1
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
2
|
Zhao D, Zhang X, Jin WF, Huang P, Wan H, He Y. Efficacy of Astragalus membranaceus-Carthamus tinctorius in cerebral ischemia/reperfusion injury: Insights from metabolomics and mass spectrometry imaging. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155881. [PMID: 39059267 DOI: 10.1016/j.phymed.2024.155881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/04/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND The combination of Astragalus membranaceus and Carthamus tinctorius (AC) exhibits significant therapeutic effects in cerebral ischemia/reperfusion injury (CIRI). Understanding the metabolic characteristics of brain microregions and disturbances in tissues and systemic circulation is crucial for elucidating the mechanisms of CIRI and the therapeutic benefits of AC. However, in situ metabolic regulation of the complex brain structure has not been adequately studied, and the therapeutic mechanism of AC requires immediate clarification. PURPOSE The present study aimed to unveil the specific metabolic reprogramming of CIRI at systemic and microregional levels, identify key metabolic pathways and metabolites, and elucidate the therapeutic mechanisms of AC. METHODS Air flow-assisted desorption electrospray ionization mass spectrometry imaging (AFADESI-MSI), a newly developed technique, was used to investigate metabolites in brain microregions. Hematoxylin-eosin, Nissl, and immunofluorescence staining were performed to visualize the microscopic changes associated with spatial metabolism. A comprehensive metabolomics study was conducted on serum, brain tissue, and microregions, along with neurological assessments, cerebral infarction measurements, and Evans blue experiments, to assess the systemic and local metabolic effects of AC treatment for CIRI. RESULTS AC significantly reduced neurological damage, minimized infarct size, and repaired blood-brain barrier damage in CIRI rats. AFADESI-MSI demonstrated that the metabolic imbalance caused by CIRI primarily occurs in the cerebral cortex, hippocampus, caudate putamen, thalamus, cerebellar cortex, and fiber tract regions. Significant changes in 16 metabolites were observed in these regions, corresponding to neuron damage, glial cell activation, and neural repair. 20 metabolites from serum and 4 from brain tissue varied significantly with the sham group. Comprehensive metabolomics analysis indicated a close relationship among serum, tissue, and microregional metabolism. CIRI-induced systemic and localized metabolic disorders involve 14 metabolic pathways. AC conferred therapeutic benefits in CIRI by reversing various metabolic imbalances. CONCLUSION AFADESI-MSI efficiently visualized brain microregion metabolism. Comprehensive metabolomics analysis revealed detailed insights into the specific metabolic reprogramming in CIRI and the therapeutic impacts of AC. AC demonstrated significant clinical potential as an adjunct therapy to existing CIRI treatments.
Collapse
Affiliation(s)
- Di Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xian Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei-Feng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
3
|
Naveed M, Smedlund K, Zhou QG, Cai W, Hill JW. Astrocyte involvement in metabolic regulation and disease. Trends Endocrinol Metab 2024:S1043-2760(24)00220-0. [PMID: 39214743 DOI: 10.1016/j.tem.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Astrocytes, the predominant glial cell type in the mammalian brain, influence a wide variety of brain parameters including neuronal energy metabolism. Exciting recent studies have shown that obesity and diabetes can impact on astrocyte function. We review evidence that dysregulation of astrocytic lipid metabolism and glucose sensing contributes to dysregulation of whole-body energy balance, thermoregulation, and insulin sensitivity. In addition, we consider the overlooked topic of the sex-specific roles of astrocytes and their response to hormonal fluctuations that provide insights into sex differences in metabolic regulation. Finally, we provide an update on potential ways to manipulate astrocyte function, including genetic targeting, optogenetic and chemogenetic techniques, transplantation, and tailored exosome-based therapies, which may lead to improved treatments for metabolic disease.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Kathryn Smedlund
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Weikang Cai
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA; Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
4
|
Zhao D, Hu M, Liu S. Glial cells in the mammalian olfactory bulb. Front Cell Neurosci 2024; 18:1426094. [PMID: 39081666 PMCID: PMC11286597 DOI: 10.3389/fncel.2024.1426094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
The mammalian olfactory bulb (OB), an essential part of the olfactory system, plays a critical role in odor detection and neural processing. Historically, research has predominantly focused on the neuronal components of the OB, often overlooking the vital contributions of glial cells. Recent advancements, however, underscore the significant roles that glial cells play within this intricate neural structure. This review discus the diverse functions and dynamics of glial cells in the mammalian OB, mainly focused on astrocytes, microglia, oligodendrocytes, olfactory ensheathing cells, and radial glia cells. Each type of glial contributes uniquely to the OB's functionality, influencing everything from synaptic modulation and neuronal survival to immune defense and axonal guidance. The review features their roles in maintaining neural health, their involvement in neurodegenerative diseases, and their potential in therapeutic applications for neuroregeneration. By providing a comprehensive overview of glial cell types, their mechanisms, and interactions within the OB, this article aims to enhance our understanding of the olfactory system's complexity and the pivotal roles glial cells play in both health and disease.
Collapse
Affiliation(s)
| | | | - Shaolin Liu
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, Department of Biomedical Sciences, University of Georgia College of Veterinary Medicine, Athens, GA, United States
| |
Collapse
|
5
|
Tang H, Li K, Lin L, Wang W, Jian W. Study on the metabolic effects of hexavalent chromium [Cr (VI)] on rat astrocytes using un-targeted metabolomics. Front Mol Biosci 2024; 11:1372783. [PMID: 39035697 PMCID: PMC11257857 DOI: 10.3389/fmolb.2024.1372783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction: Hexavalent chromium [Cr (VI)] has been identified as a human carcinogen and environmental pollutant capable of affecting multiple systems in the human body. However, the specific mechanisms by which Cr (VI) affects the human nervous system remain unclear. Objective: Following confirmation of Cr (VI)'s toxic effects on rat astrocytes, this study explores the metabolites and associated metabolic pathways of rat astrocytes under different doses of Cr (VI) exposure. Methods: Cell viability was assessed using CCK8 assays, intracellular reactive oxygen species (ROS) levels were measured using DCFH-DA fluorescent probes, intracellular 8-hydroxydeoxyguanosine (8-OHdG) content was determined by Elisa, mitochondrial membrane potential was observed using JC-1 probes, and key metabolites were identified through untargeted metabolomics analysis. Results: With increasing Cr (VI) doses, significant decreases in cell viability were observed in the 4, 8, and 16 mg/L dose groups (p < 0.05). Elevated levels of ROS and 8-OHdG, increased caspase-3 activity, and significant reductions in mitochondrial membrane potential were observed in the 2 and 4 mg/L dose groups (p < 0.05). Untargeted metabolomics analysis revealed Cr (VI)'s impact on key metabolites such as sphingosine and methionine. Enrichment analysis of KEGG pathways highlighted the critical roles of sphingolipid metabolism and the methionine-cysteine cycle in the effects of Cr (VI) on rat astrocytes. Conclusion: Our study underscores the potential neuro-health risks associated with environmental and occupational exposure to Cr (VI) and provides new perspectives and directions for investigating neurotoxic mechanisms.
Collapse
Affiliation(s)
- Hongge Tang
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, Fujian, China
| | - Kunyang Li
- Xiamen Haicang Hospital, Xiamen, Fujian, China
| | - Lin Lin
- Scientific Research Management Department, Brain Hospital of Hunan Province, The Second People’s Hospital of Hunan Province, Changsha, Hunan, China
| | - Wenying Wang
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, Fujian, China
| | - Wenjie Jian
- Department of Public Health and Medical Technology, Xiamen Medical College, Xiamen, Fujian, China
| |
Collapse
|
6
|
Roy SC, Sapkota S, Pasula MB, Katakam S, Shrestha R, Briski KP. Glucose transporter-2 regulation of VMN GABA neuron metabolic sensor and transmitter gene expression. Sci Rep 2024; 14:14220. [PMID: 38902332 PMCID: PMC11190205 DOI: 10.1038/s41598-024-64708-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: 03/20/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Glucose transporter-2 (GLUT2) monitors cellular glucose uptake. Astrocyte GLUT2 controls glucose counterregulatory hormone secretion. In vivo gene silencing and laser-catapult-microdissection tools were used here to investigate whether ventromedial hypothalamic nucleus (VMN) GLUT2 may regulate dorsomedial (VMNdm) and/or ventrolateral (VMNvl) γ-aminobutyric acid (GABA) neurotransmission to control this endocrine outflow in female rats. VMN GLUT2 gene knockdown suppressed or stimulated hypoglycemia-associated glutamate decarboxylase (GAD)1 and GAD2 mRNA expression in VMNdm versus VMNvl GABAergic neurons, respectively. GLUT2 siRNA pretreatment also modified co-expressed transmitter marker gene profiles in each cell population. VMNdm GABA neurons exhibited GLUT2 knockdown-sensitive up-regulated 5'-AMP-activated protein kinase-alpha1 (AMPKα1) and -alpha2 (AMPKα2) transcripts during hypoglycemia. Hypoglycemic augmentation of VMNvl GABA neuron AMPKα2 was refractory to GLUT2 siRNA. GLUT2 siRNA blunted (VMNdm) or exacerbated (VMNvl) hypoglycemic stimulation of GABAergic neuron steroidogenic factor-1 (SF-1) mRNA. Results infer that VMNdm and VMNvl GABA neurons may exhibit divergent, GLUT2-dependent GABA neurotransmission patterns in the hypoglycemic female rat. Data also document differential GLUT2 regulation of VMNdm versus VMNvl GABA nerve cell SF-1 gene expression. Evidence for intensification of hypoglycemic hypercorticosteronemia and -glucagonemia by GLUT2 siRNA infers that VMN GLUT2 function imposes an inhibitory tone on these hormone profiles in this sex.
Collapse
Affiliation(s)
- Sagor C Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA
| | - Subash Sapkota
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA
| | - Madhu Babu Pasula
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA
| | - Sushma Katakam
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA
| | - Rami Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Rm 356 Bienville Building, 1800 Bienville Drive, Monroe, LA, 71201, USA.
| |
Collapse
|
7
|
Karnam S, Maurya S, Ng E, Choudhary A, Thobani A, Flanagan JG, Gronert K. Dysregulation of neuroprotective lipoxin pathway in astrocytes in response to cytokines and ocular hypertension. Acta Neuropathol Commun 2024; 12:58. [PMID: 38610040 PMCID: PMC11010376 DOI: 10.1186/s40478-024-01767-2] [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: 09/12/2023] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Glaucoma leads to vision loss due to retinal ganglion cell death. Astrocyte reactivity contributes to neurodegeneration. Our recent study found that lipoxin B4 (LXB4), produced by retinal astrocytes, has direct neuroprotective actions on retinal ganglion cells. In this study, we aimed to investigate how the autacoid LXB4 influences astrocyte reactivity in the retina under inflammatory cytokine-induced activation and during ocular hypertension. The protective activity of LXB4 was investigated in vivo using the mouse silicone-oil model of chronic ocular hypertension. By employing a range of analytical techniques, including bulk RNA-seq, RNAscope in-situ hybridization, qPCR, and lipidomic analyses, we discovered the formation of lipoxins and expression of the lipoxin pathway in rodents (including the retina and optic nerve), primates (optic nerve), and human brain astrocytes, indicating the presence of this neuroprotective pathway across various species. Findings in the mouse retina identified significant dysregulation of the lipoxin pathway in response to chronic ocular hypertension, leading to an increase in 5-lipoxygenase (5-LOX) activity and a decrease in 15-LOX activity. This dysregulation was coincident with a marked upregulation of astrocyte reactivity. Reactive human brain astrocytes also showed a significant increase in 5-LOX. Treatment with LXB4 amplified the lipoxin biosynthetic pathway by restoring and amplifying the generation of another member of the lipoxin family, LXA4, and mitigated astrocyte reactivity in mouse retinas and human brain astrocytes. In conclusion, the lipoxin pathway is functionally expressed in rodents, primates, and human astrocytes, and is a resident neuroprotective pathway that is downregulated in reactive astrocytes. Novel cellular targets for LXB4's neuroprotective action are inhibition of astrocyte reactivity and restoration of lipoxin generation. Amplifying the lipoxin pathway is a potential target to disrupt or prevent astrocyte reactivity in neurodegenerative diseases, including retinal ganglion cell death in glaucoma.
Collapse
Affiliation(s)
- Shruthi Karnam
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Shubham Maurya
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Elainna Ng
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Amodini Choudhary
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Arzin Thobani
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - John G Flanagan
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA.
| | - Karsten Gronert
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA.
- Infectious Disease and Immunity Program, Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
8
|
Mitroshina E, Kalinina E, Vedunova M. Optogenetics in Alzheimer's Disease: Focus on Astrocytes. Antioxidants (Basel) 2023; 12:1856. [PMID: 37891935 PMCID: PMC10604138 DOI: 10.3390/antiox12101856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, resulting in disability and mortality. The global incidence of AD is consistently surging. Although numerous therapeutic agents with promising potential have been developed, none have successfully treated AD to date. Consequently, the pursuit of novel methodologies to address neurodegenerative processes in AD remains a paramount endeavor. A particularly promising avenue in this search is optogenetics, enabling the manipulation of neuronal activity. In recent years, research attention has pivoted from neurons to glial cells. This review aims to consider the potential of the optogenetic correction of astrocyte metabolism as a promising strategy for correcting AD-related disorders. The initial segment of the review centers on the role of astrocytes in the genesis of neurodegeneration. Astrocytes have been implicated in several pathological processes associated with AD, encompassing the clearance of β-amyloid, neuroinflammation, excitotoxicity, oxidative stress, and lipid metabolism (along with a critical role in apolipoprotein E function). The effect of astrocyte-neuronal interactions will also be scrutinized. Furthermore, the review delves into a number of studies indicating that changes in cellular calcium (Ca2+) signaling are one of the causes of neurodegeneration. The review's latter section presents insights into the application of various optogenetic tools to manipulate astrocytic function as a means to counteract neurodegenerative changes.
Collapse
Affiliation(s)
- Elena Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia (M.V.)
| | | | | |
Collapse
|
9
|
Li AJ, Wang Q, Rogers RC, Herman G, Ritter RC, Ritter S. Chemogenetic activation of ventral medullary astrocytes enhances feeding and corticosterone release in response to mild glucoprivation. Am J Physiol Regul Integr Comp Physiol 2023; 325:R229-R237. [PMID: 37424401 PMCID: PMC10396275 DOI: 10.1152/ajpregu.00079.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
To investigate the role of glial cells in the regulation of glucoprivic responses in rats, a chemogenetic approach was used to activate astrocytes neighboring catecholamine (CA) neurons in the ventromedial medulla (VLM) where A1 and C1 CA cell groups overlap (A1/C1). Previous results indicate that activation of CA neurons in this region is necessary and sufficient for feeding and corticosterone release in response to glucoprivation. However, it is not known whether astrocyte neighbors of CA neurons contribute to glucoregulatory responses. Hence, we made nanoinjections of AAV5-GFAP-hM3D(Gq)-mCherry to selectively transfect astrocytes in the A1/C1 region with the excitatory designer receptor exclusively activated by designer drugs (DREADDs), hM3D(Gq). After allowing time for DREADD expression, we evaluated the rats for increased food intake and corticosterone release in response to low systemic doses of the antiglycolytic agent, 2-deoxy-d-glucose (2DG), alone and in combination with the hM3D(Gq) activator clozapine-n-oxide (CNO). We found that DREADD-transfected rats ate significantly more food when 2DG and CNO were coadministered than when either 2DG or CNO was injected alone. We also found that CNO significantly enhanced 2DG-induced FOS expression in the A1/C1 CA neurons, and that corticosterone release also was enhanced when CNO and 2DG were administered together. Importantly, CNO-induced activation of astrocytes in the absence of 2DG did not trigger food intake or corticosterone release. Our results indicate that during glucoprivation, activation of VLM astrocytes cells markedly increases the sensitivity or responsiveness of neighboring A1/C1 CA neurons to glucose deficit, suggesting a potentially important role for VLM astrocytes in glucoregulation.
Collapse
Affiliation(s)
- Ai-Jun Li
- Programs in Neuroscience, Washington State University, Pullman, Washington, United States
| | - Qing Wang
- Programs in Neuroscience, Washington State University, Pullman, Washington, United States
| | - Richard C Rogers
- Autonomic Neuroscience Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
| | - Gerlinda Herman
- Autonomic Neuroscience Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
| | - Robert C Ritter
- Programs in Neuroscience, Washington State University, Pullman, Washington, United States
| | - Sue Ritter
- Programs in Neuroscience, Washington State University, Pullman, Washington, United States
| |
Collapse
|
10
|
Astroglial CB1 receptors, energy metabolism, and gliotransmission: an integrated signaling system? Essays Biochem 2023; 67:49-61. [PMID: 36645029 DOI: 10.1042/ebc20220089] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/17/2023]
Abstract
Astrocytes are key players in brain homeostasis and function. During the last years, several studies have cemented this notion by showing that these cells respond to neuronal signals and, via the release of molecules that modulate and support synaptic activity (gliotransmission) participates in the functions of the so-called tripartite synapse. Thus, besides their established control of brain metabolism, astrocytes can also actively control synaptic activity and behavior. Among the signaling pathways that shape the functions of astrocyte, the cannabinoid type-1 (CB1) receptor is emerging as a critical player in the control of both gliotransmission and the metabolic cooperation between astrocytes and neurons. In the present short review, we describe known and newly discovered properties of the astroglial CB1 receptors and their role in modulating brain function and behavior. Based on this evidence, we finally discuss how the functions and mode of actions of astrocyte CB1 receptors might represent a clear example of the inextricable relationship between energy metabolism and gliotransmission. These tight interactions will need to be taken into account for future research in astrocyte functions and call for a reinforcement of the theoretical and experimental bridges between studies on metabolic and synaptic functions of astrocytes.
Collapse
|
11
|
Pasula MB, Napit PR, Alhamyani A, Roy SC, Sylvester PW, Bheemanapally K, Briski KP. Sex Dimorphic Glucose Transporter-2 Regulation of Hypothalamic Astrocyte Glucose and Energy Sensor Expression and Glycogen Metabolism. Neurochem Res 2023; 48:404-417. [PMID: 36173588 PMCID: PMC9898103 DOI: 10.1007/s11064-022-03757-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/17/2022] [Accepted: 09/06/2022] [Indexed: 02/06/2023]
Abstract
The plasma membrane glucose transporter-2 (GLUT2) monitors brain cell uptake of the critical nutrient glucose, and functions within astrocytes of as-yet-unknown location to control glucose counter-regulation. Hypothalamic astrocyte-neuron metabolic coupling provides vital cues to the neural glucostatic network. Current research utilized an established hypothalamic primary astrocyte culture model along with gene knockdown tools to investigate whether GLUT2 imposes sex-specific regulation of glucose/energy sensor function and glycogen metabolism in this cell population. Data show that GLUT2 stimulates or inhibits glucokinase (GCK) expression in glucose-supplied versus -deprived male astrocytes, but does not control this protein in female. Astrocyte 5'-AMP-activated protein kinaseα1/2 (AMPK) protein is augmented by GLUT2 in each sex, but phosphoAMPKα1/2 is coincidently up- (male) or down- (female) regulated. GLUT2 effects on glycogen synthase (GS) diverges in the two sexes, but direction of this control is reversed by glucoprivation in each sex. GLUT2 increases (male) or decreases (female) glycogen phosphorylase-brain type (GPbb) protein during glucoprivation, yet simultaneously inhibits (male) or stimulates (female) GP-muscle type (GPmm) expression. Astrocyte glycogen accumulation is restrained by GLUT2 when glucose is present (male) or absent (both sexes). Outcomes disclose sex-dependent GLUT2 control of the astrocyte glycolytic pathway sensor GCK. Data show that glucose status determines GLUT2 regulation of GS (both sexes), GPbb (female), and GPmm (male), and that GLUT2 imposes opposite control of GS, GPbb, and GPmm profiles between sexes during glucoprivation. Ongoing studies aim to investigate molecular mechanisms underlying sex-dimorphic GLUT2 regulation of hypothalamic astrocyte metabolic-sensory and glycogen metabolic proteins, and to characterize effects of sex-specific astrocyte target protein responses to GLUT2 on glucose regulation.
Collapse
Affiliation(s)
- Madhu Babu Pasula
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building 1800 Bienville Drive, 71201, Monroe, LA, USA
| | - Prabhat R Napit
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building 1800 Bienville Drive, 71201, Monroe, LA, USA
| | - Abdulrahman Alhamyani
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building 1800 Bienville Drive, 71201, Monroe, LA, USA
| | - Sagor C Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building 1800 Bienville Drive, 71201, Monroe, LA, USA
| | - Paul W Sylvester
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building 1800 Bienville Drive, 71201, Monroe, LA, USA
| | - Khaggeswar Bheemanapally
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building 1800 Bienville Drive, 71201, Monroe, LA, USA
| | - Karen P Briski
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Rm 356 Bienville Building 1800 Bienville Drive, 71201, Monroe, LA, USA.
| |
Collapse
|
12
|
Rodríguez-Giraldo M, González-Reyes RE, Ramírez-Guerrero S, Bonilla-Trilleras CE, Guardo-Maya S, Nava-Mesa MO. Astrocytes as a Therapeutic Target in Alzheimer's Disease-Comprehensive Review and Recent Developments. Int J Mol Sci 2022; 23:13630. [PMID: 36362415 PMCID: PMC9654484 DOI: 10.3390/ijms232113630] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 09/20/2023] Open
Abstract
Alzheimer's disease (AD) is a frequent and disabling neurodegenerative disorder, in which astrocytes participate in several pathophysiological processes including neuroinflammation, excitotoxicity, oxidative stress and lipid metabolism (along with a critical role in apolipoprotein E function). Current evidence shows that astrocytes have both neuroprotective and neurotoxic effects depending on the disease stage and microenvironmental factors. Furthermore, astrocytes appear to be affected by the presence of amyloid-beta (Aβ), with alterations in calcium levels, gliotransmission and proinflammatory activity via RAGE-NF-κB pathway. In addition, astrocytes play an important role in the metabolism of tau and clearance of Aβ through the glymphatic system. In this review, we will discuss novel pharmacological and non-pharmacological treatments focused on astrocytes as therapeutic targets for AD. These interventions include effects on anti-inflammatory/antioxidant systems, glutamate activity, lipid metabolism, neurovascular coupling and glymphatic system, calcium dysregulation, and in the release of peptides which affects glial and neuronal function. According to the AD stage, these therapies may be of benefit in either preventing or delaying the progression of the disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauricio O. Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111711, Colombia
| |
Collapse
|
13
|
Sovrani V, Bobermin LD, Santos CL, Brondani M, Gonçalves CA, Leipnitz G, Quincozes-Santos A. Effects of long-term resveratrol treatment in hypothalamic astrocyte cultures from aged rats. Mol Cell Biochem 2022; 478:1205-1216. [PMID: 36272012 DOI: 10.1007/s11010-022-04585-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
Abstract
Aging is intrinsically related to metabolic changes and characterized by the accumulation of oxidative and inflammatory damage, as well as alterations in gene expression and activity of several signaling pathways, which in turn impact on homeostatic responses of the body. Hypothalamus is a brain region most related to these responses, and increasing evidence has highlighted a critical role of astrocytes in hypothalamic homeostatic functions, particularly during aging process. The purpose of this study was to investigate the in vitro effects of a chronic treatment with resveratrol (1 µM during 15 days, which was replaced once every 3 days), a recognized anti-inflammatory and antioxidant molecule, in primary hypothalamic astrocyte cultures obtained from aged rats (24 months old). We observed that aging process changes metabolic, oxidative, inflammatory, and senescence parameters, as well as glial markers, while long-term resveratrol treatment prevented these effects. In addition, resveratrol upregulated key signaling pathways associated with cellular homeostasis, including adenosine receptors, nuclear factor erythroid-derived 2-like 2 (Nrf2), heme oxygenase 1 (HO-1), sirtuin 1 (SIRT1), proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and phosphoinositide 3-kinase (PI3K). Our data corroborate the glioprotective effect of resveratrol in aged hypothalamic astrocytes, reinforcing the beneficial role of resveratrol in the aging process.
Collapse
Affiliation(s)
- Vanessa Sovrani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Camila Leite Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Morgana Brondani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos-Alberto Gonçalves
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 - Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 - Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil
| | - André Quincozes-Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 - Anexo, Bairro Santa Cecília, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
14
|
De Backer JF, Grunwald Kadow IC. A role for glia in cellular and systemic metabolism: insights from the fly. CURRENT OPINION IN INSECT SCIENCE 2022; 53:100947. [PMID: 35772690 DOI: 10.1016/j.cois.2022.100947] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
Excitability and synaptic transmission make neurons high-energy consumers. However, neurons do not store carbohydrates or lipids. Instead, they need support cells to fuel their metabolic demands. This role is assumed by glia, both in vertebrates and invertebrates. Many questions remain regarding the coupling between neuronal activity and energy demand on the one hand, and nutrient supply by glia on the other hand. Here, we review recent advances showing that fly glia, similar to their role in vertebrates, fuel neurons in times of high energetic demand, such as during memory formation and long-term storage. Vertebrate glia also play a role in the modulation of neurons, their communication, and behavior, including food search and feeding. We discuss recent literature pointing to similar roles of fly glia in behavior and metabolism.
Collapse
Affiliation(s)
- Jean-François De Backer
- Technical University of Munich, School of Life Sciences, Liesel-Beckmann-Str. 4, 85354 Freising, Germany; University of Bonn, Faculty of Medicine, UKB, Institute of Physiology II, Nussallee 11, 53115 Bonn, Germany
| | - Ilona C Grunwald Kadow
- Technical University of Munich, School of Life Sciences, Liesel-Beckmann-Str. 4, 85354 Freising, Germany; University of Bonn, Faculty of Medicine, UKB, Institute of Physiology II, Nussallee 11, 53115 Bonn, Germany.
| |
Collapse
|
15
|
Demais V, Pohl A, Wunderlich KA, Pfaller AM, Kaplan L, Barthélémy A, Dittrich R, Puig B, Giebel B, Hauck SM, Pfrieger FW, Grosche A. Release of VAMP5-positive extracellular vesicles by retinal Müller glia in vivo. J Extracell Vesicles 2022; 11:e12254. [PMID: 36043482 PMCID: PMC9428896 DOI: 10.1002/jev2.12254] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 11/11/2022] Open
Abstract
Cell-cell interactions in the central nervous system are based on the release of molecules mediating signal exchange and providing structural and trophic support through vesicular exocytosis and the formation of extracellular vesicles. The specific mechanisms employed by each cell type in the brain are incompletely understood. Here, we explored the means of communication used by Müller cells, a type of radial glial cells in the retina, which forms part of the central nervous system. Using immunohistochemical, electron microscopic, and molecular analyses, we provide evidence for the release of distinct extracellular vesicles from endfeet and microvilli of retinal Müller cells in adult mice in vivo. We identify VAMP5 as a Müller cell-specific SNARE component that is part of extracellular vesicles and responsive to ischemia, and we reveal differences between the secretomes of immunoaffinity-purified Müller cells and neurons in vitro. Our findings suggest extracellular vesicle-based communication as an important mediator of cellular interactions in the retina.
Collapse
Affiliation(s)
- Valerie Demais
- Plateforme Imagerie In Vitro, CNRS UAR 3156, NeuropôleUniversity of StrasbourgStrasbourgFrance
| | - Anne Pohl
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
- Institute of Human GeneticsUniversity of RegensburgRegensburgGermany
| | - Kirsten A. Wunderlich
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Anna M. Pfaller
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Lew Kaplan
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Amelie Barthélémy
- Centre National de la Recherche ScientifiqueUniversité de StrasbourgInstitut des Neurosciences Cellulaires et IntégrativesStrasbourgFrance
| | - Robin Dittrich
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Berta Puig
- Neurology DepartmentExperimental Research in Stroke and Inflammation (ERSI)University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core and Research Unit Protein ScienceHelmholtz‐Zentrum MünchenMünchenGermany
| | - Frank W. Pfrieger
- Plateforme Imagerie In Vitro, CNRS UAR 3156, NeuropôleUniversity of StrasbourgStrasbourgFrance
- Centre National de la Recherche ScientifiqueUniversité de StrasbourgInstitut des Neurosciences Cellulaires et IntégrativesStrasbourgFrance
| | - Antje Grosche
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| |
Collapse
|
16
|
Woo A, Botta A, Shi SSW, Paus T, Pausova Z. Obesity-Related Neuroinflammation: Magnetic Resonance and Microscopy Imaging of the Brain. Int J Mol Sci 2022; 23:8790. [PMID: 35955925 PMCID: PMC9368789 DOI: 10.3390/ijms23158790] [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: 07/17/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 12/01/2022] Open
Abstract
Obesity is a major risk factor of Alzheimer's disease and related dementias. The principal feature of dementia is a loss of neurons and brain atrophy. The mechanistic links between obesity and the neurodegenerative processes of dementias are not fully understood, but recent research suggests that obesity-related systemic inflammation and subsequent neuroinflammation may be involved. Adipose tissues release multiple proinflammatory molecules (fatty acids and cytokines) that impact blood and vessel cells, inducing low-grade systemic inflammation that can transition to tissues, including the brain. Inflammation in the brain-neuroinflammation-is one of key elements of the pathobiology of neurodegenerative disorders; it is characterized by the activation of microglia, the resident immune cells in the brain, and by the structural and functional changes of other cells forming the brain parenchyma, including neurons. Such cellular changes have been shown in animal models with direct methods, such as confocal microscopy. In humans, cellular changes are less tangible, as only indirect methods such as magnetic resonance (MR) imaging are usually used. In these studies, obesity and low-grade systemic inflammation have been associated with lower volumes of the cerebral gray matter, cortex, and hippocampus, as well as altered tissue MR properties (suggesting microstructural variations in cellular and molecular composition). How these structural variations in the human brain observed using MR imaging relate to the cellular variations in the animal brain seen with microscopy is not well understood. This review describes the current understanding of neuroinflammation in the context of obesity-induced systemic inflammation, and it highlights need for the bridge between animal microscopy and human MR imaging studies.
Collapse
Affiliation(s)
- Anita Woo
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Amy Botta
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Sammy S. W. Shi
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Tomas Paus
- Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, QC H3T 1C5, Canada
- Departments of Psychiatry of Neuroscience, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada
- ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A1, Canada
- ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada
| |
Collapse
|
17
|
He Z, Xu Y, Ma Q, Zhou C, Yang L, Lin M, Deng P, Yang Z, Gong M, Zhang H, Lu M, Li Y, Gao P, Lu Y, He M, Zhang L, Pi H, Zhang K, Qin S, Yu Z, Zhou Z, Chen C. SOX2 modulated astrocytic process plasticity is involved in arsenic-induced metabolic disorders. JOURNAL OF HAZARDOUS MATERIALS 2022; 435:128942. [PMID: 35468398 DOI: 10.1016/j.jhazmat.2022.128942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/02/2022] [Accepted: 04/13/2022] [Indexed: 06/14/2023]
Abstract
Metabolic disorders induced by arsenic exposure have attracted great public concern. However, it remains unclear whether hypothalamus-based central regulation mechanisms are involved in this process. Here, we exposed mice to 100 μg/L arsenic in drinking water and established a chronic arsenic exposure model. Our study revealed that chronic arsenic exposure caused metabolic disorders in mice including impaired glucose metabolism and decreased energy expenditure. Arsenic exposure also impaired glucose sensing and the activation of proopiomelanocortin (POMC) neurons in the hypothalamus. In particular, arsenic exposure damaged the plasticity of hypothalamic astrocytic process. Further research revealed that arsenic exposure inhibited the expression of sex-determining region Y-Box 2 (SOX2), which decreased the expression level of insulin receptors (INSRs) and the phosphorylation of AKT. The conditional deletion of astrocytic SOX2 exacerbated arsenic-induced effects on metabolic disorders, the impairment of hypothalamic astrocytic processes, and the inhibition of INSR/AKT signaling. Furthermore, the arsenic-induced impairment of astrocytic processes and inhibitory effects on INSR/AKT signaling were reversed by SOX2 overexpression in primary hypothalamic astrocytes. Together, we demonstrated here that chronic arsenic exposure caused metabolic disorders by impairing SOX2-modulated hypothalamic astrocytic process plasticity in mice. Our study provides evidence of novel central regulatory mechanisms underlying arsenic-induced metabolic disorders and emphasizes the crucial role of SOX2 in regulating the process plasticity of adult astrocytes.
Collapse
Affiliation(s)
- Zhixin He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Yudong Xu
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Chao Zhou
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China; Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University, Shigatse 857099, China
| | - Lingling Yang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Min Lin
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhiqi Yang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Mingyue Gong
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Huijie Zhang
- School of Medicine, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Muxue Lu
- School of Medicine, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Yanqi Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Yonghui Lu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Mindi He
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Kuan Zhang
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China
| | - Song Qin
- Department of Anatomy, Histology and Embryology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China; Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing 400030, China.
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
18
|
Gruber T, García-Cáceres C. Astroglial clean-up of satiety synapses. Nat Metab 2022; 4:505-506. [PMID: 35501600 DOI: 10.1038/s42255-022-00563-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München & German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München & German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
19
|
de Almeida Miranda D, Araripe J, de Morais Magalhães NG, de Siqueira LS, de Abreu CC, Pereira PDC, Henrique EP, da Silva Chira PAC, de Melo MAD, do Rêgo PS, Diniz DG, Sherry DF, Diniz CWP, Guerreiro-Diniz C. Shorebirds' Longer Migratory Distances Are Associated With Larger ADCYAP1 Microsatellites and Greater Morphological Complexity of Hippocampal Astrocytes. Front Psychol 2022; 12:784372. [PMID: 35185684 PMCID: PMC8855117 DOI: 10.3389/fpsyg.2021.784372] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
For the epic journey of autumn migration, long-distance migratory birds use innate and learned information and follow strict schedules imposed by genetic and epigenetic mechanisms, the details of which remain largely unknown. In addition, bird migration requires integrated action of different multisensory systems for learning and memory, and the hippocampus appears to be the integration center for this task. In previous studies we found that contrasting long-distance migratory flights differentially affected the morphological complexity of two types of hippocampus astrocytes. Recently, a significant association was found between the latitude of the reproductive site and the size of the ADCYAP1 allele in long distance migratory birds. We tested for correlations between astrocyte morphological complexity, migratory distances, and size of the ADCYAP1 allele in three long-distance migrant species of shorebird and one non-migrant. Significant differences among species were found in the number and morphological complexity of the astrocytes, as well as in the size of the microsatellites of the ADCYAP1 gene. We found significant associations between the size of the ADCYAP1 microsatellites, the migratory distances, and the degree of morphological complexity of the astrocytes. We suggest that associations between astrocyte number and morphological complexity, ADCYAP1 microsatellite size, and migratory behavior may be part of the adaptive response to the migratory process of shorebirds.
Collapse
Affiliation(s)
- Diego de Almeida Miranda
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil.,Laboratório de Genética e Conservação, Instituto de Estudos Costeiros (IECOS), Universidade Federal do Pará, Bragança, Brazil
| | - Juliana Araripe
- Laboratório de Genética e Conservação, Instituto de Estudos Costeiros (IECOS), Universidade Federal do Pará, Bragança, Brazil
| | - Nara G de Morais Magalhães
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Lucas Silva de Siqueira
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Cintya Castro de Abreu
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Patrick Douglas Corrêa Pereira
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Ediely Pereira Henrique
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Pedro Arthur Campos da Silva Chira
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Mauro A D de Melo
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| | - Péricles Sena do Rêgo
- Laboratório de Genética e Conservação, Instituto de Estudos Costeiros (IECOS), Universidade Federal do Pará, Bragança, Brazil
| | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Belém, Brazil.,Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Brazil
| | - David Francis Sherry
- Department of Psychology, Advanced Facility for Avian Research, University of Western Ontario, London, ON, Canada
| | - Cristovam W P Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Universidade Federal do Pará, Hospital Universitário João de Barros Barreto, Belém, Brazil
| | - Cristovam Guerreiro-Diniz
- Instituto Federal de Educação Ciência e Tecnologia do Pará, Campus Bragança, Laboratório de Biologia Molecular e Neuroecologia, Bragança, Brazil
| |
Collapse
|