1
|
Chen J, Liu C, Li L, Tao W, Zhang X, Zhao S, Wang C, Huang L. Exogenous leptin alleviates glutamate-excitotoxic injury caused by cerebral ischemia-reperfusion in mice by affecting the expression of glutamate transporters. Brain Res 2024; 1845:149201. [PMID: 39197570 DOI: 10.1016/j.brainres.2024.149201] [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: 06/09/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Ischemic stroke is characterized by high morbidity and mortality and a lack of effective therapeutic interventions. Leptin plays an important role in regulating oxidative stress, angiogenesis, hematopoiesis, etc. Although recent studies have found a neuroprotective effect of leptin, little is known about its role in cerebral ischemia. This study explores the possible roles and potential preventative mechanisms of leptin in cerebral ischemia-reperfusion injury (CIRI). An in vivo middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model was used to replicate the CIRI model, low (0.5 mg/kg), medium (1 mg/kg) and high (2 mg/kg) concentrations of leptin were injected intraperitoneally immediately after inserting the embolic line. After 1.5 h of ischemia and 24 h of reperfusion, we examined the neural function of the mice, collected brain tissue for histological examination, and screened for the optimal concentrations of leptin intervention. On this basis, we observed the changes of cortical apoptosis injury, intracellular calcium fluorescence intensity and astrocyte glial fibrillary acidic protein (GFAP) expression and morphological changes. In addition, we also tested the expression of transporters and metabolism-related enzymes (VGLUT-1, VGLUT-2, GLAST, GLT-1, GS, ATP1α1), the expression of inflammatory factors and the content of glutamate (Glu). Compared with the I/R group, we found that leptin improved neurological deficits, reduced the area of infarcts, maintained the normal morphology of astrocytes (AST), downregulated the expression of VGLUT-1, and upregulates the expression of GLT-1 and GLAST, thereby reducing the content of Glu in the synaptic cleft. Our studies suggest that leptin may have a neuroprotective effect by decreasing the excitotoxicity of glutamate.
Collapse
Affiliation(s)
- Jie Chen
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Chenxu Liu
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Li Li
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - WeiTing Tao
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Xiaonan Zhang
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Shidi Zhao
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China
| | - Chun Wang
- Department of General Medicine, Second Affiliated Hospital of Bengbu Medical University, Bengbu Anhui 233040, China.
| | - Li Huang
- Department of Pathophysiology, Bengbu Medical University, Bengbu Anhui 233030, China; Basic and Clinical Key Laboratory of Cardiovascular and Cerebrovascular Diseases of Bengbu Medical University, Bengbu Anhui 233030, China.
| |
Collapse
|
2
|
Medina-Vera D, López-Gambero AJ, Verheul-Campos J, Navarro JA, Morelli L, Galeano P, Suárez J, Sanjuan C, Pacheco-Sánchez B, Rivera P, Pavon-Morón FJ, Rosell-Valle C, Fonseca FRD. Therapeutic Efficacy of the Inositol D-Pinitol as a Multi-Faceted Disease Modifier in the 5×FAD Humanized Mouse Model of Alzheimer's Amyloidosis. Nutrients 2024; 16:4186. [PMID: 39683582 DOI: 10.3390/nu16234186] [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: 11/06/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Alzheimer's disease (AD), a leading cause of dementia, lacks effective long-term treatments. Current therapies offer temporary relief or fail to halt its progression and are often inaccessible due to cost. AD involves multiple pathological processes, including amyloid beta (Aβ) deposition, insulin resistance, tau protein hyperphosphorylation, and systemic inflammation accelerated by gut microbiota dysbiosis originating from a leaky gut. Given this context, exploring alternative therapeutic interventions capable of addressing the multifaceted components of AD etiology is essential. METHODS This study suggests D-Pinitol (DPIN) as a potential treatment modifier for AD. DPIN, derived from carob pods, demonstrates insulin-sensitizing, tau hyperphosphorylation inhibition, and antioxidant properties. To test this hypothesis, we studied whether chronic oral administration of DPIN (200 mg/kg/day) could reverse the AD-like disease progression in the 5×FAD mice. RESULTS Results showed that treatment of 5×FAD mice with DPIN improved cognition, reduced hippocampal Aβ and hyperphosphorylated tau levels, increased insulin-degrading enzyme (IDE) expression, enhanced pro-cognitive hormone circulation (such as ghrelin and leptin), and normalized the PI3K/Akt insulin pathway. This enhancement may be mediated through the modulation of cyclin-dependent kinase 5 (CDK5). DPIN also protected the gut barrier and microbiota, reducing the pro-inflammatory impact of the leaky gut observed in 5×FAD mice. DPIN reduced bacterial lipopolysaccharide (LPS) and LPS-associated inflammation, as well as restored intestinal proteins such as Claudin-3. This effect was associated with a modulation of gut microbiota towards a more balanced bacterial composition. CONCLUSIONS These findings underscore DPIN's promise in mitigating cognitive decline in the early AD stages, positioning it as a potential disease modifier.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
- Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Antonio J López-Gambero
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- INSERM, Neurocentre Magendie, University of Bordeaux, 33000 Bordeaux, France
| | - Julia Verheul-Campos
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Juan A Navarro
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Facultad de Medicina, Universidad de Málaga, 29010 Málaga, Spain
| | - Laura Morelli
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina
| | - Pablo Galeano
- Laboratory of Brain Aging and Neurodegeneration, Fundación Instituto Leloir (IIBBA-CONICET), Av. Patricias Argentinas 435, Ciudad Autónoma de Buenos Aires C1405BWE, Argentina
| | - Juan Suárez
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], 29001 Málaga, Spain
| | - Carlos Sanjuan
- Euronutra S.L. Calle Johannes Kepler, 3, 29590 Málaga, Spain
| | - Beatriz Pacheco-Sánchez
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Patricia Rivera
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Francisco J Pavon-Morón
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Unidad de Gestión Clínica del Corazón-CIBERCV (Enfermedades Cardiovasculares), Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Cristina Rosell-Valle
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Grupo de Neuropsicofarmacología, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Unidades Clínicas de Neurología y Salud Mental, 29010 Málaga, Spain
- Andalusian Network for Clinical and Translational Research in Neurology [NEURO-RECA], 29001 Málaga, Spain
| |
Collapse
|
3
|
Atkinson RAK, Collins JM, Sreedharan J, King AE, Fernandez-Martos CM. Alterations to metabolic hormones in amyotrophic lateral sclerosis and frontotemporal dementia postmortem human tissue. J Neuropathol Exp Neurol 2024; 83:907-916. [PMID: 38917432 PMCID: PMC11487092 DOI: 10.1093/jnen/nlae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024] Open
Abstract
Metabolic changes are observed in patients with both amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although regulation of metabolic processes in the CNS is predominantly carried out within the hypothalamus, extra-hypothalamic CNS areas contain metabolic hormone receptors, including those for leptin (LEPR), insulin (INSR), and neuropeptide Y (NPY), indicating that they may play a role in biological processes underlying pathogenic disease processes. The status of these hormones within regions vulnerable in ALS/FTD is not well described. This study sought to determine whether the expression of these hormones and their receptors is altered in pathology-rich regions in cases of human FTD (superior frontal gyrus and insular cortex) and ALS (primary motor cortex and lumbar spinal cord) with TDP-43 pathology compared to matched healthy controls. LEPR mRNA was increased within the superior frontal gyrus of FTD cases and within primary motor cortex and lumbar spinal cord of ALS cases; INSR mRNA was increased in superior frontal gyrus and insular cortex of FTD cases. NPY protein was decreased in primary motor cortex and lumbar spinal cord of ALS cases. Our results demonstrate that metabolic hormones undergo complex alterations in ALS and FTD and suggest that these hormones could play critical roles in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Rachel A K Atkinson
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jessica M Collins
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jemeen Sreedharan
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, United Kingdom
| | - Anna E King
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Carmen M Fernandez-Martos
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| |
Collapse
|
4
|
Mostaar A, Behroozi Z, MotamedNezhad A, Taherkhani S, Mojarad N, Ramezani F, Janzadeh A, Hajimirzaie P. The effect of intra spinal administration of cerium oxide nanoparticles on central pain mechanism: An experimental study. J Bioenerg Biomembr 2024; 56:505-515. [PMID: 39102102 DOI: 10.1007/s10863-024-10033-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/21/2024] [Indexed: 08/06/2024]
Abstract
This study investigated Cerium oxide nanoparticles (CeONPs) effect on central neuropathic pain (CNP). The compressive method of spinal cord injury (SCI) model was used for pain induction. Three groups were formed by a random allocation of 24 rats. In the treatment group, CeONPs were injected above and below the lesion site immediately after inducing SCI. pain symptoms were evaluated using acetone, Radian Heat, and Von Frey tests weekly for six weeks. Finally, we counted fibroblasts using H&E staining. We evaluated the expression of Cx43, GAD65 and HDAC2 proteins using the western blot method. The analysis of results was done by PRISM software. At the end of the study, we found that CeONPs reduced pain symptoms to levels similar to those observed in normal animals. CeONPs also increased the expression of GAD65 and Cx43 proteins but did not affect HDAC2 inhibition. CeONPs probably have a pain-relieving effect on chronic pain by potentially preserving GAD65 and Cx43 protein expression and hindering fibroblast infiltration.
Collapse
Affiliation(s)
- Ahmad Mostaar
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali MotamedNezhad
- College of Veterinary Medicine, Islamic Azad University, Karaj, Alborz, Iran
| | - Sourosh Taherkhani
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Negin Mojarad
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI, 48859, USA
| | - Fatemeh Ramezani
- Physiology Research Center, , Iran University of Medical Sciences, Tehran, Iran.
| | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Pooya Hajimirzaie
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Fernandes C, Forny-Germano L, Andrade MM, Lyra E Silva NM, Ramos-Lobo AM, Meireles F, Tovar-Moll F, Houzel JC, Donato J, De Felice FG. Leptin receptor reactivation restores brain function in early-life Lepr-deficient mice. Brain 2024; 147:2706-2717. [PMID: 38650574 PMCID: PMC11292908 DOI: 10.1093/brain/awae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/14/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024] Open
Abstract
Obesity is a chronic disease caused by excessive fat accumulation that impacts the body and brain health. Insufficient leptin or leptin receptor (LepR) is involved in the disease pathogenesis. Leptin is involved with several neurological processes, and it has crucial developmental roles. We have previously demonstrated that leptin deficiency in early life leads to permanent developmental problems in young adult mice, including an imbalance in energy homeostasis, alterations in melanocortin and the reproductive system and a reduction in brain mass. Given that in humans, obesity has been associated with brain atrophy and cognitive impairment, it is important to determine the long-term consequences of early-life leptin deficiency on brain structure and memory function. Here, we demonstrate that leptin-deficient (LepOb) mice exhibit altered brain volume, decreased neurogenesis and memory impairment. Similar effects were observed in animals that do not express the LepR (LepRNull). Interestingly, restoring the expression of LepR in 10-week-old mice reverses brain atrophy, in addition to neurogenesis and memory impairments in older animals. Our findings indicate that leptin deficiency impairs brain development and memory, which are reversible by restoring leptin signalling in adulthood.
Collapse
Affiliation(s)
- Caroline Fernandes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Mayara M Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Natalia M Lyra E Silva
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Fernanda Meireles
- D’Or Institute for Research and Education, Rio de Janeiro, RJ 22281-100, Brazil
| | - Fernanda Tovar-Moll
- D’Or Institute for Research and Education, Rio de Janeiro, RJ 22281-100, Brazil
| | - Jean Christophe Houzel
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen’s University, Kingston, ON K7L 3N6, Canada
- D’Or Institute for Research and Education, Rio de Janeiro, RJ 22281-100, Brazil
| |
Collapse
|
6
|
Li Y, Yang Y, Ye B, Lin Y. Maternal high fat diet programs spatial learning and central leptin signaling in mouse offspring in a sex-specific manner. Physiol Behav 2024; 281:114580. [PMID: 38714271 DOI: 10.1016/j.physbeh.2024.114580] [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: 02/08/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/09/2024]
Abstract
Environmental factors in early life have been demonstrated to increase the risk of neurodevelopmental disorders in offspring, especially the deficiency of the cognitive ability. Leptin has emerged as a key hormone that conveys information on energy stores, but there is growing appreciation that leptin signaling may also play an important role in neurodevelopment. The present study aimed to investigate whether maternal HFD exposure impairs the offspring learning and memory through the programming of central leptin system. We observed that hippocampus-dependent learning and memory were impaired in male but not female offspring from HFD-fed maternal ancestors (C57BL/6 mice), as assessed by novel object recognition and Morris water maze tests. Moreover, the chromatin immunoprecipitation results revealed the maternal HFD consumption led to the increasement in the binding of the histone marker H3K9me3 in male offspring, which mediates gene silencing in the leptin receptor promoter region. Furthermore, there was an increase in the expression of the histone methylase SUV39H1 in male but not female offspring, which regulates H3K9me3. Additionally, it has been observed that IL-6 and IL-1 also could lead to similar alternations when acting on cultured hippocampal neurons in vitro. Taken together, our data suggest that maternal HFD consumption influences male offspring hippocampal cognitive performance in a sex-specific manner, and central leptin signaling may serve as the cross-talk between maternal diet and cognitive impairment in offspring.
Collapse
Affiliation(s)
- YiQuan Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ya Yang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - BoWei Ye
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - YuanShao Lin
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
7
|
Tarhan M, Hartl T, Shchyglo O, Colitti-Klausnitzer J, Kuhla A, Breuer TM, Manahan-Vaughan D. Changes in hippocampal volume, synaptic plasticity and amylin sensitivity in an animal model of type 2 diabetes are associated with increased vulnerability to amyloid-beta in advancing age. Front Aging Neurosci 2024; 16:1373477. [PMID: 38974903 PMCID: PMC11224464 DOI: 10.3389/fnagi.2024.1373477] [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: 01/19/2024] [Accepted: 05/28/2024] [Indexed: 07/09/2024] Open
Abstract
Type-2 diabetes (T2D) is a metabolic disorder that is considered a risk factor for Alzheimer's disease (AD). Cognitive impairment can arise due to hypoglycemia associated with T2D, and hyperamylinemia associated with insulin resistance can enhance AD pathology. We explored whether changes occur in the hippocampus in aging (6-12 months old) female V-Lep○b-/- transgenic (tg) mice, comprising an animal model of T2D. We also investigated whether an increase in vulnerability to Aβ (1-42), a known pathological hallmark of AD, is evident. Using magnetic resonance imaging we detected significant decreases in hippocampal brain volume in female tg-mice compared to wild-type (wt) littermates. Long-term potentiation (LTP) was impaired in tg compared to wt mice. Treatment of the hippocampus with Aβ (1-42) elicited a stronger debilitation of LTP in tg compared to wt mice. Treatment with an amylin antagonist (AC187) significantly enhanced LTP in wt and tg mice, and rescued LTP in Aβ (1-42)-treated tg mice. Taken together our data indicate that a T2D-like state results in an increased vulnerability of the hippocampus to the debilitating effects of Aβ (1-42) and that effects are mediated in part by changes in amylin receptor signaling.
Collapse
Affiliation(s)
- Melih Tarhan
- Department of Neurophysiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- International Graduate School of Neuroscience, Bochum, Germany
| | - Tim Hartl
- Department of Neurophysiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- International Graduate School of Neuroscience, Bochum, Germany
| | - Olena Shchyglo
- Department of Neurophysiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | | | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | | | - Denise Manahan-Vaughan
- Department of Neurophysiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- International Graduate School of Neuroscience, Bochum, Germany
| |
Collapse
|
8
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [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] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l'Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
9
|
Hamilton K, Morrow K, Markantoni E, Harvey J. Leptin prevents aberrant targeting of tau to hippocampal synapses via PI 3 kinase driven inhibition of GSK3β. J Neurochem 2023; 167:520-537. [PMID: 37822142 DOI: 10.1111/jnc.15980] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Amyloid-β (Aβ) and hyper-phosphorylated tau are key hallmarks of Alzheimer's disease (AD), with an accumulation of both proteins linked to hippocampal synaptic dysfunction. Recent evidence indicates that Aβ drives mis-localisation of tau from axons to synapses, resulting in AMPA receptor (AMPAR) internalisation and impaired excitatory synaptic function. These tau-driven synaptic impairments are thought to underlie the cognitive deficits in AD. Consequently, limiting the synapto-toxic effects of tau may prevent AD-related cognitive deficits. Increasing evidence links leptin dysfunction with higher AD risk, and numerous studies have identified neuroprotective properties of leptin in AD models of Aβ-induced toxicity. However, it is unclear if leptin protects against tau-related synaptic dysfunction. Here we show that Aβ1-42 significantly increases dendritic and synaptic levels of tau and p-tau in hippocampal neurons, and these effects were blocked by leptin. In accordance with GSK-3β being involved in tau phosphorylation, the protective effects of leptin involve PI 3-kinase (PI3K) activation and inhibition of GSK-3β. Aβ1-42 -driven synaptic targeting of tau was associated with the removal of GluA1-containing AMPARs from synapses, which was also inhibited by leptin-driven inhibition of GSK-3β. Direct application of oligomeric tau to hippocampal neurons caused internalisation of GluA1-containing AMPARs and this effect was blocked by prior application of leptin. Similarly, leptin prevented the ability of tau to block induction of activity-dependent long-term potentiation (LTP) at hippocampal SC-CA1 synapses. These findings increase our understanding of the neuroprotective actions of leptin in the early pre-clinical stages of AD and further validate the leptin system as a therapeutic target in AD.
Collapse
Affiliation(s)
- Kirsty Hamilton
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Kate Morrow
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Ermione Markantoni
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
10
|
Fan X, Yuan W, Huang W, Lin Z. Recent progress in leptin signaling from a structural perspective and its implications for diseases. Biochimie 2023; 212:60-75. [PMID: 37080418 DOI: 10.1016/j.biochi.2023.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/14/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
As a multi-potency cytokine, leptin not only plays a crucial role in controlling weight and energy homeostasis but also participates in the metabolic balance in the human body. Leptin is a small helical protein with a molecular weight of 16 kDa. It can interact with multiple subtypes of its receptors to initiate intracellular signal transduction and exerts physiological effects. Disturbances in leptin signaling may lead to obesity and a variety of metabolic diseases. Leptin was also found to be a critical factor in many diseases of the elderly. In this review, we focus on recent advances in the structural and molecular mechanisms of leptin signaling through its receptors with the aim of a deeper understanding of leptin-related diseases.
Collapse
Affiliation(s)
- Xiao Fan
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China
| | - Wensu Yuan
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China
| | - Weidong Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia, 750004, PR China.
| | - Zhi Lin
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China.
| |
Collapse
|
11
|
Borges JCO, Oliveira VAB, Serdan TDA, Silva FLR, Santos CS, Pauferro JRB, Ribas ASF, Manoel R, Pereira ACG, Correa IS, Pereira JNB, Bazotte RB, Levada-Pires AC, Pithon-Curi TC, Gorjão R, Curi R, Hirabara SM, Masi LN. Brain glucose hypometabolism and hippocampal inflammation in Goto-Kakizaki rats. Braz J Med Biol Res 2023; 56:e12742. [PMID: 37377307 DOI: 10.1590/1414-431x2023e12742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/05/2023] [Indexed: 06/29/2023] Open
Abstract
Brain glucose hypometabolism and neuroinflammation are early pathogenic manifestations in neurological disorders. Neuroinflammation may also disrupt leptin signaling, an adipokine that centrally regulates appetite and energy balance by acting on the hypothalamus and exerting neuroprotection in the hippocampus. The Goto-Kakizaki (GK) rat is a non-obese type 2 diabetes mellitus (T2DM) animal model used to investigate diabetes-associated molecular mechanisms without obesity jeopardizing effects. Wistar and GK rats received the maintenance adult rodent diet. Also, an additional control group of Wistar rats received a high-fat and high-sugar diet (HFHS) provided by free consumption of condensed milk. All diets and water were provided ad libitum for eight weeks. Brain glucose uptake was evaluated by 2-deoxy-2-[fluorine-18] fluoro-D-glucose under basal (saline administration) or stimulated (CL316,243, a selective β3-AR agonist) conditions. The animals were fasted for 10-12 h, anesthetized, and euthanized. The brain was quickly dissected, and the hippocampal area was sectioned and stored at -80°C in different tubes for protein and RNA analyses on the same animal. GK rats exhibited attenuated brain glucose uptake compared to Wistar animals and the HFHS group under basal conditions. Also, the hippocampus of GK rats displayed upregulated leptin receptor, IL-1β, and IL-6 gene expression and IL-1β and the subunit of the transcription factor NF-κB (p-p65) protein expression. No significant alterations were detected in the hippocampus of HFHS rats. Our data indicated that a genetic predisposition to T2DM has significant brain deteriorating features, including brain glucose hypometabolism, neuroinflammation, and leptin signaling disruption in the hippocampal area.
Collapse
Affiliation(s)
- J C O Borges
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - V A B Oliveira
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - T D A Serdan
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - F L R Silva
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - C S Santos
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - J R B Pauferro
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - A S F Ribas
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - R Manoel
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - A C G Pereira
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - I S Correa
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - J N B Pereira
- Seção de Produção de Imunobiológicos, Bioindustrial Centro, Instituto Butantan, São Paulo, SP, Brasil
| | - R B Bazotte
- Programa de Pós-Graduação em Ciências Farmacêuticas, Departamento de Farmacologia e Terapêutica, Universidade Estadual de Maringá, Maringá, PR, Brasil
| | - A C Levada-Pires
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - T C Pithon-Curi
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - R Gorjão
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - R Curi
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
- Seção de Produção de Imunobiológicos, Bioindustrial Centro, Instituto Butantan, São Paulo, SP, Brasil
| | - S M Hirabara
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - L N Masi
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
- Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| |
Collapse
|
12
|
Hafez LM, Aboudeya HM, Matar NA, El-Sebeay AS, Nomair AM, El-Hamshary SA, Nomeir HM, Ibrahim FAR. Ameliorative effects of zinc supplementation on cognitive function and hippocampal leptin signaling pathway in obese male and female rats. Sci Rep 2023; 13:5072. [PMID: 36977735 PMCID: PMC10050324 DOI: 10.1038/s41598-023-31781-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Obesity has been associated with cognitive impairments, increasing the probability of developing dementia. Recently, zinc (Zn) supplementation has attracted an increasing attention as a therapeutic agent for cognitive disorders. Here, we investigated the potential effects of low and high doses of Zn supplementation on cognitive biomarkers and leptin signaling pathway in the hippocampus of high fat diet (HFD)-fed rats. We also explored the impact of sex difference on the response to treatment. Our results revealed a significant increase in body weight, glucose, triglycerides (TG), total cholesterol (TC), total lipids and leptin levels in obese rats as compared to controls. HFD feeding also reduced brain-derived neurotrophic factor (BDNF) levels and increased acetylcholinesterase (AChE) activity in the hippocampus of both sexes. The low and high doses of Zn supplementation improved glucose, TG, leptin, BDNF levels and AChE activity in both male and female obese rats compared to untreated ones. Additionally, downregulated expression of leptin receptor (LepR) gene and increased levels of activated signal transducer and activator of transcription 3 (p-STAT3) that observed in hippocampal tissues of obese rats were successfully normalized by both doses of Zn. In this study, the male rats were more vulnerable to HFD-induced weight gain, most of the metabolic alterations and cognition deficits than females, whereas the female obese rats were more responsive to Zn treatment. In conclusion, we suggest that Zn treatment may be effective in ameliorating obesity-related metabolic dysfunction, central leptin resistance and cognitive deficits. In addition, our findings provide evidence that males and females might differ in their response to Zn treatment.
Collapse
Affiliation(s)
- Lamia M Hafez
- Human Nutrition Department, Regional Center for Food and Feed-Agricultural Research Center, Alexandria, Egypt
| | | | - Noura A Matar
- Department of Histochemistry and Cell Biology Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Ashraf S El-Sebeay
- Human Nutrition Department, Regional Center for Food and Feed-Agricultural Research Center, Alexandria, Egypt
| | - Azhar Mohamed Nomair
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | - Hanan Mohamed Nomeir
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fawziya A R Ibrahim
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, 165, Horreya Avenue, Hadara, Alexandria, Egypt.
| |
Collapse
|
13
|
Guzmán-Ruiz MA, Guerrero-Vargas NN, Lagunes-Cruz A, González-González S, García-Aviles JE, Hurtado-Alvarado G, Mendez-Hernández R, Chavarría-Krauser A, Morin JP, Arriaga-Avila V, Buijs RM, Guevara-Guzmán R. Circadian modulation of microglial physiological processes and immune responses. Glia 2023; 71:155-167. [PMID: 35971989 PMCID: PMC10087862 DOI: 10.1002/glia.24261] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/11/2022] [Accepted: 08/01/2022] [Indexed: 11/09/2022]
Abstract
Microglia is considered the central nervous system (CNS) resident macrophages that establish an innate immune response against pathogens and toxins. However, the recent studies have shown that microglial gene and protein expression follows a circadian pattern; several immune activation markers and clock genes are expressed rhythmically without the need for an immune stimulus. Furthermore, microglia responds to an immune challenge with different magnitudes depending on the time of the day. This review examines the circadian control of microglia function and the possible physiological implications. For example, we discuss that synaptic prune is performed in the cortex at a certain moment of the day. We also consider the implications of daily microglial function for maintaining biological rhythms like general activity, body temperature, and food intake. We conclude that the developmental stage, brain region, and pathological state are not the only factors to consider for the evaluation of microglial functions; instead, emerging evidence indicates that circadian time as an essential aspect for a better understanding of the role of microglia in CNS physiology.
Collapse
Affiliation(s)
- Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Alejandra Lagunes-Cruz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Shellye González-González
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Jesús Enrique García-Aviles
- Área de Neurociencias, Departamento de Biología de la Reproducción, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, México City, Mexico
| | | | - Rebeca Mendez-Hernández
- Instituto Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, Mexico
| | - Anahí Chavarría-Krauser
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Jean-Pascal Morin
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Virginia Arriaga-Avila
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Ruud M Buijs
- Instituto Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, Mexico
| | - Rosalinda Guevara-Guzmán
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| |
Collapse
|
14
|
Liu CC, Wang QH, Xin JY, Liu YH, Zeng F, Chen DW, Li HY, Yi X, Zeng GH, Wang YJ, Xiang Y, Chen Y. Association of Adipokines with Alzheimer's Disease in a Chinese Cohort. J Alzheimers Dis 2023; 96:523-533. [PMID: 37807776 DOI: 10.3233/jad-220860] [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: 10/10/2023]
Abstract
BACKGROUND The correlation between plasma adipose factor levels and Alzheimer's patients is not entirely clear. OBJECTIVE We aimed to investigate associations between AD and plasma levels of three adipokines including plasma adiponectin, leptin, and resistin. METHODS A single-center, cross-sectional study recruited AD patients (n = 148) and cognitively normal (CN) controls (n = 110). The multivariate logistic regression analysis was applied to determine associations of adiponectin, leptin, and resistin with the presence of AD. The receiver operating characteristic (ROC) analysis was employed to determine the diagnostic power of adiponectin, leptin and resistin for AD. RESULTS After adjusted for the conventional risk factors, plasma levels of leptin (OR = 0.417, 95% CI: 0.272-0.638, p < 0.0001) and adiponectin (OR = 1.249, 95% CI: 1.151-1.354, p < 0.0001) were associated with the presence of AD. In total participants, the plasma adiponectin level was negatively correlated with MMSE scores (p < 0.0001) and was positively with CDR scores (p < 0.0001) and age (p < 0.0001). The plasma level of leptin was negatively correlated with CDR scores (p < 0.0001) and positively correlated with MMSE scores (p < 0.0001). Both adiponectin (p < 0. 0001) and leptin (p < 0. 0001) featured higher AUC than the random chance. CONCLUSIONS Plasma adiponectin and leptin were associated with the presence, symptomatic severity, and diagnostic power of AD, suggesting a potential role of adipokines in the pathogenesis of AD.
Collapse
Affiliation(s)
- Cheng-Chun Liu
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Qing-Hua Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Jia-Yan Xin
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Yu-Hao Liu
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Fan Zeng
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Dong-Wan Chen
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Hui-Yun Li
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Xu Yi
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| | - Yang Xiang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Chen
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China
| |
Collapse
|
15
|
Leptin alleviates endoplasmic reticulum stress induced by cerebral ischemia/reperfusion injury via the PI3K/Akt signaling pathway. Biosci Rep 2022; 42:232083. [PMID: 36367210 PMCID: PMC9744719 DOI: 10.1042/bsr20221443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Cerebral ischemic/reperfusion injury (CIRI) is a key factor for the prognosis of ischemic stroke (IS), the leading disease in terms of global disability and fatality rates. Recent studies have shown that endoplasmic reticulum stress (ERS) may be a target against CIRI and that leptin, a peptide hormone, has neuroprotective activity to mitigate CIRI. METHODS An in vitro CIRI model was induced in primary cortical neurons by oxygen-glucose deprivation and reoxygenation (OGD/R) after pretreatment with LY294002 (10 µmol/L) and/or leptin (0.4 mg/L), and cell viability, neuronal morphology and endoplasmic reticulum (ER) dysfunction were evaluated. An in vivo CIRI model was established in rats by middle cerebral artery occlusion and reperfusion (MCAO/R) after the injection of LY294002 (10 μmol/L) and/or leptin (1 mg/kg), and neurological function, infarct volume, cerebral pathological changes, the expression of ERS-related proteins and cell apoptosis were examined. RESULTS In vitro, leptin treatment improved the cell survival rate, ameliorated neuronal pathological morphology and alleviated OGD/R-induced ERS. In vivo, administration of leptin significantly reduced the infarct volume, neurological deficit scores and neuronal apoptosis as well as pathological alterations. In addition, leptin suppressed MCAO/R-induced ERS and may decrease apoptosis by inhibiting ERS-related death and caspase 3 activation. It also regulated expression of the antiapoptotic protein Bcl-2 and the proapoptotic protein Bax in the cortex. Furthermore, the inhibitory effect of leptin on ERS was significantly decreased by the effective phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002. CONCLUSIONS These results confirm that ERS plays an important role in CIRI and that leptin can inhibit the activation of ERS through the PI3K/Akt pathway, thereby alleviating CIRI. These findings provide novel therapeutic targets for IS.
Collapse
|
16
|
Santana LF, do Espirito Santo BLS, Tatara MB, Negrão FJ, Croda J, Alves FM, de Oliveira Filiú WF, Cavalheiro LF, Nazário CED, Asato MA, de Faria BB, do Nascimento VA, de Cássia Avellaneda Guimarães R, de Cássia Freitas K, Hiane PA. Effects of the Seed Oil of Carica papaya Linn on Food Consumption, Adiposity, Metabolic and Inflammatory Profile of Mice Using Hyperlipidic Diet. Molecules 2022; 27:6705. [PMID: 36235241 PMCID: PMC9570947 DOI: 10.3390/molecules27196705] [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: 07/28/2022] [Revised: 09/07/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Studies indicate that different parts of Carica papaya Linn have nutritional properties that mean it can be used as an adjuvant for the treatment of various pathologies. METHODS The fatty acid composition of the oil extracted from the seeds of Carica papaya Linn was evaluated by gas chromatography, and an acute toxicity test was performed. For the experiment, Swiss mice were fed a balanced or high-fat diet and supplemented with saline, soybean oil, olive oil, or papaya seed oil. Oral glucose tolerance and insulin sensitivity tests were performed. After euthanasia, adiposity, glycemia, total cholesterol and fractions, insulin, resistin, leptin, MCP-1, TNF-α, and IL-6 and the histology of the liver, pancreas, and adipose tissue were evaluated. RESULTS Papaya seed oil showed predominance of monounsaturated fatty acids in its composition. No changes were observed in the acute toxicity test. Had lower food intake in grams, and caloric intake and in the area of adipocytes without minimizing weight gain or adiposity and impacting the liver or pancreas. Reductions in total and non-HDL-c, LDL-c, and VLDL-c were also observed. The treatment had a hypoglycemic and protective effect on insulin resistance. Supplementation also resulted in higher leptin and lower insulin and cytokine resistance. CONCLUSIONS Under these experimental conditions, papaya seed oil led to higher amounts of monounsaturated fatty acids and had hypocholesterolemic, hypotriglyceridemic, and hypoglycemic effects.
Collapse
Affiliation(s)
- Lidiani Figueiredo Santana
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, Brazil
| | - Bruna Larissa Spontoni do Espirito Santo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, Brazil
| | - Mariana Bento Tatara
- Health Science Research Laboratory, Federal University of Grande Dourados, Dourados 79804-970, Brazil
| | - Fábio Juliano Negrão
- Health Science Research Laboratory, Federal University of Grande Dourados, Dourados 79804-970, Brazil
| | - Júlio Croda
- Oswaldo Cruz Foundation, Campo Grande 79074-460, Brazil
| | - Flávio Macedo Alves
- Laboratory of Botany, Institute of Biosciences, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, Brazil
| | - Wander Fernando de Oliveira Filiú
- Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79079-900, Brazil
| | | | | | - Marcel Arakaki Asato
- Medical School, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, Brazil
| | | | - Valter Aragão do Nascimento
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, Brazil
| | - Rita de Cássia Avellaneda Guimarães
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, Brazil
| | - Karine de Cássia Freitas
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, Brazil
| | - Priscila Aiko Hiane
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul (UFMS), Campo Grande 79070-900, Brazil
| |
Collapse
|
17
|
Huang X, Wang YJ, Xiang Y. Bidirectional communication between brain and visceral white adipose tissue: Its potential impact on Alzheimer's disease. EBioMedicine 2022; 84:104263. [PMID: 36122553 PMCID: PMC9490488 DOI: 10.1016/j.ebiom.2022.104263] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
A variety of axes between brain and abdominal organs have been reported, but the interaction between brain and visceral white adipose tissue (vWAT) remains unclear. In this review, we summarized human studies on the association between brain and vWAT, and generalized their interaction and the underlying mechanisms according to animal and cell experiments. On that basis, we come up with the concept of the brain-vWAT axis (BVA). Furthermore, we analyzed the potential mechanisms of involvement of BVA in the pathogenesis of Alzheimer's disease (AD), including vWAT-derived fatty acids, immunological properties of vWAT, vWAT-derived retinoic acid and vWAT-regulated insulin resistance. The proposal of BVA may expand our understanding to some extent of how the vWAT impacts on brain health and diseases, and provide a novel approach to study the pathogenesis and treatment strategies of neurodegenerative disorders.
Collapse
|
18
|
Ghosh-Swaby OR, Reichelt AC, Sheppard PAS, Davies J, Bussey TJ, Saksida LM. Metabolic hormones mediate cognition. Front Neuroendocrinol 2022; 66:101009. [PMID: 35679900 DOI: 10.1016/j.yfrne.2022.101009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 05/18/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Recent biochemical and behavioural evidence indicates that metabolic hormones not only regulate energy intake and nutrient content, but also modulate plasticity and cognition in the central nervous system. Disruptions in metabolic hormone signalling may provide a link between metabolic syndromes like obesity and diabetes, and cognitive impairment. For example, altered metabolic homeostasis in obesity is a strong determinant of the severity of age-related cognitive decline and neurodegenerative disease. Here we review the evidence that eating behaviours and metabolic hormones-particularly ghrelin, leptin, and insulin-are key players in the delicate regulation of neural plasticity and cognition. Caloric restriction and antidiabetic therapies, both of which affect metabolic hormone levels can restore metabolic homeostasis and enhance cognitive function. Thus, metabolic hormone pathways provide a promising target for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Olivia R Ghosh-Swaby
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada
| | - Amy C Reichelt
- Faculty of Health and Medical Sciences, Adelaide Medical School, Adelaide, Australia
| | - Paul A S Sheppard
- Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Jeffrey Davies
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Timothy J Bussey
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Lisa M Saksida
- Schulich School of Medicine and Dentistry, Neuroscience Program, Western University, London, ON, Canada; Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, ON, Canada.
| |
Collapse
|
19
|
Jacob L, Smith L, Koyanagi A, Konrad M, Haro JM, Shin JI, Kostev K. Sex-Differential Associations Between Body Mass Index and the Incidence of Dementia. J Alzheimers Dis 2022; 88:631-639. [PMID: 35662122 DOI: 10.3233/jad-220147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Little is known about the sex differences in the association between body mass index (BMI) and dementia in late life. OBJECTIVE Therefore, this retrospective cohort study aimed to analyze associations between BMI and dementia in older women and men separately in general practices in Germany. METHODS This study included patients followed in one of 832 general practices in Germany between 2006 and 2019 (index date: first visit date). Study variables included dementia (dependent variable), BMI (independent variable), age, sex, and comorbidities (control variables). Kaplan-Meier curves and adjusted Cox regression analyses were conducted to analyze associations between BMI and the 10-year incidence of dementia in women and men, separately. RESULTS There were 296,767 patients included in this study (mean [standard deviation] age 70.2 [5.9] years; 54.3% women). The proportion of underweight, normal weight, overweight, and obesity was 0.9%, 25.5%, 41.5%, and 32.1%, respectively. The 10-year incidence of dementia significantly decreased with increasing BMI, from 11.5% in women with underweight to 9.1% in those with obesity (log-rank p < 0.001). Respective figures in men were 12.0% and 8.2% (log-rank p < 0.001). In women, only overweight (versus normal weight) was significantly associated with dementia (HR = 0.93, 95% CI = 0.88-0.97). In contrast, in men, the only BMI category significantly associated with the incidence of dementia was underweight (HR = 1.58, 95% CI = 1.11-2.25). CONCLUSION In this study conducted in Germany, overweight was negatively associated with dementia in women, whereas there was a positive underweight-dementia relationship in men. More data are needed to confirm or refute these findings in other settings.
Collapse
Affiliation(s)
- Louis Jacob
- Research and Development Unit, Parc Sanitari Sant Joan de Déu, CIBERSAM, Dr. Antoni Pujadas, Sant Boi de Llobregat, Barcelona, Spain.,Faculty of Medicine, University of Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Lee Smith
- Centre for Health, Performance and Wellbeing, Anglia Ruskin University, Cambridge, UK
| | - Ai Koyanagi
- Research and Development Unit, Parc Sanitari Sant Joan de Déu, CIBERSAM, Dr. Antoni Pujadas, Sant Boi de Llobregat, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluis Companys, Barcelona, Spain
| | - Marcel Konrad
- Health & Social, FOM University of Applied Sciences for Economics and Management, Frankfurt am Main, Germany
| | - Josep Maria Haro
- Research and Development Unit, Parc Sanitari Sant Joan de Déu, CIBERSAM, Dr. Antoni Pujadas, Sant Boi de Llobregat, Barcelona, Spain
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | |
Collapse
|
20
|
Flores-Cordero JA, Pérez-Pérez A, Jiménez-Cortegana C, Alba G, Flores-Barragán A, Sánchez-Margalet V. Obesity as a Risk Factor for Dementia and Alzheimer's Disease: The Role of Leptin. Int J Mol Sci 2022; 23:5202. [PMID: 35563589 PMCID: PMC9099768 DOI: 10.3390/ijms23095202] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is a growing worldwide health problem, affecting many people due to excessive saturated fat consumption, lack of exercise, or a sedentary lifestyle. Leptin is an adipokine secreted by adipose tissue that increases in obesity and has central actions not only at the hypothalamic level but also in other regions and nuclei of the central nervous system (CNS) such as the cerebral cortex and hippocampus. These regions express the long form of leptin receptor LepRb, which is the unique leptin receptor capable of transmitting complete leptin signaling, and are the first regions to be affected by chronic neurocognitive deficits, such as mild cognitive impairment (MCI) and Alzheimer's Disease (AD). In this review, we discuss different leptin resistance mechanisms that could be implicated in increasing the risk of developing AD, as leptin resistance is frequently associated with obesity, which is a chronic low-grade inflammatory state, and obesity is considered a risk factor for AD. Key players of leptin resistance are SOCS3, PTP1B, and TCPTP whose signalling is related to inflammation and could be worsened in AD. However, some data are controversial, and it is necessary to further investigate the underlying mechanisms of the AD-causing pathological processes and how altered leptin signalling affects such processes.
Collapse
Affiliation(s)
| | | | | | | | | | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology and Immunology, Medical School, Virgen Macarena University Hospital, University of Seville, Av. Sánchez Pizjuan 4, 41009 Sevilla, Spain; (J.A.F.-C.); (A.P.-P.); (C.J.-C.); (G.A.); (A.F.-B.)
| |
Collapse
|
21
|
Haas SS, Myoraku A, Watson K, Robakis T, Frangou S, Abbasi F, Rasgon N. Lower functional hippocampal connectivity in healthy adults is jointly associated with higher levels of leptin and insulin resistance. Eur Psychiatry 2022; 65:e29. [PMID: 35492025 PMCID: PMC9158395 DOI: 10.1192/j.eurpsy.2022.21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Metabolic dysregulation is currently considered a major risk factor for hippocampal pathology. The aim of the present study was to characterize the influence of key metabolic drivers on functional connectivity of the hippocampus in healthy adults. METHODS Insulin resistance was directly quantified by measuring steady-state plasma glucose (SSPG) concentration during the insulin suppression test and fasting levels of insulin, glucose, leptin, and cortisol, and measurements of body mass index and waist circumference were obtained in a sample of healthy cognitively intact adults (n = 104). Resting-state neuroimaging data were also acquired for the quantification of hippocampal functional cohesiveness and integration with the major resting-state networks (RSNs). Data-driven analysis using unsupervised machine learning (k-means clustering) was then employed to identify clusters of individuals based on their metabolic and functional connectivity profiles. RESULTS K-means clustering identified two clusters of increasing metabolic deviance evidenced by cluster differences in the plasma levels of leptin (40.36 (29.97) vs. 27.59 (25.58) μg/L) and the degree of insulin resistance (SSPG concentration: 161.63 (65.27) vs. 125.72 (66.81) mg/dL). Individuals in the cluster with higher metabolic deviance showed lower functional cohesiveness within each hippocampus and lower integration of posterior and anterior components of the left and right hippocampus with the major RSNs. The two clusters did not differ in general intellectual ability or episodic memory. CONCLUSIONS We identified two clusters of individuals differentiated by abnormalities in insulin resistance, leptin levels, and hippocampal connectivity, with one of the clusters showing greater deviance. These findings support the link between metabolic dysregulation and hippocampal function even in nonclinical samples.
Collapse
Affiliation(s)
- Shalaila S. Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alison Myoraku
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kathleen Watson
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, California, USA
| | - Thalia Robakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fahim Abbasi
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Natalie Rasgon
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
22
|
Weerasinghe-Mudiyanselage PDE, Ang MJ, Kang S, Kim JS, Moon C. Structural Plasticity of the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:3349. [PMID: 35328770 PMCID: PMC8955928 DOI: 10.3390/ijms23063349] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/10/2022] Open
Abstract
Neuroplasticity is the capacity of neural networks in the brain to alter through development and rearrangement. It can be classified as structural and functional plasticity. The hippocampus is more susceptible to neuroplasticity as compared to other brain regions. Structural modifications in the hippocampus underpin several neurodegenerative diseases that exhibit cognitive and emotional dysregulation. This article reviews the findings of several preclinical and clinical studies about the role of structural plasticity in the hippocampus in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In this study, literature was surveyed using Google Scholar, PubMed, Web of Science, and Scopus, to review the mechanisms that underlie the alterations in the structural plasticity of the hippocampus in neurodegenerative diseases. This review summarizes the role of structural plasticity in the hippocampus for the etiopathogenesis of neurodegenerative diseases and identifies the current focus and gaps in knowledge about hippocampal dysfunctions. Ultimately, this information will be useful to propel future mechanistic and therapeutic research in neurodegenerative diseases.
Collapse
Affiliation(s)
- Poornima D. E. Weerasinghe-Mudiyanselage
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
- College of Veterinary Medicine, University of the Philippines Los Baños, Los Baños 4031, Philippines
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea; (P.D.E.W.-M.); (M.J.A.); (S.K.); (J.-S.K.)
| |
Collapse
|
23
|
Abedpoor N, Taghian F, Hajibabaie F. Physical activity ameliorates the function of organs via adipose tissue in metabolic diseases. Acta Histochem 2022; 124:151844. [PMID: 35045377 DOI: 10.1016/j.acthis.2022.151844] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/04/2022] [Accepted: 01/04/2022] [Indexed: 12/15/2022]
Abstract
Adipose tissue is a dynamic organ in the endocrine system that can connect organs by secreting molecules and bioactive. Hence, adipose tissue really plays a pivotal role in regulating metabolism, inflammation, energy homeostasis, and thermogenesis. Disruption of hub bioactive molecules secretion such as adipokines leads to dysregulate metabolic communication between adipose tissue and other organs in non-communicable disorders. Moreover, a sedentary lifestyle may be a risk factor for adipose tissue function. Physical inactivity leads to fat tissue accumulation and promotes obesity, Type 2 diabetes, cardiovascular disease, neurodegenerative disease, fatty liver, osteoporosis, and inflammatory bowel disease. On the other hand, physical activity may ameliorate and protect the body against metabolic disorders, triggering thermogenesis, metabolism, mitochondrial biogenesis, β-oxidation, and glucose uptake. Furthermore, physical activity provides an inter-organ association and cross-talk between different tissues by improving adipose tissue function, reprogramming gene expression, modulating molecules and bioactive factors. Also, physical activity decreases chronic inflammation, oxidative stress and improves metabolic features in adipose tissue. The current review focuses on the beneficial effect of physical activity on the cardiovascular, locomotor, digestive, and nervous systems. In addition, we visualize protein-protein interactions networks between hub proteins involved in dysregulating metabolic induced by adipose tissue.
Collapse
Affiliation(s)
- Navid Abedpoor
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Farzaneh Taghian
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Fatemeh Hajibabaie
- Department of Physiology, Medicinal Plants Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| |
Collapse
|
24
|
Frangou S, Abbasi F, Watson K, Haas SS, Antoniades M, Modabbernia A, Myoraku A, Robakis T, Rasgon N. Hippocampal volume reduction is associated with direct measure of insulin resistance in adults. Neurosci Res 2022; 174:19-24. [PMID: 34352294 PMCID: PMC9164143 DOI: 10.1016/j.neures.2021.07.006] [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: 03/30/2021] [Revised: 07/16/2021] [Accepted: 07/28/2021] [Indexed: 01/03/2023]
Abstract
Hippocampal integrity is highly susceptible to metabolic dysfunction, yet its mechanisms are not well defined. We studied 126 healthy individuals aged 23-61 years. Insulin resistance (IR) was quantified by measuring steady-state plasma glucose (SSPG) concentration during the insulin suppression test. Body mass index (BMI), adiposity, fasting insulin, glucose, leptin as well as structural neuroimaing with automatic hippocampal subfield segmentation were performed. Data analysis using unsupervised machine learning (k-means clustering) identified two subgroups reflecting a pattern of more pronounced hippocampal volume reduction being concurrently associated with greater adiposity and insulin resistance; the hippocampal volume reductions were uniform across subfields. Individuals in the most deviant subgroup were predominantly women (79 versus 42 %) with higher BMI [27.9 (2.5) versus 30.5 (4.6) kg/m2], IR (SSPG concentration, [156 (61) versus 123 (70) mg/dL] and leptinemia [21.7 (17.0) versus 44.5 (30.4) μg/L]. The use of person-based modeling in healthy individuals suggests that adiposity, insulin resistance and compromised structural hippocampal integrity behave as a composite phenotype; female sex emerged as risk factor for this phenotype.
Collapse
Affiliation(s)
- Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada,Corresponding author at: Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA., (S. Frangou), (N. Rasgon)
| | - Fahim Abbasi
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Katie Watson
- Department of Psychiatry, Stanford University School of Medicine, USA
| | - Shalaila S. Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mathilde Antoniades
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Alison Myoraku
- Department of Psychiatry, Stanford University School of Medicine, USA
| | - Thalia Robakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Natalie Rasgon
- Department of Psychiatry, Stanford University School of Medicine, USA,Corresponding author at: 401 Quarry Road, MC 5723, Palo Alto, CA 94304, USA
| |
Collapse
|
25
|
Tournissac M, Leclerc M, Valentin-Escalera J, Vandal M, Bosoi CR, Planel E, Calon F. Metabolic determinants of Alzheimer's disease: A focus on thermoregulation. Ageing Res Rev 2021; 72:101462. [PMID: 34534683 DOI: 10.1016/j.arr.2021.101462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a complex age-related neurodegenerative disease, associated with central and peripheral metabolic anomalies, such as impaired glucose utilization and insulin resistance. These observations led to a considerable interest not only in lifestyle-related interventions, but also in repurposing insulin and other anti-diabetic drugs to prevent or treat dementia. Body temperature is the oldest known metabolic readout and mechanisms underlying its maintenance fail in the elderly, when the incidence of AD rises. This raises the possibility that an age-associated thermoregulatory deficit contributes to energy failure underlying AD pathogenesis. Brown adipose tissue (BAT) plays a central role in thermogenesis and maintenance of body temperature. In recent years, the modulation of BAT activity has been increasingly demonstrated to regulate energy expenditure, insulin sensitivity and glucose utilization, which could also provide benefits for AD. Here, we review the evidence linking thermoregulation, BAT and insulin-related metabolic defects with AD, and we propose mechanisms through which correcting thermoregulatory impairments could slow the progression and delay the onset of AD.
Collapse
|
26
|
Liu M, Jiao Q, Du X, Bi M, Chen X, Jiang H. Potential Crosstalk Between Parkinson's Disease and Energy Metabolism. Aging Dis 2021; 12:2003-2015. [PMID: 34881082 PMCID: PMC8612621 DOI: 10.14336/ad.2021.0422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/22/2021] [Indexed: 01/22/2023] Open
Abstract
Parkinson's disease (PD) is characterized by the accumulation of alpha-synuclein (α-Syn) in the substantia nigra (SN) and the degeneration of nigrostriatal dopaminergic (DAergic) neurons. Some studies have reported that the pathology of PD originates from the gastrointestinal (GI) tract, which also serves as an energy portal, and develops upward along the neural pathway to the central nervous system (CNS), including the dorsal motor nucleus of vagus (DMV), SN, and hypothalamus, which are also involved in energy metabolism control. Therefore, we discuss the alterations of nuclei that regulate energy metabolism in the development of PD. In addition, due to their anti-inflammatory, antiapoptotic and antioxidative roles, metabolism-related peptides are involved in the progression of PD. Furthermore, abnormal glucose and lipid metabolism are common in PD patients and exacerbate the pathological changes in PD. Therefore, in this review, we attempt to explain the correlation between PD and energy metabolism, which may provide possible strategies for PD treatment.
Collapse
Affiliation(s)
- Meiqiu Liu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Saturated and unsaturated fat diets impair hippocampal glutamatergic transmission in adolescent mice. Psychoneuroendocrinology 2021; 133:105429. [PMID: 34624673 DOI: 10.1016/j.psyneuen.2021.105429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/10/2021] [Accepted: 09/20/2021] [Indexed: 11/22/2022]
Abstract
Consumption of high-fat diets (HFD) has been associated with neuronal plasticity deficits and cognitive disorders linked to the alteration of glutamatergic disorders in the hippocampus. As young individuals are especially vulnerable to the effects of nutrients and xenobiotics on cognition, we studied the effect of chronic consumption of saturated (SOLF) and unsaturated oil-enriched foods (UOLF) on: i) spatial memory; ii) hippocampal synaptic transmission and plasticity; and iii) gene expression of glutamatergic receptors and hormone receptors in the hippocampus of adolescent and adult mice. Our results show that both SOLF and UOLF impair spatial short-term memory. Accordingly, hippocampal synaptic plasticity mechanisms underlying memory, and gene expression of NMDA receptor subunits are modulated by both diets. On the other hand, PPARγ gene expression is specifically down-regulated in adolescent SOLF individuals and up-regulated in adult UOLF mice.
Collapse
|
28
|
Li C, Meng F, Lei Y, Liu J, Liu J, Zhang J, Liu F, Liu C, Guo M, Lu XY. Leptin regulates exon-specific transcription of the Bdnf gene via epigenetic modifications mediated by an AKT/p300 HAT cascade. Mol Psychiatry 2021; 26:3701-3722. [PMID: 33106599 PMCID: PMC8550971 DOI: 10.1038/s41380-020-00922-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 01/17/2023]
Abstract
Leptin is an adipocyte-derived hormone with pleiotropic functions affecting appetite and mood. While leptin's role in the regulation of appetite has been extensively studied in hypothalamic neurons, its function in the hippocampus, where it regulates mood-related behaviors, is poorly understood. Here, we show that the leptin receptor (LepRb) colocalizes with brain-derived neurotrophic factor (BDNF), a key player in the pathophysiology of major depression and the action of antidepressants, in the dentate gyrus of the hippocampus. Leptin treatment increases, whereas deficiency of leptin or leptin receptors decreases, total Bdnf mRNA levels, with distinct expression profiles of specific exons, in the hippocampus. Epigenetic analyses reveal that histone modifications, but not DNA methylation, underlie exon-specific transcription of the Bdnf gene induced by leptin. This is mediated by stimulation of AKT signaling, which in turn activates histone acetyltransferase p300 (p300 HAT), leading to changes in histone H3 acetylation and methylation at specific Bdnf promoters. Furthermore, deletion of Bdnf in the dentate gyrus, or specifically in LepRb-expressing neurons, abolishes the antidepressant-like effects of leptin. These findings indicate that leptin, acting via an AKT-p300 HAT epigenetic cascade, induces exon-specific Bdnf expression, which in turn is indispensable for leptin-induced antidepressant-like effects.
Collapse
Affiliation(s)
- Chen Li
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Shandong, China.
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Fantao Meng
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Shandong, China
| | - Yun Lei
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jing Liu
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jing Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Shandong, China
| | - Jingyan Zhang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Fang Liu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Cuilan Liu
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Shandong, China
| | - Ming Guo
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
29
|
Benefits of Exercise and Astaxanthin Supplementation: Are There Additive or Synergistic Effects? Antioxidants (Basel) 2021; 10:antiox10060870. [PMID: 34071514 PMCID: PMC8229412 DOI: 10.3390/antiox10060870] [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: 05/12/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 11/30/2022] Open
Abstract
A healthy lifestyle is essential for maintaining physical and mental health. Health promotion, with a particular emphasis on regular exercise and a healthy diet, is one of the emerging trends in healthcare. However, the way in which exercise training and nutrients from dietary intake interact with each other to promote additive, synergistic, or antagonistic effects on physiological functions leading to health promotion, and the possible underlying biomolecular mechanisms of such interactions, remain poorly understood. A healthy diet is characterized by a high intake of various bioactive compounds usually found in natural, organic, and fresh foodstuffs. Among these bioactive compounds, astaxanthin (ASX), a red carotenoid pigment especially found in seafood, has been recognized in the scientific literature as a potential nutraceutical due to its antioxidant, anti-inflammatory, and neurotrophic properties. Therefore, scientists are currently exploring whether this promising nutrient can increase the well-known benefits of exercise on health and disease prevention. Hence, the present review aimed to compile and summarize the current scientific evidence for ASX supplementation in association with exercise regimes, and evaluate the additive or synergistic effects on physiological functions and health when both interventions are combined. The new insights into the combination paradigm of exercise and nutritional supplementation raise awareness of the importance of integrative studies, particularly for future research directions in the field of health and sports nutrition science.
Collapse
|
30
|
Sahin GS, Luis Rodriguez-Llamas J, Dillon C, Medina I, Appleyard SM, Gaiarsa JL, Wayman GA. Leptin increases GABAergic synaptogenesis through the Rho guanine exchange factor β-PIX in developing hippocampal neurons. Sci Signal 2021; 14:14/683/eabe4111. [PMID: 34006608 DOI: 10.1126/scisignal.abe4111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Developing hippocampal neurons undergo rapid synaptogenesis in response to neurotrophic signals to form and refine circuit connections. The adipokine leptin is a satiety factor with neurotrophic actions, which potentiates both glutamatergic and GABAergic synaptogenesis in the hippocampus during neonatal development. Brief exposure to leptin enhances GABAA receptor-dependent synaptic currents in hippocampal neurons. Here, using molecular and electrophysiological techniques, we found that leptin increased the surface localization of GABAA receptors and the number of functional GABAergic synapses in hippocampal cultures from male and female rat pups. Leptin increased the interaction between GABAA receptors and the Rho guanine exchange factor β-PIX (a scaffolding protein at GABAergic postsynaptic sites) in a manner dependent on the kinase CaMKK. We also found that the leptin receptor and β-PIX formed a complex, the amount of which transiently increased upon leptin receptor activation. Furthermore, Tyr985 in the leptin receptor and the SH3 domain of β-PIX are crucial for this interaction, which was required for the developmental increase in GABAergic synaptogenesis. Our results suggest a mechanism by which leptin promotes GABAergic synaptogenesis in hippocampal neurons and reveal further complexity in leptin receptor signaling and its interactome.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Jose Luis Rodriguez-Llamas
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Crystal Dillon
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Igor Medina
- Aix-Marseille University UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Suzanne M Appleyard
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Jean-Luc Gaiarsa
- Aix-Marseille University UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Gary A Wayman
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA.
| |
Collapse
|
31
|
Almaguer-Mederos LE, Pérez-Ávila I, Aguilera-Rodríguez R, Velázquez-Garcés M, Almaguer-Gotay D, Hechavarría-Pupo R, Rodríguez-Estupiñán A, Auburger G. Body Mass Index Is Significantly Associated With Disease Severity in Spinocerebellar Ataxia Type 2 Patients. Mov Disord 2021; 36:1372-1380. [PMID: 33548146 DOI: 10.1002/mds.28498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/09/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Spinocerebellar ataxia type 2 is a progressive neurodegenerative disorder due to an unstable expansion of a CAG repeat in the ATXN2 gene. Although weight loss has been associated with disease progression in several neurodegenerative conditions, it has been barely assessed in patients with spinocerebellar ataxia type 2. OBJECTIVE The objective of this study was to test whether body mass index is altered in patients with spinocerebellar ataxia type 2 with varying expansion sizes from early to late disease stages. METHODS A cross-sectional case-control study was performed, which included 222 clinically and molecularly diagnosed patients and 214 sex- and age-matched healthy individuals. ATXN2 genotypes and sex were considered as risk factors. Clinical outcomes included the body mass index, age at onset, disease duration, Scale for the Assessment and Rating of Ataxia score, disease stage, dysphagia, and progression rate. Multiple linear regression models were generated. RESULTS Body mass index was significantly decreased in male patients, but not in female patients, relative to control subjects. In addition to sex, body mass index was significantly associated with age at onset and progression rate. Conversely, body mass index, along with repeat length in ATXN2 expanded alleles and disease duration, was associated with Scale for the Assessment and Rating of Ataxia score. In addition, body mass index, along with the age at onset and the repeat length in ATXN2 normal and expanded alleles, has a significant influence on progression rate. CONCLUSIONS Body mass index might be a useful biomarker of disease severity, particularly in male patients with spinocerebellar ataxia type 2 in the context of nutritional interventions or clinical trials assessing the efficacy of promising new drugs. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Ilbedis Pérez-Ávila
- Center for the Investigation and Rehabilitation of Hereditary Ataxias, Holguín, Cuba.,Center for Sports Medicine, Holguín, Cuba
| | | | | | - Dennis Almaguer-Gotay
- Center for the Investigation and Rehabilitation of Hereditary Ataxias, Holguín, Cuba
| | | | | | - Georg Auburger
- Experimental Neurology, Goethe University Medical Faculty, Frankfurt, Germany
| |
Collapse
|
32
|
Cecon E, Lhomme T, Maurice T, Luka M, Chen M, Silva A, Wauman J, Zabeau L, Tavernier J, Prévot V, Dam J, Jockers R. Amyloid Beta Peptide Is an Endogenous Negative Allosteric Modulator of Leptin Receptor. Neuroendocrinology 2021; 111:370-387. [PMID: 32335558 DOI: 10.1159/000508105] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/23/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Metabolic dysfunction is now recognized as a pivotal component of Alzheimer's disease (AD), the most common dementia worldwide. However, the precise molecular mechanisms linking metabolic dysfunction to AD remain elusive. OBJECTIVE Here, we investigated the direct impact of soluble oligomeric amyloid beta (Aβ) peptides, the main molecular hallmark of AD, on the leptin system, a major component of central energy metabolism regulation. METHODS We developed a new time-resolved fluorescence resonance energy transfer-based Aβ binding assay for the leptin receptor (LepR) and studied the effect of Aβ on LepR function in several in vitro assays. The in vivo effect of Aβ on LepR function was studied in an Aβ-specific AD mouse model and in pro-opiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus. RESULTS We revealed specific and high-affinity (Ki = 0.1 nM) binding of Aβ to LepR. Pharmacological characterization of this interaction showed that Aβ binds allosterically to the extracellular domain of LepR and negatively affects receptor function. Negative allosteric modulation of LepR by Aβ was detected at the level of signaling pathways (STAT-3, AKT, and ERK) in vitroand in vivo. Importantly, the leptin-induced response of POMC neurons, key players in the regulation of metabolic function, was completely abolished in the presence of Aβ. CONCLUSION Our data indicate that Aβ is a negative allosteric modulator of LepR, resulting in impaired leptin action, and qualify LepR as a new and direct target of Aβ oligomers. Preventing the interaction of Aβ with LepR might improve both the metabolic and cognitive dysfunctions in AD condition.
Collapse
Affiliation(s)
- Erika Cecon
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Tori Lhomme
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, EGID, DistAlz, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, UMR_S1198, Montpellier, France
| | - Marine Luka
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Min Chen
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Anisia Silva
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Joris Wauman
- VIB Center for Medical Biotechnology, Department of Biomolecular Medicine, University of Ghent, Ghent, Belgium
| | - Lennart Zabeau
- VIB Center for Medical Biotechnology, Department of Biomolecular Medicine, University of Ghent, Ghent, Belgium
| | - Jan Tavernier
- VIB Center for Medical Biotechnology, Department of Biomolecular Medicine, University of Ghent, Ghent, Belgium
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, EGID, DistAlz, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
| | - Julie Dam
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Ralf Jockers
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France,
| |
Collapse
|
33
|
Sung R, Heo YS. Sandwich ELISA-Based Electrochemical Biosensor for Leptin in Control and Diet-Induced Obesity Mouse Model. BIOSENSORS-BASEL 2020; 11:bios11010007. [PMID: 33374256 PMCID: PMC7823388 DOI: 10.3390/bios11010007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
Leptin is a peptide hormone produced primarily in adipose tissues. Leptin is considered a biomarker associated with obesity and obesity-mediated diseases. Biosensor detection of leptin in the blood may play a critical role as an indicator of dynamic pathological changes. In this paper, we introduce an electrochemical biosensor that adopts o-Phenylenediamine (oPD) on screen-printed gold electrodes (SPGEs) for detecting the leptin from a mouse model of diet-induced obesity (DIO). A linear calibration curve for the leptin concentration was obtained in the ranges from 0.1 to 20 ng/mL with a lower detection limit of 0.033 ng/mL. The leptin concentration was quantified with HRP (horseradish peroxidase)-catalyzed oxidation of oPD by two voltammetry methods: cyclic voltammetry (CV) and square-wave voltammetry (SWV). The proposed sandwich enzyme-linked immunosorbent assay (ELISA)-based electrochemical biosensor for the leptin in mouse blood serum showed high stability, sensitivity, selectivity, and effectivity compared to the commercial Leptin ELISA measurement. Thus, we believe that this leptin biosensor can be a sensitive analytical tool to detect low-levels of biomarkers in clinics and point-of-care testing (POCT).
Collapse
Affiliation(s)
- Ryong Sung
- Obesity-Mediated Disease Research (ODR) Center, School of Medicine, Keimyung University, Daegu 42601, Korea;
| | - Yun Seok Heo
- Department of Biomedical Engineering, School of Medicine, Keimyung University, Daegu 42601, Korea
- Correspondence:
| |
Collapse
|
34
|
Chandramowlishwaran P, Vijay A, Abraham D, Li G, Mwangi SM, Srinivasan S. Role of Sirtuins in Modulating Neurodegeneration of the Enteric Nervous System and Central Nervous System. Front Neurosci 2020; 14:614331. [PMID: 33414704 PMCID: PMC7783311 DOI: 10.3389/fnins.2020.614331] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022] Open
Abstract
Neurodegeneration of the central and enteric nervous systems is a common feature of aging and aging-related diseases, and is accelerated in individuals with metabolic dysfunction including obesity and diabetes. The molecular mechanisms of neurodegeneration in both the CNS and ENS are overlapping. Sirtuins are an important family of histone deacetylases that are important for genome stability, cellular response to stress, and nutrient and hormone sensing. They are activated by calorie restriction (CR) and by the coenzyme, nicotinamide adenine dinucleotide (NAD+). Sirtuins, specifically the nuclear SIRT1 and mitochondrial SIRT3, have been shown to have predominantly neuroprotective roles in the CNS while the cytoplasmic sirtuin, SIRT2 is largely associated with neurodegeneration. A systematic study of sirtuins in the ENS and their effect on enteric neuronal growth and survival has not been conducted. Recent studies, however, also link sirtuins with important hormones such as leptin, ghrelin, melatonin, and serotonin which influence many important processes including satiety, mood, circadian rhythm, and gut homeostasis. In this review, we address emerging roles of sirtuins in modulating the metabolic challenges from aging, obesity, and diabetes that lead to neurodegeneration in the ENS and CNS. We also highlight a novel role for sirtuins along the microbiota-gut-brain axis in modulating neurodegeneration.
Collapse
Affiliation(s)
- Pavithra Chandramowlishwaran
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Anitha Vijay
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Daniel Abraham
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Ge Li
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Simon Musyoka Mwangi
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Research-Gastroenterology, Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| |
Collapse
|
35
|
Meta-analysis of cognitive and behavioral tests in leptin- and leptin receptor-deficient mice. Neurosci Res 2020; 170:217-235. [PMID: 33316303 DOI: 10.1016/j.neures.2020.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 01/19/2023]
Abstract
Leptin is a hormone produced by adipocytes that regulates food intake and metabolism. Leptin-related gene-deficient mice, such as db/db and ob/ob mice, are widely used to study diabetes and its related diseases. However, broad effects of leptin appear to cause variability in behavioral test results. We performed a meta-analysis of major behavioral tests in db/db and ob/ob mice. These mice exhibited significant impairments in the Morris water maze, forced swim, novel object recognition, Y-maze, tail suspension, and light-dark box tests, whereas the elevated plus maze and open field tests did not reveal significant changes. We also performed correlation and regression analyses between the animals' performances and the experimental protocols and conditions. The memory-related tests were characterized by the correlations of their results with animal age, while the performances in the elevated plus-maze and forced swim tests were affected by the width of the devices used. In conclusion, db/db and ob/ob mice mainly exhibit memory deficits and depression-like behavior, although experimenters should be aware of animal age and device size in conducting experiments.
Collapse
|
36
|
Leptin enhances adult neurogenesis and reduces pathological features in a transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2020; 148:105219. [PMID: 33301880 DOI: 10.1016/j.nbd.2020.105219] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/18/2020] [Accepted: 12/03/2020] [Indexed: 01/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common dementia worldwide and is characterized by the presence of senile plaques by amyloid-beta (Aβ) and neurofibrillary tangles of hyperphosphorylated Tau protein. These changes lead to progressive neuronal degeneration and dysfunction, resulting in severe brain atrophy and cognitive deficits. With the discovery that neurogenesis persists in the adult mammalian brain, including brain regions affected by AD, studies of the use of neural stem cells (NSCs) for the treatment of neurodegenerative diseases to repair or prevent neuronal cell loss have increased. Here we demonstrate that leptin administration increases the neurogenic process in the dentate gyrus of the hippocampus as well as in the subventricular zone of lateral ventricles of adult and aged mice. Chronic treatment with leptin increased NSCs proliferation with significant effects on proliferation and differentiation of newborn cells. The expression of the long form of the leptin receptor, LepRb, was detected in the neurogenic niches by reverse qPCR and immunohistochemistry. Moreover, leptin modulated astrogliosis, microglial cell number and the formation of senile plaques. Additionally, leptin led to attenuation of Aβ-induced neurodegeneration and superoxide anion production as revealed by Fluoro-Jade B and dihydroethidium staining. Our study contributes to the understanding of the effects of leptin in the brain that may lead to the development of new therapies to treat Alzheimer's disease.
Collapse
|
37
|
Tian J, Wang T, Wang Q, Guo L, Du H. MK0677, a Ghrelin Mimetic, Improves Neurogenesis but Fails to Prevent Hippocampal Lesions in a Mouse Model of Alzheimer's Disease Pathology. J Alzheimers Dis 2020; 72:467-478. [PMID: 31594237 DOI: 10.3233/jad-190779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hippocampal lesions including synaptic injury, neuroinflammation, and impaired neurogenesis are featured pathology closely associated with neuronal stress and cognitive impairment in Alzheimer's disease (AD). Previous studies suggest that ghrelin and its receptor, growth hormone secretagogue receptor 1α (GHSR1α), promote hippocampal synaptic function and neurogenesis. GHSR1α activation thus holds the potential to be a therapeutic avenue for the treatment of hippocampal pathology in AD; however, a comprehensive study on the preventive effect of MK0677 on hippocampal lesions in AD-related conditions is still lacking. In this study, we treated a transgenic mouse model of AD-like amyloidosis (5xFAD mice) at the asymptomatic stage with MK0677, a potent ghrelin mimetic. We found that MK0677 fostered hippocampal neurogenesis in 5xFAD mice but observed little preventive function with regards to the development of hippocampal amyloid-β (Aβ) deposition, synaptic loss, microglial activation, or cognitive impairment. Furthermore, MK0677 at a dose of 3 mg/kg significantly increased 5xFAD mouse mortality. Despite enhanced hippocampal neurogenesis, MK0677 treatment has little beneficial effect to prevent hippocampal lesions or cognitive deficits against Aβ toxicity. This study, together with a failed large-scale clinical trial, suggests the ineffectiveness of MK0677 alone for AD prevention and treatment.
Collapse
Affiliation(s)
- Jing Tian
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Tienju Wang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Qi Wang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Department of Neurology, Qianfoshan Hospital, Shandong First Medical University, Jinan, China
| | - Lan Guo
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Heng Du
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
38
|
Dumon C, Belaidouni Y, Diabira D, Appleyard SM, Wayman GA, Gaiarsa JL. Leptin down-regulates KCC2 activity and controls chloride homeostasis in the neonatal rat hippocampus. Mol Brain 2020; 13:151. [PMID: 33183317 PMCID: PMC7661183 DOI: 10.1186/s13041-020-00689-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/28/2020] [Indexed: 11/10/2022] Open
Abstract
The canonical physiological role of leptin is to regulate hunger and satiety acting on specific hypothalamic nuclei. Beyond this key metabolic function; leptin also regulates many aspects of development and functioning of neuronal hippocampal networks throughout life. Here we show that leptin controls chloride homeostasis in the developing rat hippocampus in vitro. The effect of leptin relies on the down-regulation of the potassium/chloride extruder KCC2 activity and is present during a restricted period of postnatal development. This study confirms and extends the role of leptin in the ontogenesis of functional GABAergic inhibition and helps understanding how abnormal levels of leptin may contribute to neurological disorders.
Collapse
Affiliation(s)
- Camille Dumon
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France
- Neurochlore Parc Scientifique et Technologique de Luminy, Bâtiment Beret Delaage, Zone Luminy Entreprises Biotech, Marseille, France
| | - Yasmine Belaidouni
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Diabe Diabira
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Suzanne M Appleyard
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Gary A Wayman
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Jean-Luc Gaiarsa
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France.
| |
Collapse
|
39
|
Fadó R, Rodríguez-Rodríguez R, Casals N. The return of malonyl-CoA to the brain: Cognition and other stories. Prog Lipid Res 2020; 81:101071. [PMID: 33186641 DOI: 10.1016/j.plipres.2020.101071] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
Nutrients, hormones and the energy sensor AMP-activated protein kinase (AMPK) tightly regulate the intracellular levels of the metabolic intermediary malonyl-CoA, which is a precursor of fatty acid synthesis and a negative regulator of fatty acid oxidation. In the brain, the involvement of malonyl-CoA in the control of food intake and energy homeostasis has been known for decades. However, recent data uncover a new role in cognition and brain development. The sensing of malonyl-CoA by carnitine palmitoyltransferase 1 (CPT1) proteins regulates a variety of functions, such as the fate of neuronal stem cell precursors, the motility of lysosomes in developing axons, the trafficking of glutamate receptors to the neuron surface (necessary for proper synaptic function) and the metabolic coupling between astrocytes and neurons. We discuss the relevance of those recent findings evidencing how nutrients and metabolic disorders impact cognition. We also enumerate all nutritional and hormonal conditions that are known to regulate malonyl-CoA levels in the brain, reflect on protein malonylation as a new post-translational modification, and give a reasoned vision of the opportunities and challenges that future research in the field could address.
Collapse
Affiliation(s)
- Rut Fadó
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Bellaterra, Cerdanyola del Vallès, Spain.
| | - Rosalía Rodríguez-Rodríguez
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain.
| | - Núria Casals
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, E-08195 Sant Cugat del Vallès, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain.
| |
Collapse
|
40
|
Page KC, Anday EK. Dietary Exposure to Excess Saturated Fat During Early Life Alters Hippocampal Gene Expression and Increases Risk for Behavioral Disorders in Adulthood. Front Neurosci 2020; 14:527258. [PMID: 33013310 PMCID: PMC7516040 DOI: 10.3389/fnins.2020.527258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 08/21/2020] [Indexed: 02/01/2023] Open
Abstract
Purpose Maternal and postnatal diets result in long-term changes in offspring brain and behavior; however, the key mediators of these developmental changes are not well-defined. In this study, we investigated the impact of maternal and post-weaning high-fat diets on gene expression of key components mediating hippocampal synaptic efficacy. In addition, we evaluated the risk for impaired stress-coping and anxiety-like behaviors in adult offspring exposed to obesogenic diets during early life. Methods Dams were fed a control (C) or high-fat (HF) diet prior to mating, pregnancy, and lactation. Male offspring from control chow and high-fat fed dams were weaned to control chow or HF diets. The forced swim test (FST) and the elevated-plus maze (EPM) were used to detect stress-coping and anxiety-like behavior, respectively. Real-time RT-PCR and ELISA were used to analyze hippocampal expression of genes mediating synaptic function. Results Animals fed a HF diet post-weaning spent more time immobile in the FST. Swimming time was reduced in response to both maternal and post-weaning HF diets. Both maternal and post-weaning HF diets contributed to anxiety-like behavior in animals exposed to the EPM. Maternal and post-weaning HF diets were associated with a significant decrease in mRNA and protein expression for hippocampal GDNF, MAP2, SNAP25, and synaptophysin. Hippocampal mRNA expression of key serotonergic and glutamatergic receptors also exhibited differential responses to maternal and post-weaning HF diets. Hippocampal serotonergic receptor 5HT1A mRNA was reduced in response to both the maternal and post-weaning diet, whereas, 5HT2A receptor mRNA expression was increased in response to the maternal HF diet. The glutamate AMPA receptor subunit, GluA1, mRNA expression was significantly reduced in response to both diets, whereas no change was detected in GluA2 subunit mRNA expression. Conclusion These data demonstrate that the expression of genes mediating synaptic function are differentially affected by maternal and post-weaning high-fat diets. The post-weaning high-fat diet clearly disturbs both behavior and gene expression. In addition, although the transition to control diet at weaning partially compensates for the adverse effects of the maternal HF diet, the negative consequence of the maternal HF diet is exacerbated by continuing the high-fat diet post-weaning. We present evidence to support the claim that these dietary influences increase the risk for anxiety and impaired stress-coping abilities in adulthood.
Collapse
Affiliation(s)
- Kathleen C Page
- Department of Biology, Bucknell University, Lewisburg, PA, United States
| | - Endla K Anday
- College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
41
|
Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB, Beal MF, Bergersen LH, Brinton RD, de la Monte S, Eckert A, Harvey J, Jeggo R, Jhamandas JH, Kann O, la Cour CM, Martin WF, Mithieux G, Moreira PI, Murphy MP, Nave KA, Nuriel T, Oliet SHR, Saudou F, Mattson MP, Swerdlow RH, Millan MJ. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov 2020; 19:609-633. [PMID: 32709961 PMCID: PMC7948516 DOI: 10.1038/s41573-020-0072-x] [Citation(s) in RCA: 498] [Impact Index Per Article: 99.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
The brain requires a continuous supply of energy in the form of ATP, most of which is produced from glucose by oxidative phosphorylation in mitochondria, complemented by aerobic glycolysis in the cytoplasm. When glucose levels are limited, ketone bodies generated in the liver and lactate derived from exercising skeletal muscle can also become important energy substrates for the brain. In neurodegenerative disorders of ageing, brain glucose metabolism deteriorates in a progressive, region-specific and disease-specific manner - a problem that is best characterized in Alzheimer disease, where it begins presymptomatically. This Review discusses the status and prospects of therapeutic strategies for countering neurodegenerative disorders of ageing by improving, preserving or rescuing brain energetics. The approaches described include restoring oxidative phosphorylation and glycolysis, increasing insulin sensitivity, correcting mitochondrial dysfunction, ketone-based interventions, acting via hormones that modulate cerebral energetics, RNA therapeutics and complementary multimodal lifestyle changes.
Collapse
Affiliation(s)
- Stephen C Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Research Center on Aging, Sherbrooke, QC, Canada.
| | | | - Cecilie Morland
- Department of Pharmaceutical Biosciences, Institute of Pharmacy, University of Oslo, Oslo, Norway
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University of Dusseldorf, Dusseldorf, Germany
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Zane B Andrews
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| | - M Flint Beal
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Linda H Bergersen
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | | | - Jenni Harvey
- Ninewells Hospital, University of Dundee, Dundee, UK
- Medical School, University of Dundee, Dundee, UK
| | - Ross Jeggo
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France
| | - Jack H Jhamandas
- Department of Medicine, University of Albeta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Albeta, Edmonton, AB, Canada
| | - Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Clothide Mannoury la Cour
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France
| | - William F Martin
- Institute of Molecular Evolution, University of Dusseldorf, Dusseldorf, Germany
| | | | - Paula I Moreira
- CNC Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Klaus-Armin Nave
- Department of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Tal Nuriel
- Columbia University Medical Center, New York, NY, USA
| | - Stéphane H R Oliet
- Neurocentre Magendie, INSERM U1215, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Frédéric Saudou
- University of Grenoble Alpes, Grenoble, France
- INSERM U1216, CHU Grenoble Alpes, Grenoble Institute Neurosciences, Grenoble, France
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, Croissy sur Seine, France.
| |
Collapse
|
42
|
Abstract
The investigation of hormones, brain function and behavior over the past 50 years has played a major role in elucidating how the brain and body communicate reciprocally via hormones and other mediators and how this impacts brain and body health both positively and negatively. This is illustrated here for the hippocampus, a uniquely sensitive and vulnerable brain region, study of which as a hormone target has provided a gateway into the rest of the brain. Hormone actions on the brain and hormones generated within the brain are now recognized to include not only steroid hormones but also metabolic hormones and chemical signals from bone and muscle. Moreover, steroid hormones, and some metabolic hormones, and their receptors, are generated by the brain for specific functions that synergize with effects of those circulating hormones. Hormone actions in hippocampus have revealed its capacity, and that of other brain regions, for adaptive plasticity, loss of which needs external intervention in, for example, mood disorders. Early life experiences as well as in utero and transgenerational effects are now appreciated for their lasting effects at the level of gene expression affecting the capacity for adaptive plasticity. Moreover sex differences are recognized as affecting the whole brain via both genetic and epigenetic mechanisms. The demonstrated plasticity of a healthy brain gives hope that interventions throughout the life course can ameliorate negative effects by reactivating that plasticity and the underlying epigenetic activity to produce compensatory changes in the brain with more positive consequences for the body.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, United States of America.
| |
Collapse
|
43
|
Cárdenas D, Madinabeitia I, Vera J, de Teresa C, Alarcón F, Jiménez R, Catena A. Better brain connectivity is associated with higher total fat mass and lower visceral adipose tissue in military pilots. Sci Rep 2020; 10:610. [PMID: 31953480 PMCID: PMC6969099 DOI: 10.1038/s41598-019-57345-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 12/23/2019] [Indexed: 12/17/2022] Open
Abstract
A lack of exercise leads to being overweight or obese affecting regional brain structure and functional connectivity associated with impaired cognitive function and dementia. In recent decades, several studies of healthy individuals suggest that adiposity may also produce negative independent effects on the brain. We aimed to investigate the relationship between body composition – total fat mass (TFM) and visceral adipose tissue (VAT) – with white matter (WM) integrity using a whole-brain approach in military pilots. Twenty-three military helicopter pilots (Mage = 36.79; SD = 8.00; MBMI = 25.48; SD = 2.49) took part in the study. Brain volumes were studied using diffusion tensor imaging technique by means of a 3T Magnetom Tim Trio. Measurements of body mass index (BMI), TFM and VAT were obtained using Dual-energy X-ray Absorptiometry (DXA). The results showed that, on one hand, higher TFM was associated with higher white matter fractional anisotropy (FA) and, on the other hand, higher VAT was associated with lower FA. Data showed that TFM and VAT are the critical factors underlying WM integrity in combat helicopter pilots. The authors suggest that fat presence enhance brain connectivity while there is no excess, specifically in VAT.
Collapse
Affiliation(s)
- David Cárdenas
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain. .,Sport and Health University Research Institute (iMUDS), Granada, Spain.
| | - Iker Madinabeitia
- Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain.,Sport and Health University Research Institute (iMUDS), Granada, Spain
| | - Jesús Vera
- Sport and Health University Research Institute (iMUDS), Granada, Spain.,Department of Optics, Faculty of Sciences, University of Granada, Granada, Spain
| | - Carlos de Teresa
- Sport and Health University Research Institute (iMUDS), Granada, Spain.,Andalusian Centre of Sports Medicine, Consejería de Turismo y Deporte, Junta de Andalucía, Granada, Spain
| | - Francisco Alarcón
- Department of Didactic General and Specific Training, Faculty of Education, University of Alicante, Alicante, Spain
| | - Raimundo Jiménez
- Sport and Health University Research Institute (iMUDS), Granada, Spain.,Department of Optics, Faculty of Sciences, University of Granada, Granada, Spain
| | - Andrés Catena
- Department of Experimental Psychology, Faculty of Psychology, University of Granada, Granada, Spain.,Mind, Brain and Behaviour Research Centre (CIMCYC), University of Granada, Granada, Spain
| |
Collapse
|
44
|
Stewart AFR, Chen HH. Activation of tyrosine phosphatases in the progression of Alzheimer's disease. Neural Regen Res 2020; 15:2245-2246. [PMID: 32594039 PMCID: PMC7749463 DOI: 10.4103/1673-5374.284986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
45
|
An JH, Jang EH, Kim AY, Fava M, Mischoulon D, Papakostas GI, Na EJ, Jang J, Yu HY, Hong JP, Jeon HJ. Ratio of plasma BDNF to leptin levels are associated with treatment response in major depressive disorder but not in panic disorder: A 12-week follow-up study. J Affect Disord 2019; 259:349-354. [PMID: 31465895 DOI: 10.1016/j.jad.2019.08.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND A link between brain-derived neurotrophic factor (BDNF) expression and the mood regulatory effect of leptin has been suggested in the pathophysiology of major depressive disorder (MDD). We investigated treatment response and pre-treatment leptin and BDNF in patients with MDD and with panic disorder (PD). METHODS We recruited 41 patients with MDD, 52 patients with PD, and 59 matched healthy controls. All subjects completed five visits (at baseline, 2, 4, 8, and 12 weeks), and both MDD and PD patients were treated with standard pharmacotherapy for 12 weeks. Plasma BDNF (pBDNF) and blood leptin levels were obtained along with a 17-item Hamilton Depression Scale rating (HDRS-17) score at every visit. RESULTS The ratio of pre-treatment pBDNF to leptin was significantly lower in patients with MDD and PD compared to healthy controls (p = 0.024), but was not associated with severity of depressive or anxiety symptoms. Pre-treatment pBDNF:leptin ratio was significantly higher in treatment responders than in non-responders (p = 0.012) in MDD but not in PD. This difference was larger in MDD patients with appetite loss (p = 0.034). In multivariate analysis, pre-treatment pBDNF:leptin ratio was significantly associated with treatment responsiveness (Adjusted Odds Ration [AOR] = 2.50, 95% CI 1.02-6.14) in MDD. LIMITATION small sample size; limited information on detailed pharmacological effects. CONCLUSIONS A relatively higher ratio of pre-treatment pBDNF to leptin was associated with greater treatment response in MDD but not in PD. Further research should focus on exploration of a link between BDNF and leptin underlying neuronal plasticity in depression.
Collapse
Affiliation(s)
- Ji Hyun An
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Hye Jang
- Bio-Medical IT Convergence Research Division, Electronics and Telecommunications Research Institute (ETRI), Daejeon, Republic of Korea
| | - Ah Young Kim
- Bio-Medical IT Convergence Research Division, Electronics and Telecommunications Research Institute (ETRI), Daejeon, Republic of Korea
| | - Maurizio Fava
- Depression Clinical and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Mischoulon
- Depression Clinical and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - George I Papakostas
- Depression Clinical and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eun Jin Na
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jihoon Jang
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Han Young Yu
- Bio-Medical IT Convergence Research Division, Electronics and Telecommunications Research Institute (ETRI), Daejeon, Republic of Korea
| | - Jin Pyo Hong
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hong Jin Jeon
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences & Technology, Department of Medical Device Management & Research, and Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
46
|
Kim K, Jang EH, Kim AY, Fava M, Mischoulon D, Papakostas GI, Kim H, Na EJ, Yu HY, Jeon HJ. Pre-treatment peripheral biomarkers associated with treatment response in panic symptoms in patients with major depressive disorder and panic disorder: A 12-week follow-up study. Compr Psychiatry 2019; 95:152140. [PMID: 31669792 DOI: 10.1016/j.comppsych.2019.152140] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/11/2019] [Accepted: 10/15/2019] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Peripheral biomarkers have been studied to predict treatment response of panic symptoms. We hypothesized that depressive disorder (MDD) vs. panic disorder (PD) would exhibit different peripheral biomarkers, and their correlation with severity of panic attacks (PA) would also differ. METHODS Forty-one MDD patients, 52 PD patients, and 59 healthy controls were followed for 12 weeks. We measured peripheral biomarkers along with the Panic Disorder Severity Scale (PDSS) at each visit-pre-treatment, 2, 4, 8, and 12 weeks on a regular schedule. Peripheral biomarkers including serum cytokines, plasma and serum brain-derived neurotrophic factor (BDNF), leptin, adiponectin, and C-reactive protein (CRP) were quantified using enzyme-linked immunosorbent assay (ELISA). RESULTS Patients with MDD and PD demonstrated significantly higher levels of pre-treatment IL-6 compared to controls, but no differences were seen in plasma and serum BDNF, leptin, adiponectin, and CRP. Pre-treatment leptin showed a significant clinical correlation with reduction of panic symptoms in MDD patients at visit 5 (p=0.011), whereas pre-treatment IL-6 showed a negative correlation with panic symptom reduction in PD patients (p=0.022). An improvement in three panic-related items was observed to be positively correlated with pre-treatment leptin in MDD patients: distress during PA, anticipatory anxiety, and occupational interference. CONCLUSION Higher pre-treatment leptin was associated with better response to treatment regarding panic symptoms in patients with MDD, while higher IL-6 was associated with worse response regarding panic symptoms in PD patients. Different predictive peripheral biomarkers observed in MDD and PD suggest the need for establishing individualized predictive biomarkers, even in cases of similar symptoms observed in different disorders.
Collapse
Affiliation(s)
- Kiwon Kim
- Department of Psychiatry, Veteran Health Service Medical Center, Seoul, South Korea; Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Hye Jang
- Bio-Medical IT Convergence Research Division, Electronics and Telecommunications Research Institute (ETRI), Republic of Korea
| | - Ah Young Kim
- Bio-Medical IT Convergence Research Division, Electronics and Telecommunications Research Institute (ETRI), Republic of Korea
| | - Maurizio Fava
- Depression Clinical and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - David Mischoulon
- Depression Clinical and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - George I Papakostas
- Depression Clinical and Research Program, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Hyewon Kim
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun Jin Na
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Han Young Yu
- Bio-Medical IT Convergence Research Division, Electronics and Telecommunications Research Institute (ETRI), Republic of Korea
| | - Hong Jin Jeon
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences & Technology, Department of Medical Device Management & Research, and Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
47
|
Anderson BM, Hirschstein Z, Novakovic ZM, Grasso P. MA-[d-Leu-4]-OB3, a Small Molecule Synthetic Peptide Leptin Mimetic, Mirrors the Cognitive Enhancing Action of Leptin in a Mouse Model of Type 1 Diabetes Mellitus and Alzheimer’s Disease-Like Cognitive Impairment. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09929-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
48
|
Fujita Y, Yamashita T. The Effects of Leptin on Glial Cells in Neurological Diseases. Front Neurosci 2019; 13:828. [PMID: 31447640 PMCID: PMC6692660 DOI: 10.3389/fnins.2019.00828] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022] Open
Abstract
It is known that various endocrine modulators, including leptin and ghrelin, have neuroprotective roles in neurological diseases. Leptin is a hormone produced by adipocytes and was originally identified as a gene related to obesity in mice. The leptin receptors in the hypothalamus are the main target for the homeostatic regulation of body weight. Recent studies have demonstrated that leptin receptors are also expressed in other regions of the central nervous system (CNS), such as the hippocampus, cerebral cortex, and spinal cord. Accordingly, these studies identified the involvement of leptin in the regulation of neuronal survival and neural development. Furthermore, leptin has been shown to have neuroprotective functions in animal models of neurological diseases and demyelination. These observations also suggest that dysregulation of leptin signaling may be involved in the association between neurodegeneration and obesity. In this review, we summarize novel functions of leptin in animal models of neurodegenerative diseases. Specifically, we focus on the emerging evidence for the role of leptin in non-neuronal cells in the CNS, including astrocytes, microglia, and oligodendrocytes. Understanding leptin-mediated neuroprotective signals and molecular mechanisms underlying remyelination will be helpful to establish therapeutic strategies against neurological diseases.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.,Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
49
|
Lloret A, Monllor P, Esteve D, Cervera-Ferri A, Lloret MA. Obesity as a Risk Factor for Alzheimer's Disease: Implication of Leptin and Glutamate. Front Neurosci 2019; 13:508. [PMID: 31191220 PMCID: PMC6540965 DOI: 10.3389/fnins.2019.00508] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/02/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity is known to induce leptin and insulin resistance. Leptin is a peptide hormone synthesized in adipose tissue that mainly regulates food intake. It has been shown that insulin stimulates the production of leptin when adipocytes are exposed to glucose to encourage satiety; while leptin, via a negative feedback, decreases the insulin release and enhances tissue sensitivity to it, leading to glucose uptake for energy utilization or storage. Therefore, resistance to insulin is closely related to leptin resistance. Obesity in middle age has also been related to Alzheimer's disease (AD). In recent years, the relation between impaired leptin signaling pathway and the onset of AD has been studied. In all this context the role of the blood brain barrier (BBB) is crucial. Slow excitotoxicity happens in AD due to an excess of the neurotransmitter glutamate. Since leptin has been shown to regulate N-methyl-D-aspartate (NMDA) receptors, we want to review the link between these pathological pathways, and how they are affected by other AD triggering factors and its role in the onset of AD.
Collapse
Affiliation(s)
- Ana Lloret
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Paloma Monllor
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Daniel Esteve
- Department of Physiology, Faculty of Medicine, University of Valencia, Health Research Institute INCLIVA, Valencia, Spain
| | - Ana Cervera-Ferri
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Maria-Angeles Lloret
- Department of Clinic Neurophysiology, University Clinic Hospital of Valencia, Valencia, Spain
| |
Collapse
|