADAM10, which were decreased compared to mice fed an HFHS diet. In addition, levels of 4G8, which detects protein levels of amyloid-β, were decreased with combination treatment. Our results indicate that exercise training, genistein supplementation, or combination treatment provide varying degrees of neuroprotection from HFHS feeding-induced Alzheimer's pathology. Future perspectives could include evaluating moderate exercise regimens in combination with dietary supplementation with genistein in humans to determine whether the same benefits translate clinically.
Collapse
Affiliation(s)
- Juhi Shah
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Tyler Orosz
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Avneet Singh
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Savan Parameshwar Laxma
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Rachel E. Gross
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Nicholas Smith
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Spencer Vroegop
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - Sydney Sudler
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
| | - James T. Porter
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce 00732, Puerto Rico; (J.T.P.); (M.C.)
| | - Maria Colon
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce 00732, Puerto Rico; (J.T.P.); (M.C.)
| | - Lauren Jun
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Jeganathan R. Babu
- Department of Nutritional Sciences, Auburn University, Auburn, AL 36849, USA
| | - Minsub Shim
- Department of Biochemistry, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Thomas L. Broderick
- Department of Physiology, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Layla Al-Nakkash
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA (T.O.); (S.P.L.); (R.E.G.); (S.V.); (S.S.)
- Department of Physiology, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| |
Collapse
|
7
|
Liu S, Deng Y, Liu H, Fu Z, Wang Y, Zhou M, Feng Z. Causal Relationship between Meat Intake and Biological Aging: Evidence from Mendelian Randomization Analysis. Nutrients 2024; 16:2433. [PMID: 39125314 PMCID: PMC11313912 DOI: 10.3390/nu16152433] [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: 06/23/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Existing research indicates that different types of meat have varying effects on health and aging, but the specific causal relationships remain unclear. This study aimed to explore the causal relationship between different types of meat intake and aging-related phenotypes. This study employed Mendelian randomization (MR) to select genetic variants associated with meat intake from large genomic databases, ensuring the independence and pleiotropy-free nature of these instrumental variables (IVs), and calculated the F-statistic to evaluate the strength of the IVs. The validity of causal estimates was assessed through sensitivity analyses and various MR methods (MR-Egger, weighted median, inverse-variance weighted (IVW), simple mode, and weighted mode), with the MR-Egger regression intercept used to test for pleiotropy bias and Cochran's Q test employed to evaluate the heterogeneity of the results. The findings reveal a positive causal relationship between meat consumers and DNA methylation PhenoAge acceleration, suggesting that increased meat intake may accelerate the biological aging process. Specifically, lamb intake is found to have a positive causal effect on mitochondrial DNA copy number, while processed meat consumption shows a negative causal effect on telomere length. No significant causal relationships were observed for other types of meat intake. This study highlights the significant impact that processing and cooking methods have on meat's role in health and aging, enhancing our understanding of how specific types of meat and their preparation affect the aging process, providing a theoretical basis for dietary strategies aimed at delaying aging and enhancing quality of life.
Collapse
Affiliation(s)
| | | | | | | | | | - Meijuan Zhou
- Department of Radiation medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (Y.D.); (H.L.); (Z.F.); (Y.W.)
| | - Zhijun Feng
- Department of Radiation medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (Y.D.); (H.L.); (Z.F.); (Y.W.)
| |
Collapse
|
8
|
Nasab MG, Heidari A, Sedighi M, Shakerian N, Mirbeyk M, Saghazadeh A, Rezaei N. Dietary inflammatory index and neuropsychiatric disorders. Rev Neurosci 2024; 35:21-33. [PMID: 37459114 DOI: 10.1515/revneuro-2023-0047] [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: 04/17/2023] [Accepted: 06/24/2023] [Indexed: 01/10/2024]
Abstract
Neuropsychiatric disorders (NPDs) are considered a potential threat to mental health. Inflammation predominantly plays a role in the pathophysiology of NPDs. Dietary patterns are widely postulated to be involved in the physiological response to inflammation. This review aims to discuss the literature on how dietary inflammatory index (DII) is related to inflammation and, consequently, NPDs. After comprehensive scrutiny in different databases, the articles that investigated the relation of DII score and various NPDs and psychological circumstances were included. The association between dietary patterns and mental disorders comprising depression, anxiety, and stress proved the role of a proinflammatory diet in these conditions' exacerbation. Aging is another condition closely associated with DII. The impact of proinflammatory and anti-inflammatory diet on sleep quality indicated related disorders like sleep latency and day dysfunctions among the different populations are in relation with the high DII score. The potential effects of genetic backgrounds, dietary patterns, and the gut microbiome on DII are discussed as well. To plan preventive or therapeutic interventions considering the DII, these factors, especially genetic variations, should be considered as there is a growing body of literature indicating the role of personalized medicine in different NPDs. To the best of our knowledge, there is a limited number of RCTs on this subject, so future research should evaluate the causality via RCTs and look for therapeutic interventions with an eye on personalized medicine using information about DII in NPDs.
Collapse
Affiliation(s)
- Mahsa Golshani Nasab
- Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | - Arash Heidari
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammadreza Sedighi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Narges Shakerian
- Student Research Committee, School of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Musculoskeletal Rehabilitation Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Ahvaz, Iran
| | - Mona Mirbeyk
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amene Saghazadeh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Meta Cognition Interest Group (MCIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
9
|
Mishra PS, Kumar A, Kaur K, Jaitak V. Recent Developments in Coumarin Derivatives as Neuroprotective Agents. Curr Med Chem 2024; 31:5702-5738. [PMID: 37455459 DOI: 10.2174/0929867331666230714160047] [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: 03/01/2023] [Revised: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Neurodegenerative diseases are among the diseases that cause the foremost burden on the health system of the world. The diseases are multifaceted and difficult to treat because of their complex pathophysiology, which includes protein aggregation, neurotransmitter breakdown, metal dysregulation, oxidative stress, neuroinflammation, excitotoxicity, etc. None of the currently available therapies has been found to be significant in producing desired responses without any major side effects; besides, they only give symptomatic relief otherwise indicated off-episode relief. Targeting various pathways, namely choline esterase, monoamine oxidase B, cannabinoid system, metal chelation, β-secretase, oxidative stress, etc., may lead to neurodegeneration. By substituting various functional moieties over the coumarin nucleus, researchers are trying to produce safer and more effective neuroprotective agents. OBJECTIVES This study aimed to review the current literature to produce compounds with lower side effects using coumarin as a pharmacophore. METHODS In this review, we have attempted to compile various synthetic strategies that have been used to produce coumarin and various substitutional strategies used to produce neuroprotective agents from the coumarin pharmacophore. Moreover, structure-activity relationships of substituting coumarin scaffold at various positions, which could be instrumental in designing new compounds, were also discussed. RESULTS The literature review suggested that coumarins and their derivatives can act as neuroprotective agents following various mechanisms. CONCLUSION Various studies have demonstrated the neuroprotective activity of coumarin due to an oxaheterocyclic loop, which allows binding with a broad array of proteins, thus motivating researchers to explore its potential as a lead against various neurodegenerative diseases.
Collapse
Affiliation(s)
- Prakash Shyambabu Mishra
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda (Pb.), 151401, India
| | - Amit Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda (Pb.), 151401, India
| | - Kamalpreet Kaur
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda (Pb.), 151401, India
| | - Vikas Jaitak
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda (Pb.), 151401, India
| |
Collapse
|
10
|
Denman CR, Park SM, Jo J. Gut-brain axis: gut dysbiosis and psychiatric disorders in Alzheimer's and Parkinson's disease. Front Neurosci 2023; 17:1268419. [PMID: 38075261 PMCID: PMC10704039 DOI: 10.3389/fnins.2023.1268419] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/30/2023] [Indexed: 02/17/2025] Open
Abstract
Gut dysbiosis and psychiatric symptoms are common early manifestations of Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases, characterised by progressive neuron loss and pathological protein accumulation, impose debilitating effects on patients. Recently, these pathological proteins have been linked with gut dysbiosis and psychiatric disorders. The gut-brain axis links the enteric and central nervous systems, acting as a bidirectional communication pathway to influence brain function and behavior. The relationship triad between gut dysbiosis, psychiatric disorders, and neurodegeneration has been investigated in pairs; however, evidence suggests that they are all interrelated and a deeper understanding is required to unravel the nuances of neurodegenerative diseases. Therefore, this review aims to summarise the current literature on the roles of gut dysbiosis and psychiatric disorders in pathological protein-related neurodegenerative diseases. We discussed how changes in the gut environment can influence the development of psychiatric symptoms and the progression of neurodegeneration and how these features overlap in AD and PD. Moreover, research on the interplay between gut dysbiosis, psychiatric disorders, and neurodegeneration remains in its early phase. In this review, we highlighted potential therapeutic approaches aimed at mitigating gastrointestinal problems and psychiatric disorders to alter the rate of neurodegeneration. Further research to assess the molecular mechanisms underlying AD and PD pathogenesis remains crucial for developing more effective treatments and achieving earlier diagnoses. Moreover, exploring non-invasive, early preventive measures and interventions is a relatively unexplored but important avenue of research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Charlotte R. Denman
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Junghyun Jo
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, Republic of Korea
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
11
|
Mechanism of metformin regulation in central nervous system: Progression and future perspectives. Biomed Pharmacother 2022; 156:113686. [DOI: 10.1016/j.biopha.2022.113686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022] Open
|
12
|
Alzahrani JM, Murray KO, Gambino BJ, Garcia CK, Sheikh LH, Cusack KJ, Laitano O, Clanton TL. Neuromotor deficits and altered physiological responses to repeated exertional heat stroke exposures in mice. Am J Physiol Regul Integr Comp Physiol 2022; 323:R951-R961. [PMID: 36279505 PMCID: PMC9722251 DOI: 10.1152/ajpregu.00152.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022]
Abstract
Exertional heat stroke (EHS) is a life-threatening illness that can lead to negative health outcomes. Using a "severe" preclinical mouse model of EHS, we tested the hypotheses that one EHS exposure results in altered susceptibility to a subsequent EHS and reduced neuromotor performance. Female C57BL/6 mice underwent two protocols, 2 wk apart, either an EHS trial (EHS) or a sham exercise control trial (EXC). For EHS, mice ran in a forced running wheel at 37.5°C/40% relative humidity until loss of consciousness, followed by a slow cooling protocol (2 h recovery at 37.5°C). EXC mice exercised equally but in ∼22°C. Mice were randomized into three groups: 1) EXC-EXC (two consecutive EXC, n = 6, 2) EHS-EXC (EHS followed by EXC, n = 5), and 3) EHS-EHS (repeated EHS, n = 9). Mice underwent noninvasive neuromotor and behavioral tests during recovery and isolated soleus force measurements at the end of recovery. At the first EHS, mice reached average peak core temperatures (Tc,max) of 42.4°C, (46% mortality). On the second EHS, average Tc,max was reduced by ∼0.7°C (P < 0.05; mortality 18%). After the first EHS, both EHS-EX and EHS-EHS showed significant reductions in maximum strength (24 h and 1 wk post). After the second EHS, strength, horizontal rotation, hindlimb tone, suspended hindlimb splay, trunk curl, and provoked biting continued to decline in the EHS-EHS group. In conclusion, exposure to a second EHS after 2 wk leads to increased exercise times in the heat, symptom limitation at a lower Tc,max, and greater deficits in neuromotor and behavioral function during recovery.
Collapse
Affiliation(s)
- Jamal M Alzahrani
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Kevin O Murray
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Bryce J Gambino
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Christian K Garcia
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Laila H Sheikh
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Kevin J Cusack
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Thomas L Clanton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida
| |
Collapse
|
13
|
Wang K, Liu H, Hu Q, Wang L, Liu J, Zheng Z, Zhang W, Ren J, Zhu F, Liu GH. Epigenetic regulation of aging: implications for interventions of aging and diseases. Signal Transduct Target Ther 2022; 7:374. [PMID: 36336680 PMCID: PMC9637765 DOI: 10.1038/s41392-022-01211-8] [Citation(s) in RCA: 232] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
Aging is accompanied by the decline of organismal functions and a series of prominent hallmarks, including genetic and epigenetic alterations. These aging-associated epigenetic changes include DNA methylation, histone modification, chromatin remodeling, non-coding RNA (ncRNA) regulation, and RNA modification, all of which participate in the regulation of the aging process, and hence contribute to aging-related diseases. Therefore, understanding the epigenetic mechanisms in aging will provide new avenues to develop strategies to delay aging. Indeed, aging interventions based on manipulating epigenetic mechanisms have led to the alleviation of aging or the extension of the lifespan in animal models. Small molecule-based therapies and reprogramming strategies that enable epigenetic rejuvenation have been developed for ameliorating or reversing aging-related conditions. In addition, adopting health-promoting activities, such as caloric restriction, exercise, and calibrating circadian rhythm, has been demonstrated to delay aging. Furthermore, various clinical trials for aging intervention are ongoing, providing more evidence of the safety and efficacy of these therapies. Here, we review recent work on the epigenetic regulation of aging and outline the advances in intervention strategies for aging and age-associated diseases. A better understanding of the critical roles of epigenetics in the aging process will lead to more clinical advances in the prevention of human aging and therapy of aging-related diseases.
Collapse
Affiliation(s)
- Kang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Huicong Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Qinchao Hu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China
- Hospital of Stomatology, Sun Yat-sen University, 510060, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 510060, Guangzhou, China
| | - Lingna Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Jiaqing Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China
| | - Zikai Zheng
- University of Chinese Academy of Sciences, 100049, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, 100049, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China
| | - Jie Ren
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, 100101, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Fangfang Zhu
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, Shanghai, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 100101, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China.
| |
Collapse
|
14
|
Hong S, Nagayach A, Lu Y, Peng H, Duong QVA, Pham NB, Vuong CA, Bazan NG. A high fat, sugar, and salt Western diet induces motor-muscular and sensory dysfunctions and neurodegeneration in mice during aging: Ameliorative action of metformin. CNS Neurosci Ther 2021; 27:1458-1471. [PMID: 34510763 PMCID: PMC8611779 DOI: 10.1111/cns.13726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/18/2023] Open
Abstract
Aims To explore the novel linkage between a Western diet combining high saturated fat, sugar, and salt (HFSS) and neurological dysfunctions during aging as well as Metformin intervention, we assessed cerebral cortex abnormalities associated with sensory and motor dysfunctions and cellular and molecular insights in brains using HFSS‐fed mice during aging. We also explored the effect of Metformin treatment on these mice. Methods C57BL/6 mice were fed with HFSS and treated with metformin from 20 to 22 months of age, resembling human aging from 56 to 68 years of age (an entry phase of the aged portion of lifespan). Results The motor and sensory cortexes in mice during aging after HFSS diet showed: (A) decreased motor‐muscular and sensory functions; (B) reduced inflammation‐resolving Arg‐1+ microglia; (C) increased inflammatory iNOs+ microglia and TNFα levels; (D) enhanced abundance of amyloid‐β peptide and of phosphorylated Tau. Metformin attenuated these changes. Conclusion A HFSS‐combined diet caused motor‐muscular and sensory dysfunctions, neuroinflammation, and neurodegeneration, whereas metformin counteracted these effects. Our findings show neuroinflammatory consequences of a HFSS diet in aging. Metformin curbs the HFSS‐related neuroinflammation eliciting neuroprotection.
Collapse
Affiliation(s)
- Song Hong
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Aarti Nagayach
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Yan Lu
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Hongying Peng
- Biostatistics, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Quoc-Viet A Duong
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Nicholas B Pham
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Christopher A Vuong
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.,Department of Ophthalmology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|