1
|
Prasad SK, Acharjee A, Singh VV, Trigun SK, Acharjee P. Modulation of brain energy metabolism in hepatic encephalopathy: impact of glucose metabolic dysfunction. Metab Brain Dis 2024; 39:1649-1665. [PMID: 39120853 DOI: 10.1007/s11011-024-01407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Cerebral function is linked to a high level of metabolic activity and relies on glucose as its primary energy source. Glucose aids in the maintenance of physiological brain activities; as a result, a disruption in metabolism has a significant impact on brain function, launching a chain of events that leads to neuronal death. This metabolic insufficiency has been observed in a variety of brain diseases and neuroexcitotoxicity disorders, including hepatic encephalopathy. It is a significant neurological complication that develops in people with liver disease, ranging from asymptomatic abnormalities to coma. Hyperammonemia is the main neurotoxic villain in the development of hepatic encephalopathy and induces a wide range of complications in the brain. The neurotoxic effects of ammonia on brain function are thought to be mediated by impaired glucose metabolism. Accordingly, in this review, we provide an understanding of deranged brain energy metabolism, emphasizing the role of glucose metabolic dysfunction in the pathogenesis of hepatic encephalopathy. We also highlighted the differential metabolic profiles of brain cells and the status of metabolic cooperation between them. The major metabolic pathways that have been explored are glycolysis, glycogen metabolism, lactate metabolism, the pentose phosphate pathway, and the Krebs cycle. Furthermore, the lack of efficacy in current hepatic encephalopathy treatment methods highlights the need to investigate potential therapeutic targets for hepatic encephalopathy, with regulating deficient bioenergetics being a viable alternative in this case. This review also demonstrates the importance of the development of glucose metabolism-focused disease diagnostics and treatments, which are now being pursued for many ailments.
Collapse
Affiliation(s)
- Shambhu Kumar Prasad
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Arup Acharjee
- Department of Zoology, University of Allahabad, Prayagraj, 211002, India.
| | - Vishal Vikram Singh
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Surendra Kumar Trigun
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Papia Acharjee
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
2
|
Chang CP, Wu CW, Chern Y. Metabolic dysregulation in Huntington's disease: Neuronal and glial perspectives. Neurobiol Dis 2024; 201:106672. [PMID: 39306013 DOI: 10.1016/j.nbd.2024.106672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a mutant huntingtin protein with an abnormal CAG/polyQ expansion in the N-terminus of HTT exon 1. HD is characterized by progressive neurodegeneration and metabolic abnormalities, particularly in the brain, which accounts for approximately 20 % of the body's resting metabolic rate. Dysregulation of energy homeostasis in HD includes impaired glucose transporters, abnormal functions of glycolytic enzymes, changes in tricarboxylic acid (TCA) cycle activity and enzyme expression in the basal ganglia and cortical regions of both HD mouse models and HD patients. However, current understanding of brain cell behavior during energy dysregulation and its impact on neuron-glia crosstalk in HD remains limited. This review provides a comprehensive summary of the current understanding of the differences in glucose metabolism between neurons and glial cells in HD and how these differences contribute to disease development compared with normal conditions. We also discuss the potential impact of metabolic shifts on neuron-glia communication in HD. A deeper understanding of these metabolic alterations may reveal potential therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Wen Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
3
|
Fernandes MGF, Pernin F, Antel JP, Kennedy TE. From BBB to PPP: Bioenergetic requirements and challenges for oligodendrocytes in health and disease. J Neurochem 2024. [PMID: 39253904 DOI: 10.1111/jnc.16219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/24/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Abstract
Mature myelinating oligodendrocytes, the cells that produce the myelin sheath that insulates axons in the central nervous system, have distinct energetic and metabolic requirements compared to neurons. Neurons require substantial energy to execute action potentials, while the energy needs of oligodendrocytes are directed toward building the lipid-rich components of myelin and supporting neuronal metabolism by transferring glycolytic products to axons as additional fuel. The utilization of energy metabolites in the brain parenchyma is tightly regulated to meet the needs of different cell types. Disruption of the supply of metabolites can lead to stress and oligodendrocyte injury, contributing to various neurological disorders, including some demyelinating diseases. Understanding the physiological properties, structures, and mechanisms involved in oligodendrocyte energy metabolism, as well as the relationship between oligodendrocytes and neighboring cells, is crucial to investigate the underlying pathophysiology caused by metabolic impairment in these disorders. In this review, we describe the particular physiological properties of oligodendrocyte energy metabolism and the response of oligodendrocytes to metabolic stress. We delineate the relationship between oligodendrocytes and other cells in the context of the neurovascular unit, and the regulation of metabolite supply according to energetic needs. We focus on the specific bioenergetic requirements of oligodendrocytes and address the disruption of metabolic energy in demyelinating diseases. We encourage further studies to increase understanding of the significance of metabolic stress on oligodendrocyte injury, to support the development of novel therapeutic approaches for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Milton Guilherme Forestieri Fernandes
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Florian Pernin
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jack P Antel
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Timothy E Kennedy
- Neuroimmunological Diseases and Glia Biology Research Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Marangoni D, Placidi G, D'Agostino E, De Siena E, Attinà G, Mastrangelo S, Ruggiero A, Colosimo C, Falsini B. Longitudinal changes in retinal ganglion cell function in optic pathway glioma evaluated by photopic negative response. Exp Eye Res 2024; 246:110012. [PMID: 39059735 DOI: 10.1016/j.exer.2024.110012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Photopic negative response (PhNR), an index of retinal ganglion cell (RGC) function, is impaired in patients with optic pathway gliomas (OPGs). The aim of this longitudinal study was to evaluate whether PhNR deteriorates over time in OPG patients. Fourteen pediatric patients affected by OPG (4 males and 10 females, mean age 12.4 ± 5.7 years, 8 with neurofibromatosis type 1 [NF1]) with ≥12 months of follow-up and ≥2 evaluations, were included in this retrospective study. All patients had received chemotherapy, with or without OPG surgical resection, at least 5 years prior to the study. At baseline, all patients underwent a complete ophthalmological examination. Follow-up included clinical examination and PhNR measurement as well as brain MRI (according to pediatric oncologist indications) every 6 or 12 months. Mean follow-up duration was 16.7 ± 7.5 months (range 12-36 months). Photopic electroretinograms were elicited by 2.0 cd-s/m2 Ganzfeld white flashes presented on a steady 20 cd/m2 white background. The PhNR amplitude was measured as the difference between baseline and the maximal negative amplitude (minimum) of the negative wave, following the photopic b-wave. Compared to baseline, mean PhNR amplitude was significantly decreased at the end of follow-up (p = 0.008). NF1-related OPGs exhibited a decline in PhNR amplitude (p = 0.005) and an increase in PhNR peak-time during the follow-up (p = 0.013), whereas sporadic OPGs showed no significant changes. Tumor size remained stable in all patients on MRI. PhNR amplitude decreased over the observation period, suggesting progressive RGC dysfunction in NF1-related pediatric OPGs, despite stable size on MRI imaging. PhNR could serve as a non-invasive objective tool for assessing longitudinal changes in RGC function in the clinical management of childhood OPG.
Collapse
Affiliation(s)
- Dario Marangoni
- University Eye Clinic, Department of Medicine, Surgery and Health Science, University of Trieste, Strada di Fiume, 447, 34129, Trieste, Italy.
| | - Giorgio Placidi
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Elena D'Agostino
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Elisa De Siena
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Cesare Colosimo
- Radiology and Neuroradiology Department, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| | - Benedetto Falsini
- Ophthalmology Unit, Department of NeuroScience, Sensory Organs and Chest, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli, 8, 00136, Rome, Italy
| |
Collapse
|
5
|
Oka M, Nakajima S, Suzuki E, Yamamoto S, Ando K. Glucose uptake in pigment glia suppresses tau-induced inflammation and photoreceptor degeneration in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607919. [PMID: 39229232 PMCID: PMC11370381 DOI: 10.1101/2024.08.14.607919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Brain inflammation contributes to the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD). Glucose hypometabolism and glial activation are pathological features seen in AD brains; however, the connection between the two is not fully understood. Using a Drosophila model of AD, we identified that glucose metabolism in glia plays a critical role in neuroinflammation under disease conditions. Expression of human tau in the retinal cells, including photoreceptor neurons and pigment glia, causes photoreceptor degeneration accompanied by inclusion formation and swelling of lamina glial cells. We found that inclusions are formed by glial phagocytosis, and swelling of the laminal cortex correlates with the expression of antimicrobial peptides (AMPs). Co-expression of human glucose transporter 3 ( GLUT3 ) with tau in the retina does not affect tau levels but suppresses these inflammatory responses and photoreceptor degeneration. We also found that expression of GLUT3 , specifically in the pigment glia, is sufficient to suppress inflammatory phenotypes and mitigate photoreceptor degeneration in the tau-expressing retina. Our results suggest that glial glucose metabolism contributes to inflammatory responses and neurodegeneration in tauopathy. Highlights Tau expression in the fly retina induces glial activationPigment glial cells mediate inflammatory phenotypes in the degenerating retinaEnhanced glucose uptake in the pigment glia suppresses inflammation and photoreceptor neurodegeneration caused by tau expression.
Collapse
|
6
|
Oft HC, Simon DW, Sun D. New insights into metabolism dysregulation after TBI. J Neuroinflammation 2024; 21:184. [PMID: 39075578 PMCID: PMC11288120 DOI: 10.1186/s12974-024-03177-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024] Open
Abstract
Traumatic brain injury (TBI) remains a leading cause of death and disability that places a great physical, social, and financial burden on individuals and the health system. In this review, we summarize new research into the metabolic changes described in clinical TBI trials, some of which have already shown promise for informing injury classification and staging. We focus our discussion on derangements in glucose metabolism, cell respiration/mitochondrial function and changes to ketone and lipid metabolism/oxidation to emphasize potentially novel biomarkers for clinical outcome prediction and intervention and offer new insights into possible underlying mechanisms from preclinical research of TBI pathology. Finally, we discuss nutrition supplementation studies that aim to harness the gut/microbiome-brain connection and manipulate systemic/cellular metabolism to improve post-TBI recovery. Taken together, this narrative review summarizes published TBI-associated changes in glucose and lipid metabolism, highlighting potential metabolite biomarkers for clinical use, the cellular processes linking these markers to TBI pathology as well as the limitations and future considerations for TBI "omics" work.
Collapse
Affiliation(s)
- Helena C Oft
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dennis W Simon
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
7
|
Liao Z, Zhang Q, Ren N, Zhao H, Zheng X. Progress in mitochondrial and omics studies in Alzheimer's disease research: from molecular mechanisms to therapeutic interventions. Front Immunol 2024; 15:1418939. [PMID: 39040111 PMCID: PMC11260616 DOI: 10.3389/fimmu.2024.1418939] [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: 04/17/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Alzheimer's disease (Alzheimer's disease, AD) is a progressive neurological disorder characterized by memory loss and cognitive impairment. It is characterized by the formation of tau protein neurofibrillary tangles and β-amyloid plaques. Recent studies have found that mitochondria in neuronal cells of AD patients exhibit various dysfunctions, including reduced numbers, ultrastructural changes, reduced enzyme activity, and abnormal kinetics. These abnormal mitochondria not only lead to the loss of normal neuronal cell function, but are also a major driver of AD progression. In this review, we will focus on the advances of mitochondria and their multi-omics in AD research, with particular emphasis on how mitochondrial dysfunction in AD drives disease progression. At the same time, we will focus on summarizing how mitochondrial genomics technologies have revealed specific details of these dysfunctions and how therapeutic strategies targeting mitochondria may provide new directions for future AD treatments. By delving into the key mechanisms of mitochondria in AD related to energy metabolism, altered kinetics, regulation of cell death, and dysregulation of calcium-ion homeostasis, and how mitochondrial multi-omics technologies can be utilized to provide us with a better understanding of these processes. In the future, mitochondria-centered therapeutic strategies will be a key idea in the treatment of AD.
Collapse
Affiliation(s)
- Zuning Liao
- Department of Neurology, Fourth People’s Hospital of Jinan, Jinan, China
| | - Qiying Zhang
- Department of Internal Medicine, Jinan Municipal Government Hospital, Jinan, China
| | - Na Ren
- Pharmacy Department, Jinan Municipal People’s Government Organs Outpatient Department, Jinan, China
| | - Haiyan Zhao
- Department of Pharmacy, Qihe County People’s Hospital, Dezhou, China
| | - Xueyan Zheng
- Department of Pharmacy, Jinan Second People’s Hospital, Jinan, China
| |
Collapse
|
8
|
Ruan X, Yan W, Cao M, Daza RAM, Fong MY, Yang K, Wu J, Liu X, Palomares M, Wu X, Li A, Chen Y, Jandial R, Spitzer NC, Hevner RF, Wang SE. Breast cancer cell-secreted miR-199b-5p hijacks neurometabolic coupling to promote brain metastasis. Nat Commun 2024; 15:4549. [PMID: 38811525 PMCID: PMC11137082 DOI: 10.1038/s41467-024-48740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/09/2024] [Indexed: 05/31/2024] Open
Abstract
Breast cancer metastasis to the brain is a clinical challenge rising in prevalence. However, the underlying mechanisms, especially how cancer cells adapt a distant brain niche to facilitate colonization, remain poorly understood. A unique metabolic feature of the brain is the coupling between neurons and astrocytes through glutamate, glutamine, and lactate. Here we show that extracellular vesicles from breast cancer cells with a high potential to develop brain metastases carry high levels of miR-199b-5p, which shows higher levels in the blood of breast cancer patients with brain metastases comparing to those with metastatic cancer in other organs. miR-199b-5p targets solute carrier transporters (SLC1A2/EAAT2 in astrocytes and SLC38A2/SNAT2 and SLC16A7/MCT2 in neurons) to hijack the neuron-astrocyte metabolic coupling, leading to extracellular retention of these metabolites and promoting cancer cell growth. Our findings reveal a mechanism through which cancer cells of a non-brain origin reprogram neural metabolism to fuel brain metastases.
Collapse
Affiliation(s)
- Xianhui Ruan
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Wei Yan
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Minghui Cao
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Ray Anthony M Daza
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Miranda Y Fong
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Cancer Biology, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Kaifu Yang
- School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jun Wu
- Center for Comparative Medicine, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Xuxiang Liu
- Department of Cancer Biology, City of Hope Beckman Research Institute, Duarte, CA, USA
| | | | - Xiwei Wu
- Department of Computational and Quantitative Medicine, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Arthur Li
- Division of Biostatistics, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Yuan Chen
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Rahul Jandial
- Department of Surgery; City of Hope, Duarte, CA, USA
| | - Nicholas C Spitzer
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Kavli Institute for Brain and Mind, University of California San Diego, La Jolla, CA, USA
| | - Robert F Hevner
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Benarroch E. What Is the Role of Lactate in Brain Metabolism, Plasticity, and Neurodegeneration? Neurology 2024; 102:e209378. [PMID: 38574305 DOI: 10.1212/wnl.0000000000209378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
|
10
|
He Y, He T, Li H, Chen W, Zhong B, Wu Y, Chen R, Hu Y, Ma H, Wu B, Hu W, Han Z. Deciphering mitochondrial dysfunction: Pathophysiological mechanisms in vascular cognitive impairment. Biomed Pharmacother 2024; 174:116428. [PMID: 38599056 DOI: 10.1016/j.biopha.2024.116428] [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: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Vascular cognitive impairment (VCI) encompasses a range of cognitive deficits arising from vascular pathology. The pathophysiological mechanisms underlying VCI remain incompletely understood; however, chronic cerebral hypoperfusion (CCH) is widely acknowledged as a principal pathological contributor. Mitochondria, crucial for cellular energy production and intracellular signaling, can lead to numerous neurological impairments when dysfunctional. Recent evidence indicates that mitochondrial dysfunction-marked by oxidative stress, disturbed calcium homeostasis, compromised mitophagy, and anomalies in mitochondrial dynamics-plays a pivotal role in VCI pathogenesis. This review offers a detailed examination of the latest insights into mitochondrial dysfunction within the VCI context, focusing on both the origins and consequences of compromised mitochondrial health. It aims to lay a robust scientific groundwork for guiding the development and refinement of mitochondrial-targeted interventions for VCI.
Collapse
Affiliation(s)
- Yuyao He
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Tiantian He
- Sichuan Academy of Chinese Medicine Sciences, China
| | - Hongpei Li
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wei Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Biying Zhong
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yue Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Runming Chen
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yuli Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Huaping Ma
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Bin Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Wenyue Hu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Zhenyun Han
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
11
|
Zheng C, Xiao X, Zhao W, Yang Z, Guo S. Functional brain network controllability dysfunction in Alzheimer's disease and its relationship with cognition and gene expression profiling. J Neural Eng 2024; 21:026018. [PMID: 38502960 DOI: 10.1088/1741-2552/ad357e] [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: 09/11/2023] [Accepted: 03/19/2024] [Indexed: 03/21/2024]
Abstract
Objective. In recent studies, network control theory has been applied to clarify transitions between brain states, emphasizing the significance of assessing the controllability of brain networks in facilitating transitions from one state to another. Despite these advancements, the potential alterations in functional network controllability associated with Alzheimer's disease (AD), along with the underlying genetic mechanisms responsible for these alterations, remain unclear.Approach. We conducted a comparative analysis of functional network controllability measures between patients with AD (n= 64) and matched normal controls (NCs,n= 64). We investigated the association between altered controllability measures and cognitive function in AD. Additionally, we conducted correlation analyses in conjunction with the Allen Human Brain Atlas to identify genes whose expression was correlated with changes in functional network controllability in AD, followed by a set of analyses on the functional features of the identified genes.Main results. In comparison to NCs, patients with AD exhibited a reduction in average controllability, predominantly within the default mode network (DMN) (63% of parcellations), and an increase in average controllability within the limbic (LIM) network (33% of parcellations). Conversely, AD patients displayed a decrease in modal controllability within the LIM network (27% of parcellations) and an increase in modal controllability within the DMN (80% of parcellations). In AD patients, a significant positive correlation was found between the average controllability of the salience network and the mini-mental state examination scores. The changes in controllability measures exhibited spatial correlation with transcriptome profiles. The significant genes identified exhibited enrichment in neurobiologically relevant pathways and demonstrated preferential expression in various tissues, cell types, and developmental periods.Significance. Our findings have the potential to offer new insights into the genetic mechanisms underlying alterations in the controllability of functional networks in AD. Additionally, these results offered perspectives for a deeper understanding of the pathogenesis and the development of therapeutic strategies for AD.
Collapse
Affiliation(s)
- Chuchu Zheng
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Xiaoxia Xiao
- School of Informatics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, People's Republic of China
| | - Wei Zhao
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Zeyu Yang
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| | - Shuixia Guo
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha 410006, People's Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha 410006, People's Republic of China
| |
Collapse
|
12
|
Zhou P, Yu ZC, Cao C, Cui HR, Ding MC, Yang CX, Liao M. Pyruvate maintains and enhances the pro-inflammatory response of microglia caused by glucose deficiency in early stroke. J Cell Biochem 2024; 125:e30524. [PMID: 38226453 DOI: 10.1002/jcb.30524] [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: 11/07/2023] [Accepted: 12/29/2023] [Indexed: 01/17/2024]
Abstract
Pro-inflammatory microglia mainly rely on glycolysis to maintain cytokine production during ischemia, accompanied by an increase in inducible nitric oxide synthase (iNOS) and monocarboxylate transporter 1 (MCT1). The role of energy metabolism in the pro-inflammatory response of microglia is currently unclear. In this study, we tested the response of microglia in mice after cerebral ischemia and simulated an energy environment in vitro using low glucose culture medium. The research results indicate that the expression levels of iNOS and arginase 1 (ARG1) increase in the ischemic mouse brain, but the upregulation of MCT1 expression is mainly present in iNOS positive microglia. In microglia exposed to low glucose conditions, iNOS and MCT1 levels increased, while ARG1 levels decreased. Under the same conditions, knocking down MCT1 in microglia leads to a decrease in iNOS levels, while overexpression of MCT1 leads to the opposite result. The use of NF-κB inhibitors reduced the expression levels of iNOS and MCT1 in microglia. In summary, our data indicate that pyruvate maintains and enhances the NF-κB regulated pro-inflammatory response of microglia induced by low glucose.
Collapse
Affiliation(s)
- Peng Zhou
- Institute of Neuroscience, Basic Medical College of Wenzhou Medical University, Wenzhou, China
- Department of Anatomy, Basic Medical College of Wenzhou Medical University, Wenzhou, China
| | - Zhe-Cheng Yu
- Institute of Neuroscience, Basic Medical College of Wenzhou Medical University, Wenzhou, China
| | - Cong Cao
- Institute of Neuroscience, Basic Medical College of Wenzhou Medical University, Wenzhou, China
| | - Huai-Rui Cui
- Department of Anatomy, Basic Medical College of Wenzhou Medical University, Wenzhou, China
| | - Mao-Chao Ding
- Department of Anatomy, Basic Medical College of Wenzhou Medical University, Wenzhou, China
| | - Chao-Xian Yang
- Department of Anatomy, Southwest Medical University, Luzhou, China
| | - Min Liao
- Institute of Neuroscience, Basic Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Makhlouf M, Souza DG, Kurian S, Bellaver B, Ellis H, Kuboki A, Al-Naama A, Hasnah R, Venturin GT, Costa da Costa J, Venugopal N, Manoel D, Mennella J, Reisert J, Tordoff MG, Zimmer ER, Saraiva LR. Short-term consumption of highly processed diets varying in macronutrient content impair the sense of smell and brain metabolism in mice. Mol Metab 2024; 79:101837. [PMID: 37977411 PMCID: PMC10724696 DOI: 10.1016/j.molmet.2023.101837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/29/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
OBJECTIVE Food processing greatly contributed to increased food safety, diversity, and accessibility. However, the prevalence of highly palatable and highly processed food in our modern diet has exacerbated obesity rates and contributed to a global health crisis. While accumulating evidence suggests that chronic consumption of such foods is detrimental to sensory and neural physiology, it is unclear whether its short-term intake has adverse effects. Here, we assessed how short-term consumption (<2 months) of three diets varying in composition and macronutrient content influence olfaction and brain metabolism in mice. METHODS The diets tested included a grain-based standard chow diet (CHOW; 54% carbohydrate, 32% protein, 14% fat; #8604 Teklad Rodent diet , Envigo Inc.), a highly processed control diet (hpCTR; 70% carbohydrate, 20% protein, 10% fat; #D12450B, Research Diets Inc.), and a highly processed high-fat diet (hpHFD; 20% carbohydrate, 20% protein, 60% fat; #D12492, Research Diets Inc.). We performed behavioral and metabolic phenotyping, electro-olfactogram (EOG) recordings, brain glucose metabolism imaging, and mitochondrial respirometry in different brain regions. We also performed RNA-sequencing (RNA-seq) in the nose and across several brain regions, and conducted differential expression analysis, gene ontology, and network analysis. RESULTS We show that short-term consumption of the two highly processed diets, but not the grain-based diet, regardless of macronutrient content, adversely affects odor-guided behaviors, physiological responses to odorants, transcriptional profiles in the olfactory mucosa and brain regions, and brain glucose metabolism and mitochondrial respiration. CONCLUSIONS Even short periods of highly processed food consumption are sufficient to cause early olfactory and brain abnormalities, which has the potential to alter food choices and influence the risk of developing metabolic disease.
Collapse
Affiliation(s)
| | - Débora G Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Bruna Bellaver
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Hillary Ellis
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Akihito Kuboki
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | | | - Reem Hasnah
- Sidra Medicine, PO Box 26999, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Gianina Teribele Venturin
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Julie Mennella
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Johannes Reisert
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Michael G Tordoff
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil; Department of Pharmacology, UFRGS, Porto Alegre, Brazil; Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil; McGill Centre for Studies in Aging, Montreal, Canada.
| | - Luis R Saraiva
- Sidra Medicine, PO Box 26999, Doha, Qatar; Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
14
|
Nave KA, Asadollahi E, Sasmita A. Expanding the function of oligodendrocytes to brain energy metabolism. Curr Opin Neurobiol 2023; 83:102782. [PMID: 37703600 DOI: 10.1016/j.conb.2023.102782] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023]
Abstract
Oligodendrocytes are best known for wrapping myelin, a unique specialization that enables energy-efficient and fast axonal impulse propagation in white matter tracts and fibers of the cortical circuitry. However, myelinating oligodendrocytes have additional metabolic functions that are only gradually understood, including the regulated release of pyruvate/lactate and extracellular vesicles, both of which are in support of the axonal energy balance. The axon-supportive functions of glial cells are older than myelin in nervous system evolution and implicate oligodendrocyte dysfunction and loss of myelin integrity as a risk factor for progressive neurodegeneration in brain diseases.
Collapse
Affiliation(s)
- Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Multidisciplinary Sciences, Göttingen.
| | - Ebrahim Asadollahi
- Department of Neurogenetics, Max Planck Institute of Multidisciplinary Sciences, Göttingen. https://twitter.com/EbrahimAsadoll3
| | - Andrew Sasmita
- Department of Neurogenetics, Max Planck Institute of Multidisciplinary Sciences, Göttingen. https://twitter.com/AOSasmita
| |
Collapse
|
15
|
Preziuso A, Piccirillo S, Cerqueni G, Serfilippi T, Terenzi V, Vinciguerra A, Orciani M, Amoroso S, Magi S, Lariccia V. Exploring the Role of NCX1 and NCX3 in an In Vitro Model of Metabolism Impairment: Potential Neuroprotective Targets for Alzheimer's Disease. BIOLOGY 2023; 12:1005. [PMID: 37508434 PMCID: PMC10376230 DOI: 10.3390/biology12071005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a widespread neurodegenerative disorder, affecting a large number of elderly individuals worldwide. Mitochondrial dysfunction, metabolic alterations, and oxidative stress are regarded as cooperating drivers of the progression of AD. In particular, metabolic impairment amplifies the production of reactive oxygen species (ROS), resulting in detrimental alterations to intracellular Ca2+ regulatory processes. The Na+/Ca2+ exchanger (NCX) proteins are key pathophysiological determinants of Ca2+ and Na+ homeostasis, operating at both the plasma membrane and mitochondria levels. Our study aimed to explore the role of NCX1 and NCX3 in retinoic acid (RA) differentiated SH-SY5Y cells treated with glyceraldehyde (GA), to induce impairment of the default glucose metabolism that typically precedes Aβ deposition or Tau protein phosphorylation in AD. By using an RNA interference-mediated approach to silence either NCX1 or NCX3 expression, we found that, in GA-treated cells, the knocking-down of NCX3 ameliorated cell viability, increased the intracellular ATP production, and reduced the oxidative damage. Remarkably, NCX3 silencing also prevented the enhancement of Aβ and pTau levels and normalized the GA-induced decrease in NCX reverse-mode activity. By contrast, the knocking-down of NCX1 was totally ineffective in preventing GA-induced cytotoxicity except for the increase in ATP synthesis. These findings indicate that NCX3 and NCX1 may differently influence the evolution of AD pathology fostered by glucose metabolic dysfunction, thus providing a potential target for preventing AD.
Collapse
Affiliation(s)
- Alessandra Preziuso
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Giorgia Cerqueni
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Valentina Terenzi
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Antonio Vinciguerra
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences-Histology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health-Pharmacology, School of Medicine, University "Politecnica delle Marche", Via Tronto 10/A, 60126 Ancona, Italy
| |
Collapse
|
16
|
Nemcova M, Seidlova V, Zukal J, Dundarova H, Bednarikova S, Pikula J. Bat-derived cells use glucose as a cryoprotectant. J Therm Biol 2023; 115:103652. [PMID: 37451039 DOI: 10.1016/j.jtherbio.2023.103652] [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: 11/18/2022] [Revised: 06/20/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023]
Abstract
Evolution of heterothermy in environments with variable temperatures has allowed bats to survive food scarcity during seasonal climatic extremes by using torpor as a hibernation strategy. The controlled reduction of body temperature and metabolism through complex behavioural and physiological adaptations at organismal, organ, cellular and molecular levels includes the ability of tissues and cells to adapt to temperature alterations. Based on the prediction that cells of different tissues cultured in vitro would differ in their ability to withstand freezing and thawing of the medium, we determined the survival rate of bat-derived cells following exposure to -20 °C for 24 h in media with no cryoprotective agents or medium supplemented by glucose in concentration range 0-3333 mM. Cell survival rates were determined in relation to availability of glucose in the medium, organ origin, cell concentration and bat species. In general, increased glucose helped cells survive at sub-zero temperatures, though concentrations up to 80-fold higher than those found in chiropterans were needed. However, cells in glucose-free phosphate buffered saline also survived, suggesting that other mechanisms may be contributing to cell survival at low temperatures. Highest in vitro viability was observed in nervus olfactorius-derived cell cultures, with high survival rates and rapid re-growth under optimal conditions after exposure to -20 °C. Kidney cells from different bat species showed comparable overall survival rate patterns, though smaller chiropteran species appeared to utilise lower glucose levels as a cryoprotectant than larger species. Our in vitro data provide evidence that cells of heterothermic bats can survive sub-zero temperatures and that higher glucose levels in important tissues significantly improve hibernation survival at extremely low temperatures.
Collapse
Affiliation(s)
- Monika Nemcova
- Department of Ecology and Diseases of Zoo Animals, Game, Fish and Bees, University of Veterinary Sciences Brno, Palackého tř. 1946/1, 612 42 Brno, Czech Republic.
| | - Veronika Seidlova
- Department of Ecology and Diseases of Zoo Animals, Game, Fish and Bees, University of Veterinary Sciences Brno, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
| | - Jan Zukal
- Institute of Vertebrate Biology, Czech Academy of Sciences, Květná 8, 603 65 Brno, Czech Republic
| | - Heliana Dundarova
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, 1 Tsar Osvoboditel Blvd., 1000 Sofia, Bulgaria
| | - Sarka Bednarikova
- Department of Ecology and Diseases of Zoo Animals, Game, Fish and Bees, University of Veterinary Sciences Brno, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
| | - Jiri Pikula
- Department of Ecology and Diseases of Zoo Animals, Game, Fish and Bees, University of Veterinary Sciences Brno, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
| |
Collapse
|
17
|
Iino S, Oya S, Kakutani T, Kohno H, Kubo T. Identification of ecdysone receptor target genes in the worker honey bee brains during foraging behavior. Sci Rep 2023; 13:10491. [PMID: 37380789 DOI: 10.1038/s41598-023-37001-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 06/14/2023] [Indexed: 06/30/2023] Open
Abstract
Ecdysone signaling plays central roles in morphogenesis and female ovarian development in holometabolous insects. In the European honey bee (Apis mellifera L.), however, ecdysone receptor (EcR) is expressed in the brains of adult workers, which have already undergone metamorphosis and are sterile with shrunken ovaries, during foraging behavior. Aiming at unveiling the significance of EcR signaling in the worker brain, we performed chromatin-immunoprecipitation sequencing of EcR to search for its target genes using the brains of nurse bees and foragers. The majority of the EcR targets were common between the nurse bee and forager brains and some of them were known ecdysone signaling-related genes. RNA-sequencing analysis revealed that some EcR target genes were upregulated in forager brains during foraging behavior and some were implicated in the repression of metabolic processes. Single-cell RNA-sequencing analysis revealed that EcR and its target genes were expressed mostly in neurons and partly in glial cells in the optic lobes of the forager brain. These findings suggest that in addition to its role during development, EcR transcriptionally represses metabolic processes during foraging behavior in the adult worker honey bee brain.
Collapse
Affiliation(s)
- Shiori Iino
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Satoyo Oya
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tetsuji Kakutani
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroki Kohno
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takeo Kubo
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
18
|
Xu L, Liu R, Qin Y, Wang T. Brain metabolism in Alzheimer's disease: biological mechanisms of exercise. Transl Neurodegener 2023; 12:33. [PMID: 37365651 DOI: 10.1186/s40035-023-00364-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
Alzheimer's disease (AD) is a major subtype of neurodegenerative dementia caused by long-term interactions and accumulation of multiple adverse factors, accompanied by dysregulation of numerous intracellular signaling and molecular pathways in the brain. At the cellular and molecular levels, the neuronal cellular milieu of the AD brain exhibits metabolic abnormalities, compromised bioenergetics, impaired lipid metabolism, and reduced overall metabolic capacity, which lead to abnormal neural network activity and impaired neuroplasticity, thus accelerating the formation of extracellular senile plaques and intracellular neurofibrillary tangles. The current absence of effective pharmacological therapies for AD points to the urgent need to investigate the benefits of non-pharmacological approaches such as physical exercise. Despite the evidence that regular physical activity can improve metabolic dysfunction in the AD state, inhibit different pathophysiological molecular pathways associated with AD, influence the pathological process of AD, and exert a protective effect, there is no clear consensus on the specific biological and molecular mechanisms underlying the advantages of physical exercise. Here, we review how physical exercise improves crucial molecular pathways and biological processes associated with metabolic disorders in AD, including glucose metabolism, lipid metabolism, Aβ metabolism and transport, iron metabolism and tau pathology. How metabolic states influence brain health is also presented. A better knowledge on the neurophysiological mechanisms by which exercise improves AD metabolism can contribute to the development of novel drugs and improvement of non-pharmacological interventions.
Collapse
Affiliation(s)
- Longfei Xu
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
| | - Ran Liu
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China
| | - Yingkai Qin
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China.
| | - Tianhui Wang
- Institute of Environmental and Operational Medicine, Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, 300050, China.
- Tianjin Key Laboratory of Exercise Physiology & Sports Medicine, Tianjin University of Sport, Tianjin, 301617, China.
| |
Collapse
|
19
|
Cheataini F, Ballout N, Al Sagheer T. The effect of neuroinflammation on the cerebral metabolism at baseline and after neural stimulation in neurodegenerative diseases. J Neurosci Res 2023. [PMID: 37186320 DOI: 10.1002/jnr.25198] [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: 11/06/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023]
Abstract
Neuroinflammation is a reaction of nervous tissue to an attack caused by an infection, a toxin, or a neurodegenerative disease. It involves brain metabolism adaptation in order to meet the increased energy needs of glial cell activation, but the nature of these adaptations is still unknown. Increasing interest concerning neuroinflammation leads to the identification of its role in neurodegenerative diseases. Few reports studied the effect of metabolic alteration on neuroinflammation. Metabolic damage initiates a pro-inflammatory response by microglial activation. Moreover, the exact neuroinflammation effect on cerebral cell metabolism remains unknown. In this study, we reviewed systematically the neuroinflammation effect in animal models' brains. All articles showing the relationship of neuroinflammation with brain metabolism, or with neuronal stimulation in neurodegenerative diseases were considered. Moreover, this review examines also the mitochondrial damage effect in neurodegeneration diseases. Then, different biosensors are classified regarding their importance in the determination of metabolite change. Finally, some therapeutic drugs inhibiting neuroinflammation are cited. Neuroinflammation increases lymphocyte infiltration and cytokines' overproduction, altering cellular energy homeostasis. This review demonstrates the importance of neuroinflammation as a mediator of disease progression. Further, the spread of depolarization effects pro-inflammatory genes expression and microglial activation, which contribute to the degeneration of neurons, paving the road to better management and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Fatima Cheataini
- Neuroscience Research Center (NRC), Faculty of Medical Science, Lebanese University, Hadath, Beirut, Lebanon
| | - Nissrine Ballout
- Neuroscience Research Center (NRC), Faculty of Medical Science, Lebanese University, Hadath, Beirut, Lebanon
| | - Tareq Al Sagheer
- Neuroscience Research Center (NRC), Faculty of Medical Science, Lebanese University, Hadath, Beirut, Lebanon
| |
Collapse
|
20
|
Sun Y, Zabihi M, Li Q, Li X, Kim BJ, Ubogu EE, Raja SN, Wesselmann U, Zhao C. Drug Permeability: From the Blood-Brain Barrier to the Peripheral Nerve Barriers. ADVANCED THERAPEUTICS 2023; 6:2200150. [PMID: 37649593 PMCID: PMC10465108 DOI: 10.1002/adtp.202200150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Indexed: 01/20/2023]
Abstract
Drug delivery into the peripheral nerves and nerve roots has important implications for effective local anesthesia and treatment of peripheral neuropathies and chronic neuropathic pain. Similar to drugs that need to cross the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) to gain access to the central nervous system (CNS), drugs must cross the peripheral nerve barriers (PNB), formed by the perineurium and blood-nerve barrier (BNB) to modulate peripheral axons. Despite significant progress made to develop effective strategies to enhance BBB permeability in therapeutic drug design, efforts to enhance drug permeability and retention in peripheral nerves and nerve roots are relatively understudied. Guided by knowledge describing structural, molecular and functional similarities between restrictive neural barriers in the CNS and peripheral nervous system (PNS), we hypothesize that certain CNS drug delivery strategies are adaptable for peripheral nerve drug delivery. In this review, we describe the molecular, structural and functional similarities and differences between the BBB and PNB, summarize and compare existing CNS and peripheral nerve drug delivery strategies, and discuss the potential application of selected CNS delivery strategies to improve efficacious drug entry for peripheral nerve disorders.
Collapse
Affiliation(s)
- Yifei Sun
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Mahmood Zabihi
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Qi Li
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Xiaosi Li
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Brandon J. Kim
- Department of Biological Sciences, The University of Alabama, Tuscaloosa AL 35487, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham AL 35294, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa AL 35487, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa AL 35487, USA
| | - Eroboghene E. Ubogu
- Division of Neuromuscular Disease, Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Srinivasa N. Raja
- Division of Pain Medicine, Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Ursula Wesselmann
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, and Department of Neurology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Consortium for Neuroengineering and Brain-Computer Interfaces, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa AL 35487, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa AL 35487, USA
| |
Collapse
|
21
|
Cross-talk between energy and redox metabolism in astrocyte-neuron functional cooperation. Essays Biochem 2023; 67:17-26. [PMID: 36805653 PMCID: PMC10011404 DOI: 10.1042/ebc20220075] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023]
Abstract
Astrocytes show unique anatomical, morphological, and metabolic features to take up substrates from the blood and metabolize them for local delivery to active synapses to sustain neuron function. In the present review, we specifically focus on key molecular aspects of energy and redox metabolism that facilitate this astrocyte-neuronal coupling in a controlled manner. Basal glycolysis is co-ordinated by the anaphase-promoting complex/cyclosome (APC/C)-Cdh1, a ubiquitin ligase that targets the proglycolytic enzyme 6-phosphofructokinase-2,6-bisphosphastate-3 (PFKFB3) for degradation. APC/C-Cdh1 activity is more robust in neurons than in astrocytes, which determine that PFKFB3 abundance and glycolytic rate are weaker in neurons. The low PFKFB3 activity in neurons facilitates glucose-6-phosphate oxidation via the pentose-phosphate pathway, which promotes antioxidant protection. Conversely, the high PFKFB3 activity in astrocytes allows the production and release of glycolytic lactate, which is taken up by neurons that use it as an oxidizable substrate. Importantly, the mitochondrial respiratory chain is tighter assembled in neurons than in astrocytes, thus the bioenergetic efficiency of mitochondria is higher in neurons. Because of this, the production of reactive oxygen species (mROS) by mitochondrial complex I is very low in neurons and very high in astrocytes. Such a naturally occurring high abundance of mROS in astrocytes physiologically determines a specific transcriptional fingerprint that contributes to sustaining cognitive performance. We conclude that the energy and redox metabolism of astrocytes must complementarily match that of neurons to regulate brain function and animal welfare.
Collapse
|
22
|
Yassine HN, Self W, Kerman BE, Santoni G, Navalpur Shanmugam N, Abdullah L, Golden LR, Fonteh AN, Harrington MG, Gräff J, Gibson GE, Kalaria R, Luchsinger JA, Feldman HH, Swerdlow RH, Johnson LA, Albensi BC, Zlokovic BV, Tanzi R, Cunnane S, Samieri C, Scarmeas N, Bowman GL. Nutritional metabolism and cerebral bioenergetics in Alzheimer's disease and related dementias. Alzheimers Dement 2023; 19:1041-1066. [PMID: 36479795 PMCID: PMC10576546 DOI: 10.1002/alz.12845] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/12/2022] [Accepted: 10/05/2022] [Indexed: 12/13/2022]
Abstract
Disturbances in the brain's capacity to meet its energy demand increase the risk of synaptic loss, neurodegeneration, and cognitive decline. Nutritional and metabolic interventions that target metabolic pathways combined with diagnostics to identify deficits in cerebral bioenergetics may therefore offer novel therapeutic potential for Alzheimer's disease (AD) prevention and management. Many diet-derived natural bioactive components can govern cellular energy metabolism but their effects on brain aging are not clear. This review examines how nutritional metabolism can regulate brain bioenergetics and mitigate AD risk. We focus on leading mechanisms of cerebral bioenergetic breakdown in the aging brain at the cellular level, as well as the putative causes and consequences of disturbed bioenergetics, particularly at the blood-brain barrier with implications for nutrient brain delivery and nutritional interventions. Novel therapeutic nutrition approaches including diet patterns are provided, integrating studies of the gut microbiome, neuroimaging, and other biomarkers to guide future personalized nutritional interventions.
Collapse
Affiliation(s)
- Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wade Self
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bilal E Kerman
- Department of Medicine, Keck School of Medicine, University of Southern, California, Los Angeles, California, USA
| | - Giulia Santoni
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - NandaKumar Navalpur Shanmugam
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Lesley R Golden
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alfred N Fonteh
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Huntington Medical Research Institutes, Pasadena, California, USA
| | - Michael G Harrington
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale Lausanne (EPFL), Lausanne, Switzerland
| | - Gary E Gibson
- Brain and Mind Research Institute, Weill Cornell Medicine, Burke Neurological Institute, White Plains, New York, USA
| | - Raj Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jose A Luchsinger
- Department of Medicine and Epidemiology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Howard H Feldman
- Department of Neurosciences, University of California, San Diego, California, USA
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Benedict C Albensi
- Nova Southeastern Univ. College of Pharmacy, Davie, Florida, USA
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rudolph Tanzi
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Cunnane
- Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Cécilia Samieri
- Univ. Bordeaux, INSERM, BPH, U1219, F-33000, Bordeaux, France
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, Columbia University, New York City, New York, USA
| | - Gene L Bowman
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Helfgott Research Institute, National University of Natural Medicine, Portland, Oregon, USA
| |
Collapse
|
23
|
Jiang C, Lu Y, Zhu R, Zong Y, Huang Y, Wang D, Da Z, Yu B, Shen L, Cao Q. Pyruvate dehydrogenase beta subunit (Pdhb) promotes peripheral axon regeneration by regulating energy supply and gene expression. Exp Neurol 2023; 363:114368. [PMID: 36863478 DOI: 10.1016/j.expneurol.2023.114368] [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: 11/23/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023]
Abstract
Key metabolic enzymes not only regulate Glucose, lipid, amino acid metabolism to serve the cellular energy needs, but also modulate noncanonical or nonmetabolic signaling pathway such as gene expression, cell-cycle progression, DNA repair, apoptosis and cell proliferation in regulating the pathologic progression of disease. However, the role of glycometabolism in peripheral nerve axon regeneration is little known. In this study, we investigated the expression of Pyruvate dehydrogenase E1(PDH), a key enzyme linking glycolysis and the tricarboxylic acid (TCA) cycle, with qRT-PCR and found that pyruvate dehydrogenase beta subunit (Pdhb) is up-regulated at the early stage during peripheral nerve injury. The knockdown of Pdhb inhibits neurite outgrowth of primary DRG neurons in vitro and restrains axon regeneration of sciatic nerve after crush injury. Pdhb overexpression promoting axonal regeneration is reversed by knockdown of Monocarboxylate transporter 2(Mct2), a transporter involved in the transport and metabolism of lactate, indicating Pdhb promoting axon regeneration depends on lactate for energy supply. Given the nucleus-localization of Pdhb, further analysis revealed that Pdhb enhances the acetylation of H3K9 and affecting the expression of genes involved in arachidonic acid metabolism and Ras signaling pathway, such as Rsa-14-44 and Pla2g4a, thereby promoting axon regeneration. Collectively, our data indicates that Pdhb is a positive dual modulator of energy generation and gene expression in regulating peripheral axon regeneration.
Collapse
Affiliation(s)
- Chunyi Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yan Lu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Clinical Laboratory, Nantong Third Hospital Affiliated to Nantong University, Nantong 226001, China
| | - Ran Zhu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Ying Zong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yuchen Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Dong Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Zhanyun Da
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Longxiang Shen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Qianqian Cao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
24
|
Lamanna-Rama N, MacDowell KS, López G, Leza JC, Desco M, Ambrosio E, Soto-Montenegro ML. Neuroimaging revealed long-lasting glucose metabolism changes to morphine withdrawal in rats pretreated with the cannabinoid agonist CP-55,940 during periadolescence. Eur Neuropsychopharmacol 2023; 69:60-76. [PMID: 36780817 DOI: 10.1016/j.euroneuro.2023.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 02/13/2023]
Abstract
This study evaluates the long-term effects of a six and 14-week morphine withdrawal in rats pretreated with a cannabinoid agonist (CP-55,940, CP) during periadolescence. Wistar rats (33 males; 32 females) were treated with CP or its vehicle (VH) from postnatal day (PND) 28-38. At PND100, rats performed morphine self-administration (MSA, 15d/12 h/session). Eight groups were defined according to pretreatment (CP), treatment (morphine), and sex. Three [18F]FDG-PET brain images were acquired: after MSA, and after six and 14 weeks of withdrawal. PET data were analyzed with SPM12. Endocannabinoid (EC) markers were evaluated in frozen brain tissue at endpoint. Females showed a higher mean number of self-injections than males. A main Sex effect on global brain metabolism was found. FDG uptake in males was discrete, whereas females showed greater brain metabolism changes mainly in areas of the limbic system after morphine treatment. Moreover, the morphine-induced metabolic pattern in females was exacerbated when CP was previously present. In addition, the CP-Saline male group showed reduced CB1R, MAGL expression, and NAPE/FAAH ratio compared to the control group, and morphine was able to reverse CB1R and MAGL expression almost to control levels. In conclusion, females showed greater and longer-lasting metabolic changes after morphine withdrawal than males, indicating a higher vulnerability and a different sensitivity to morphine in subjects pre-exposed to CP. In contrast, males primarily showed changes in EC markers. Together, our results suggest that CP pre-exposure contributes to the modulation of brain metabolism and EC systems in a sex-dependent manner.
Collapse
Affiliation(s)
- N Lamanna-Rama
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain
| | - K S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento de Farmacología & Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Imas12, IUIN, Spain
| | - G López
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Psicobiología, Facultad de Psicología, National University for Distance Learning (UNED), Madrid, Spain; Faculty of Health Science, Universidad Internacional de La Rioja (UNIR), Spain
| | - J C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento de Farmacología & Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Imas12, IUIN, Spain
| | - M Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| | - E Ambrosio
- Departamento de Psicobiología, Facultad de Psicología, National University for Distance Learning (UNED), Madrid, Spain.
| | - M L Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Grupo de Fisiopatología y Farmacología del Sistema Digestivo de la Universidad Rey Juan Carlos (NEUGUT), Madrid, España.
| |
Collapse
|
25
|
Xu H, Chen K, Zhu H, Bu J, Yang L, Chen F, Ma H, Qu X, Zhang R, Liu H. Neurovascular coupling changes in patients with magnetic resonance imaging negative focal epilepsy. Epilepsy Behav 2023; 138:109035. [PMID: 36535109 DOI: 10.1016/j.yebeh.2022.109035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
Brain neuron activity is closely related to cerebral blood flow (CBF) changes. Alterations in the regional homogeneity (ReHo) and CBF occur in patients with magnetic resonance imaging negative focal epilepsy (FEP-MRI-). However, the coupling alterations of ReHo and CBF in FEP-MRI- remain unclear. The study aims to explore neurovascular coupling alterations and their clinical implication in FEP-MRI-. We collected resting-state magnetic resonance imaging (MRI) data from 31 healthy controls (HCs) and 48 patients with FEP-MRI-,including three-dimensional (3D) T1-weighted imaging, 3D arterial spin labeling (ASL) imaging,and resting-state functional MRI (rs-fMRI). The CBF and ReHo values were calculated from the ASL and rs-fMRI data, respectively. The CBF/ReHo ratio per voxel and whole-brain CBF-ReHo coupling were compared between the two groups. Correlation analysis involved the CBF/ReHo ratio and clinical indicators in FEP-MRI-. Patients with FEP-MRI- showed significantly increased cross-subject CBF-ReHo and global cross-voxel CBF-ReHo coupling. The CBF/ReHo ratio was higher in the bilateral orbitofrontal gyrus, right parietal lobe, and right middle frontal gyrus of patients with FEP-MRI-. Nevertheless, this ratio was lower in the bilateral supplementary motor areas, the left middle and posterior cingulate gyrus, and the right central sulcus cover. The CBF/ReHo ratio was markedly correlated with cognitive function, memory, intelligence, and epilepsy duration in the above abnormal brain regions. CBF/ReHo ratio may be useful as an indicator of neuropathological mechanisms. These results support the hypothesis that CBF/ReHo ratio relates to the neuropathological mechanisms of FEP-MRI-. Furthermore, it offers new perspectives for studying the mechanisms of MRI-negative epilepsy.
Collapse
Affiliation(s)
- Honghao Xu
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Kefan Chen
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Haitao Zhu
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Jinxin Bu
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Lu Yang
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Fangqing Chen
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Haiyan Ma
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Xuefeng Qu
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Rui Zhang
- Department of Functional Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China.
| | - Hongyi Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
26
|
Li M, Tang H, Li Z, Tang W. Emerging Treatment Strategies for Cerebral Ischemia-Reperfusion Injury. Neuroscience 2022; 507:112-124. [PMID: 36341725 DOI: 10.1016/j.neuroscience.2022.10.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Cerebral ischemia-reperfusion injury (CI/RI) injury is a common feature of ischemic stroke which occurs when the blood supply is restored after a period of ischemia in the brain. Reduced blood-flow to the brain during CI/RI compromises neuronal cell health as a result of mitochondrial dysfunction, oxidative stress, cytokine production, inflammation and tissue damage. Reperfusion therapy during CI/RI can restore the blood flow to ischemic regions of brain which are not yet infarcted. The long-term goal of CI/RI therapy is to reduce stroke-related neuronal cell death, disability and mortality. A range of drug and interventional therapies have emerged that can alleviate CI/RI mediated oxidative stress, inflammation and apoptosis in the brain. Herein, we review recent studies on CI/RI interventions for which a mechanism of action has been described and the potential of these therapeutic modalities for future use in the clinic.
Collapse
Affiliation(s)
- Mengxing Li
- College of Acupuncture and Massage (Rehabilitation Medical College), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Heyong Tang
- College of Integrated Chinese and Western Medicine (School of Life Sciences), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zhen Li
- College of Acupuncture and Massage (Rehabilitation Medical College), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wei Tang
- College of Acupuncture and Massage (Rehabilitation Medical College), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
27
|
Gale JR, Gedeon JY, Donnelly CJ, Gold MS. Local translation in primary afferents and its contribution to pain. Pain 2022; 163:2302-2314. [PMID: 35438669 PMCID: PMC9579217 DOI: 10.1097/j.pain.0000000000002658] [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: 09/30/2021] [Accepted: 04/08/2022] [Indexed: 02/06/2023]
Abstract
ABSTRACT Chronic pain remains a significant problem due to its prevalence, impact, and limited therapeutic options. Progress in addressing chronic pain is dependent on a better understanding of underlying mechanisms. Although the available evidence suggests that changes within the central nervous system contribute to the initiation and maintenance of chronic pain, it also suggests that the primary afferent plays a critical role in all phases of the manifestation of chronic pain in most of those who suffer. Most notable among the changes in primary afferents is an increase in excitability or sensitization. A number of mechanisms have been identified that contribute to primary afferent sensitization with evidence for both increases in pronociceptive signaling molecules, such as voltage-gated sodium channels, and decreases in antinociceptive signaling molecules, such as voltage-dependent or calcium-dependent potassium channels. Furthermore, these changes in signaling molecules seem to reflect changes in gene expression as well as posttranslational processing. A mechanism of sensitization that has received far less attention, however, is the local or axonal translation of these signaling molecules. A growing body of evidence indicates that this process not only is dynamically regulated but also contributes to the initiation and maintenance of chronic pain. Here, we review the biology of local translation in primary afferents and its relevance to pain pathobiology.
Collapse
Affiliation(s)
- Jenna R Gale
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Jeremy Y Gedeon
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | | | - Michael S Gold
- Corresponding author: Michael S Gold, PhD, Department of Neurobiology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, P: 412-383-5367,
| |
Collapse
|
28
|
Glyceryl triacetate feeding in mice increases plasma acetate levels but has no anticonvulsant effects in acute electrical seizure models. Epilepsy Behav 2022; 137:108964. [PMID: 36343532 DOI: 10.1016/j.yebeh.2022.108964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Acetate has been shown to have neuroprotective and anti-inflammatory effects. It is oxidized by astrocytes and can thus provide auxiliary energy to the brain in addition to glucose. Therefore, we hypothesized that it may protect against seizures, which is investigated here by feeding glyceryl triacetate (GTA), to provide high amounts of acetate without raising sodium or acid levels. METHOD CD1 male mice were fed controlled diets with or without GTA for up to three weeks. Body weights, blood glucose levels, plasma short-chain fatty acid levels, and other hematological parameters were monitored. Seizure thresholds were determined in 6 Hz and maximal electroshock seizure threshold (MEST) tests. Antioxidant capacities were evaluated in the cerebral cortex and plasma using a ferric reducing antioxidant power (FRAP) assay and Trolox equivalent antioxidant capacity assay. RESULTS Body weight gain was similar with both diets with and without GTA in two experiments. Glyceryl triacetate-fed groups showed 2-3- and 1.6-fold increased acetate and propionate levels in plasma, respectively. Glucose levels were unaltered in blood collected from the tail tip but increased in trunk blood. No differences were found in the activity of cerebral cortex acetyl-CoA synthetase. In the 6 Hz threshold test, seizure thresholds were lower by 3 mA and 2.4 mA after 8 and 14 days, respectively, in the GTA compared to the control diet-fed group, but showed no difference on day 16, showing that GTA has small, but inconsistent proconvulsant effects in this model. In MEST tests, a slightly increased seizure threshold (1 mA) was found on day 19 in the GTA-fed group, but not in another experiment on day 21. There were no differences in antioxidant capacity in plasma or cortex between the two groups. CONCLUSION Glyceryl triacetate feeding showed no antioxidant effects nor beneficial changes in acute electrical seizure threshold mouse models, despite its ability to increase plasma acetate levels.
Collapse
|
29
|
Shackleford G, Marziali LN, Sasaki Y, Claessens A, Ferri C, Weinstock NI, Rossor AM, Silvestri NJ, Wilson ER, Hurley E, Kidd GJ, Manohar S, Ding D, Salvi RJ, Feltri ML, D’Antonio M, Wrabetz L. A new mouse model of Charcot-Marie-Tooth 2J neuropathy replicates human axonopathy and suggest alteration in axo-glia communication. PLoS Genet 2022; 18:e1010477. [PMID: 36350884 PMCID: PMC9707796 DOI: 10.1371/journal.pgen.1010477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/29/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022] Open
Abstract
Myelin is essential for rapid nerve impulse propagation and axon protection. Accordingly, defects in myelination or myelin maintenance lead to secondary axonal damage and subsequent degeneration. Studies utilizing genetic (CNPase-, MAG-, and PLP-null mice) and naturally occurring neuropathy models suggest that myelinating glia also support axons independently from myelin. Myelin protein zero (MPZ or P0), which is expressed only by Schwann cells, is critical for myelin formation and maintenance in the peripheral nervous system. Many mutations in MPZ are associated with demyelinating neuropathies (Charcot-Marie-Tooth disease type 1B [CMT1B]). Surprisingly, the substitution of threonine by methionine at position 124 of P0 (P0T124M) causes axonal neuropathy (CMT2J) with little to no myelin damage. This disease provides an excellent paradigm to understand how myelinating glia support axons independently from myelin. To study this, we generated targeted knock-in MpzT124M mutant mice, a genetically authentic model of T124M-CMT2J neuropathy. Similar to patients, these mice develop axonopathy between 2 and 12 months of age, characterized by impaired motor performance, normal nerve conduction velocities but reduced compound motor action potential amplitudes, and axonal damage with only minor compact myelin modifications. Mechanistically, we detected metabolic changes that could lead to axonal degeneration, and prominent alterations in non-compact myelin domains such as paranodes, Schmidt-Lanterman incisures, and gap junctions, implicated in Schwann cell-axon communication and axonal metabolic support. Finally, we document perturbed mitochondrial size and distribution along MpzT124M axons suggesting altered axonal transport. Our data suggest that Schwann cells in P0T124M mutant mice cannot provide axons with sufficient trophic support, leading to reduced ATP biosynthesis and axonopathy. In conclusion, the MpzT124M mouse model faithfully reproduces the human neuropathy and represents a unique tool for identifying the molecular basis for glial support of axons.
Collapse
Affiliation(s)
- Ghjuvan’Ghjacumu Shackleford
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Leandro N. Marziali
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Yo Sasaki
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, Missouri, United States of America
| | - Anke Claessens
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Ferri
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Nadav I. Weinstock
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Alexander M. Rossor
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Nicholas J. Silvestri
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Emma R. Wilson
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Edward Hurley
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Grahame J. Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Senthilvelan Manohar
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Dalian Ding
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Richard J. Salvi
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - M. Laura Feltri
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Maurizio D’Antonio
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Lawrence Wrabetz
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| |
Collapse
|
30
|
Marmolejo-Garza A, Medeiros-Furquim T, Rao R, Eggen BJL, Boddeke E, Dolga AM. Transcriptomic and epigenomic landscapes of Alzheimer's disease evidence mitochondrial-related pathways. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119326. [PMID: 35839870 DOI: 10.1016/j.bbamcr.2022.119326] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 02/06/2023]
Abstract
Alzheimers disease (AD) is the main cause of dementia and it is defined by cognitive decline coupled to extracellular deposit of amyloid-beta protein and intracellular hyperphosphorylation of tau protein. Historically, efforts to target such hallmarks have failed in numerous clinical trials. In addition to these hallmark-targeted approaches, several clinical trials focus on other AD pathological processes, such as inflammation, mitochondrial dysfunction, and oxidative stress. Mitochondria and mitochondrial-related mechanisms have become an attractive target for disease-modifying strategies, as mitochondrial dysfunction prior to clinical onset has been widely described in AD patients and AD animal models. Mitochondrial function relies on both the nuclear and mitochondrial genome. Findings from omics technologies have shed light on AD pathophysiology at different levels (e.g., epigenome, transcriptome and proteome). Most of these studies have focused on the nuclear-encoded components. The first part of this review provides an updated overview of the mechanisms that regulate mitochondrial gene expression and function. The second part of this review focuses on evidence of mitochondrial dysfunction in AD. We have focused on published findings and datasets that study AD. We analyzed published data and provide examples for mitochondrial-related pathways. These pathways are strikingly dysregulated in AD neurons and glia in sex-, cell- and disease stage-specific manners. Analysis of mitochondrial omics data highlights the involvement of mitochondria in AD, providing a rationale for further disease modeling and drug targeting.
Collapse
Affiliation(s)
- Alejandro Marmolejo-Garza
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands; Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tiago Medeiros-Furquim
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands; Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ramya Rao
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Erik Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen N, Denmark.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands.
| |
Collapse
|
31
|
Afridi R, Rahman MH, Suk K. Implications of glial metabolic dysregulation in the pathophysiology of neurodegenerative diseases. Neurobiol Dis 2022; 174:105874. [PMID: 36154877 DOI: 10.1016/j.nbd.2022.105874] [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: 12/13/2021] [Revised: 08/28/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Glial cells are the most abundant cells of the brain, outnumbering neurons. These multifunctional cells are crucial for maintaining brain homeostasis by providing trophic and nutritional support to neurons, sculpting synapses, and providing an immune defense. Glia are highly plastic and undergo both structural and functional alterations in response to changes in the brain microenvironment. Glial phenotypes are intimately regulated by underlying metabolic machinery, which dictates the effector functions of these cells. Altered brain energy metabolism and chronic neuroinflammation are common features of several neurodegenerative diseases. Microglia and astrocytes are the major glial cells fueling the ongoing neuroinflammatory process, exacerbating neurodegeneration. Distinct metabolic perturbations in microglia and astrocytes, including altered carbohydrate, lipid, and amino acid metabolism have been documented in neurodegenerative diseases. These disturbances aggravate the neurodegenerative process by potentiating the inflammatory activation of glial cells. This review covers the recent advances in the molecular aspects of glial metabolic changes in the pathophysiology of neurodegenerative diseases. Finally, we discuss studies exploiting glial metabolism as a potential therapeutic avenue in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
32
|
Harnessing conserved signaling and metabolic pathways to enhance the maturation of functional engineered tissues. NPJ Regen Med 2022; 7:44. [PMID: 36057642 PMCID: PMC9440900 DOI: 10.1038/s41536-022-00246-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
The development of induced-pluripotent stem cell (iPSC)-derived cell types offers promise for basic science, drug testing, disease modeling, personalized medicine, and translatable cell therapies across many tissue types. However, in practice many iPSC-derived cells have presented as immature in physiological function, and despite efforts to recapitulate adult maturity, most have yet to meet the necessary benchmarks for the intended tissues. Here, we summarize the available state of knowledge surrounding the physiological mechanisms underlying cell maturation in several key tissues. Common signaling consolidators, as well as potential synergies between critical signaling pathways are explored. Finally, current practices in physiologically relevant tissue engineering and experimental design are critically examined, with the goal of integrating greater decision paradigms and frameworks towards achieving efficient maturation strategies, which in turn may produce higher-valued iPSC-derived tissues.
Collapse
|
33
|
Song J, Nilsson G, Xu Y, Zelco A, Rocha-Ferreira E, Wang Y, Zhang X, Zhang S, Ek J, Hagberg H, Zhu C, Wang X. Temporal brain transcriptome analysis reveals key pathological events after germinal matrix hemorrhage in neonatal rats. J Cereb Blood Flow Metab 2022; 42:1632-1649. [PMID: 35491813 PMCID: PMC9441725 DOI: 10.1177/0271678x221098811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Germinal matrix hemorrhage (GMH) is a common complication in preterm infants and is associated with high risk of adverse neurodevelopmental outcomes. We used a rat GMH model and performed RNA sequencing to investigate the signaling pathways and biological processes following hemorrhage. GMH induced brain injury characterized by early hematoma and subsequent tissue loss. At 6 hours after GMH, gene expression indicated an increase in mitochondrial activity such as ATP metabolism and oxidative phosphorylation along with upregulation of cytoprotective pathways and heme metabolism. At 24 hours after GMH, the expression pattern suggested an increase in cell cycle progression and downregulation of neurodevelopmental-related pathways. At 72 hours after GMH, there was an increase in genes related to inflammation and an upregulation of ferroptosis. Hemoglobin components and genes related to heme metabolism and ferroptosis such as Hmox1, Alox15, and Alas2 were among the most upregulated genes. We observed dysregulation of processes involved in development, mitochondrial function, cholesterol biosynthesis, and inflammation, all of which contribute to neurodevelopmental deterioration following GMH. This study is the first temporal transcriptome profile providing a comprehensive overview of the molecular mechanisms underlying brain injury following GMH, and it provides useful guidance in the search for therapeutic interventions.
Collapse
Affiliation(s)
- Juan Song
- Centre for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Gisela Nilsson
- Centre for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Aura Zelco
- Centre for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Yafeng Wang
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Joakim Ek
- Centre for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China.,Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Xiaoyang Wang
- Centre for Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China.,Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
34
|
Signorelli CM, Boils JD, Tagliazucchi E, Jarraya B, Deco G. From Brain-Body Function to Conscious Interactions. Neurosci Biobehav Rev 2022; 141:104833. [PMID: 36037978 DOI: 10.1016/j.neubiorev.2022.104833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/06/2022] [Accepted: 08/18/2022] [Indexed: 11/15/2022]
Abstract
In this review, we discuss empirical results inspiring the introduction of a formal mathematical multilayer model for the biological neuroscience of conscious experience. First, we motivate the discussion through evidence regarding the dynamic brain. Second, we review different brain-body couplings associated with conscious experience and its potential role in driving brain dynamics. Third, we introduce the machinery of multilayer networks to account for several types of interactions in brain-body systems. Then, a multilayer structure consists of two main generalizations: a formal semantic to study biological systems, and an integrative account for several signatures and models of consciousness. Finally, under this framework, we define composition of layers to account for entangled features of brain-body systems related to conscious experience. As such, a multilayer mathematical framework is highly integrative and thus may be more complete than other models. In this short review, we discuss a variety of empirical results inspiring the introduction of a formal mathematical multilayer model for the biological neuroscience of conscious experience.
Collapse
Affiliation(s)
- Camilo Miguel Signorelli
- Department of Computer Science, University of Oxford, Oxford, 7 Parks Rd, OxfordOX1 3QG, United Kingdom; Physiology of Cognition, GIGA-CRC In Vivo Imaging, Allée du 6 Août, 8 (B30), 4000 Sart Tilman, University of Liège, Belgium; Cognitive Neuroimaging Unit, INSERM, CEA, CNRS, Université Paris-Saclay, NeuroSpin center, 91191 Gif/Yvette, France; Computational Neuroscience Group, Center for Brain and Cognition, Universitat Pompeu Fabra, Barcelona, Spain.
| | - Joaquín Díaz Boils
- Universidad Internacional de La Rioja, Avda La Paz, 137, Logroño, La Rioja, Spain
| | - Enzo Tagliazucchi
- Physics Department, University of Buenos Aires, Buenos Aires, Argentina
| | - Bechir Jarraya
- Cognitive Neuroimaging Unit, INSERM, CEA, CNRS, Université Paris-Saclay, NeuroSpin center, 91191 Gif/Yvette, France
| | - Gustavo Deco
- Computational Neuroscience Group, Center for Brain and Cognition, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
35
|
Monocarboxylate transporters (MCTs) in skeletal muscle and hypothalamus of less or more physically active mice exposed to aerobic training. Life Sci 2022; 307:120872. [PMID: 35948119 DOI: 10.1016/j.lfs.2022.120872] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 11/22/2022]
Abstract
AIMS The synthesis of monocarboxylate transporters (MCTs) can be stimulated by aerobic training, but few is known about this effect associated or not with non-voluntary daily activities. We examined the effect of eight weeks of aerobic training in MCTs on the skeletal muscle and hypothalamus of less or more physically active mice, which can be achieved by keeping them in two different housing models, a small cage (SC) and a large cage (LC). MAIN METHODS Forty male C57BL/6J mice were divided into four groups. In each housing condition, mice were divided into untrained (N) and trained (T). For 8 weeks, the trained animals ran on a treadmill with an intensity equivalent to 80 % of the individual critical velocity (CV), considered aerobic capacity, 40 min/day, 5 times/week. Protein expression of MCTs was determined with fluorescence Western Blot. KEY FINDINGS T groups had higher hypothalamic MCT2 than N groups (ANOVA, P = 0.032). Significant correlations were detected between hypothalamic MCT2 and CV. There was a difference between the SC and LC groups in relation to MCT4 in the hypothalamus (LC > SC, P = 0.044). Trained mice housed in LC (but not SC-T) exhibited a reduction in MCT4 muscle (P < 0.001). SIGNIFICANCE Our findings indicate that aerobically trained mice increased the expression of MCT2 protein in the hypothalamus, which has been related to the uptake of lactate in neurons. Changes in energy metabolism in physically active mice (kept in LC) may be related to upregulation of hypothalamic MCT4, probably participating in the regulation of satiety.
Collapse
|
36
|
Comparative Metabolomics Study of the Impact of Articaine and Lidocaine on the Metabolism of SH-SY5Y Neuronal Cells. Metabolites 2022; 12:metabo12070581. [PMID: 35888705 PMCID: PMC9323911 DOI: 10.3390/metabo12070581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/06/2023] Open
Abstract
Articaine (ATC) and lidocaine (LDC) are the local anesthetics (LAs) currently most employed in dentistry. Cases of paresthesia, reported more frequently for ATC, have raised concerns about their potential neurotoxicity, calling for further investigation of their biological effects in neuronal cells. In this work, the impact of ATC and LDC on the metabolism of SH-SY5Y cells was investigated through 1H NMR metabolomics. For each LA, in vitro cultured cells were exposed to concentrations causing 10 and 50% reductions in cell viability, and their metabolic intracellular and extracellular profiles were characterized. Most effects were common to ATC and LDC, although with varying magnitudes. The metabolic variations elicited by the two LAs suggested (i) downregulation of glycolysis and of glucose-dependent pathways (e.g., one-carbon metabolism and hexosamine biosynthetic pathway), (ii) disturbance of branched chain amino acids (BCAA) catabolism, (iii) downregulation of TCA cycle anaplerotic fueling and activation of alternative energy producing pathways, (iv) interference with choline metabolism and (v) lipid droplet build-up. Interestingly, LDC had a greater impact on membrane phospholipid turnover, as suggested by higher phosphatidylcholine to phosphocholine conversion. Moreover, LDC elicited an increase in triglycerides, whereas cholesteryl esters accumulated in ATC-exposed cells, suggesting a different composition and handling of lipid droplets.
Collapse
|
37
|
Stanescu S, Bravo-Alonso I, Belanger-Quintana A, Pérez B, Medina-Diaz M, Ruiz-Sala P, Flores NP, Buenache R, Arrieta F, Rodríguez-Pombo P. Mitochondrial bioenergetic is impaired in Monocarboxylate transporter 1 deficiency: a new clinical case and review of the literature. Orphanet J Rare Dis 2022; 17:243. [PMID: 35729663 PMCID: PMC9215049 DOI: 10.1186/s13023-022-02389-4] [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: 03/03/2022] [Accepted: 06/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background Monocarboxylate transporter 1 (MCT1) deficiency has recently been described as a rare cause of recurrent ketosis, the result of impaired ketone utilization in extrahepatic tissues. To date, only six patients with this condition have been identified, and clinical and biochemical details remain incomplete. Results The present work reports a patient suffering from severe, recurrent episodes of metabolic acidosis and psychomotor delay, showing a pathogenic loss-of-function variation c.747_750del in homozygosity in SLC16A1 (which codes for MCT1). Persistent ketotic and lactic acidosis was accompanied by an abnormal excretion of organic acids related to redox balance disturbances. Together with an altered bioenergetic profile detected in patient-derived fibroblasts, this suggests possible mitochondrial dysfunction. Brain MRI revealed extensive, diffuse bilateral, symmetric signal alterations for the subcortical white matter and basal ganglia, together with corpus callosum agenesia. Conclusions These findings suggest that the clinical spectrum of MCT1 deficiency not only involves recurrent atacks of ketoacidosis, but may also cause lactic acidosis and neuromotor delay with a distinctive neuroimaging pattern including agenesis of corpus callosum and other brain signal alterations. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02389-4.
Collapse
Affiliation(s)
- Sinziana Stanescu
- Unidad de Enfermedades Metabólicas, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain.
| | - Irene Bravo-Alonso
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular Severo Ochoa, UAM-CSIC, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| | - Amaya Belanger-Quintana
- Unidad de Enfermedades Metabólicas, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Belen Pérez
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular Severo Ochoa, UAM-CSIC, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| | - Montserrat Medina-Diaz
- Department of Neuroradiology, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Pedro Ruiz-Sala
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular, Universidad Autónoma de Madrid, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| | - Nathaly Paola Flores
- Paediatric Department, Hospital General La Mancha Centro, Av. Constitución, 3, 13600, Alcázar de San Juan, Ciudad Real, Spain
| | - Raquel Buenache
- Neuropediatric Department, Hospital Universitario Ramón y Cajal, IRYCIS, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Francisco Arrieta
- Unidad de Enfermedades Metabólicas, Hospital Universitario Ramón y Cajal, IRYCIS, CIBER-OBN, Crta de Colmenar Viejo, km 9,100, 28034, Madrid, Spain
| | - Pilar Rodríguez-Pombo
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular Severo Ochoa, UAM-CSIC, CIBERER, IdiPAZ, C/Francisco Tomás y Valiente, 7, 28049, Madrid, Spain
| |
Collapse
|
38
|
Maly IV, Hofmann WA, Pletnikov MV. Experimental and computational analyses of calcium dynamics in 22q11.2 deletion model astrocytes. Neurosci Lett 2022; 783:136711. [PMID: 35671915 DOI: 10.1016/j.neulet.2022.136711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/31/2022] [Accepted: 06/02/2022] [Indexed: 12/29/2022]
Abstract
Methods for deriving mechanistic information from intracellular calcium dynamics have largely been applied to neuronal data despite the knowledge of roles of glial cells in behavior, cognition, and psychiatric disorders. Using calcium imaging, computer vision, and Bayesian kinetic inference (BKI), we analyzed calcium dynamics in primary astrocytes derived from control or Df1/+ mice, a model of 22q11.2 deletion (DiGeorge syndrome). Inference of the highest-likelihood molecular kinetic characteristics of intracellular calcium dynamics identified changes in the activity of the sarcoendoplasmic reticulum calcium ATPase (SERCA). Application of a SERCA inhibitor to wild-type astrocytes reproduced the differences detected in Df1/+ astrocytes. Our work reveals the molecular changes driving the calcium kinetics in astrocytes from a 22q11.2 deletion model. BKI can be useful for mechanistically dissecting calcium dynamics in glial cells and formulating and testing hypotheses about underlying molecular mechanisms.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Wilma A Hofmann
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21225, USA.
| |
Collapse
|
39
|
High-resolution NMR metabolomics of patients with subjective cognitive decline plus: Perturbations in the metabolism of glucose and branched-chain amino acids. Neurobiol Dis 2022; 171:105782. [DOI: 10.1016/j.nbd.2022.105782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 11/20/2022] Open
|
40
|
Giglia G, Agliani G, Lepri E, Baldoni E, Gobbi M, Ceccherelli R, Gröne A, van den Brand JMA, Mandara MT. Neuronal satellitosis is a common finding in the avian brain. Avian Pathol 2022; 51:381-387. [PMID: 35503252 DOI: 10.1080/03079457.2022.2073193] [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: 10/18/2022]
Abstract
AbstractPerineuronal or neuronal satellitosis is the term describing the presence of glial cells in the satellite space surrounding the neuronal perikaryon. Confusingly, this finding has been described both as a physiologic and pathologic condition in humans and animals. In animals, neuronal satellitosis has been described in mammals, as well as in avian species. For the latter, authors wondered whether this finding can be expressed in the normal telencephalon of different avian orders and families and whether this pattern in different species shows a specific brain-region association. For these aims, this study explored the presence of neuronal satellitosis in the major areas of the healthy telencephalon in wild avian species of different orders and families, evaluating its grade in different brain regions. Neuronal satellitosis was seen in the Hyperpallium and Mesopallium as areas with the highest grade. Passeriformes showed the highest grade of neuronal satellitosis compared to Diurnal, Nocturnal raptors, and Charadriiformes. To clarify the exact role of neuronal satellitosis in animals without neurological disease further studies are needed.
Collapse
Affiliation(s)
- Giuseppe Giglia
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo, 4, 06126 Perugia, Italy.,Division of Pathology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Gianfilippo Agliani
- Division of Pathology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Elvio Lepri
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo, 4, 06126 Perugia, Italy
| | - Erika Baldoni
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo, 4, 06126 Perugia, Italy
| | - Marco Gobbi
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo, 4, 06126 Perugia, Italy.,Diagnostic Department, Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche, Via Gaetano Salvemini, 1, 06126 Perugia, Italy
| | - Renato Ceccherelli
- Centro Recupero Uccelli Marini e Acquatici (CRUMA), Via delle Sorgenti, 430, 57121 Livorno, Italy
| | - Andrea Gröne
- Division of Pathology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Judith M A van den Brand
- Division of Pathology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Maria Teresa Mandara
- Department of Veterinary Medicine, University of Perugia, Via San Costanzo, 4, 06126 Perugia, Italy
| |
Collapse
|
41
|
Inflammation: A New Look at an Old Problem. Int J Mol Sci 2022; 23:ijms23094596. [PMID: 35562986 PMCID: PMC9100490 DOI: 10.3390/ijms23094596] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 02/07/2023] Open
Abstract
Pro-inflammatory stress is inherent in any cells that are subject to damage or threat of damage. It is defined by a number of universal components, including oxidative stress, cellular response to DNA damage, unfolded protein response to mitochondrial and endoplasmic reticulum stress, changes in autophagy, inflammasome formation, non-coding RNA response, formation of an inducible network of signaling pathways, and epigenetic changes. The presence of an inducible receptor and secretory phenotype in many cells is the cause of tissue pro-inflammatory stress. The key phenomenon determining the occurrence of a classical inflammatory focus is the microvascular inflammatory response (exudation, leukocyte migration to the alteration zone). This same reaction at the systemic level leads to the development of life-critical systemic inflammation. From this standpoint, we can characterize the common mechanisms of pathologies that differ in their clinical appearance. The division of inflammation into alternative variants has deep evolutionary roots. Evolutionary aspects of inflammation are also described in the review. The aim of the review is to provide theoretical arguments for the need for an up-to-date theory of the relationship between key human pathological processes based on the integrative role of the molecular mechanisms of cellular and tissue pro-inflammatory stress.
Collapse
|
42
|
Aoyama BB, Zanetti GG, Dias EV, Athié MCP, Lopes-Cendes I, Schwambach Vieira A. Transcriptomic analysis of dorsal and ventral subiculum after induction of acute seizures by electric stimulation of the perforant pathway in rats. Hippocampus 2022; 32:436-448. [PMID: 35343006 DOI: 10.1002/hipo.23417] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 11/09/2022]
Abstract
Preconditioning is a mechanism in which injuries induced by non-lethal hypoxia or seizures trigger cellular resistance to subsequent events. Norwood et al., in a 2010 study, showed that an 8-h-long period of electrical stimulation of the perforant pathway in rats is required for the induction of hippocampal sclerosis. However, in order to avoid generalized seizures, status epilepticus (SE), and death, a state of resistance to seizures must be induced in the hippocampus by a preconditioning paradigm consisting of two daily 30-min stimulation periods. Due to the importance of the subiculum in the hippocampal formation, this study aims to investigate differential gene expression patterns in the dorsal and ventral subiculum using RNA-sequencing, after induction of a preconditioning protocol by electrical stimulation of the perforant pathway. The dorsal (dSub) and ventral (vSub) subiculum regions were collected by laser-microdissection 24 h after preconditioning protocol induction in rats. RNA sequencing was performed in a Hiseq 4000 platform, reads were aligned using the STAR and DESEq2 statistics package was used to estimate gene expression. We identified 1176 differentially expressed genes comparing control to preconditioned subiculum regions, 204 genes were differentially expressed in dSub and 972 in vSub. The gene ontology enrichment analysis showed that the most significant common enrichment pathway considering up-regulated genes in dSub and vSub was steroid metabolism. In contrast, the most significant enrichment pathway considering down-regulated genes in vSub was axon guidance. Our results indicate that preconditioning induces changes in the expression of genes related to synaptic reorganization, increased cholesterol metabolism, and astrogliosis in both dSub and vSub. Both regions also presented a decrease in the expression of genes related to glutamatergic transmission and an increase in expression of genes related to complement system activation and GABAergic transmission. The down-regulation of proapoptotic and axon guidance genes in the ventral subiculum suggests that preconditioning may induce a neuroprotective environment in this region.
Collapse
Affiliation(s)
- Beatriz B Aoyama
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Gabriel G Zanetti
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Elayne V Dias
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Maria C P Athié
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Iscia Lopes-Cendes
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil.,Department of Translational Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - André Schwambach Vieira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| |
Collapse
|
43
|
Rudge JD. A New Hypothesis for Alzheimer's Disease: The Lipid Invasion Model. J Alzheimers Dis Rep 2022; 6:129-161. [PMID: 35530118 PMCID: PMC9028744 DOI: 10.3233/adr-210299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023] Open
Abstract
This paper proposes a new hypothesis for Alzheimer's disease (AD)-the lipid invasion model. It argues that AD results from external influx of free fatty acids (FFAs) and lipid-rich lipoproteins into the brain, following disruption of the blood-brain barrier (BBB). The lipid invasion model explains how the influx of albumin-bound FFAs via a disrupted BBB induces bioenergetic changes and oxidative stress, stimulates microglia-driven neuroinflammation, and causes anterograde amnesia. It also explains how the influx of external lipoproteins, which are much larger and more lipid-rich, especially more cholesterol-rich, than those normally present in the brain, causes endosomal-lysosomal abnormalities and overproduction of the peptide amyloid-β (Aβ). This leads to the formation of amyloid plaques and neurofibrillary tangles, the most well-known hallmarks of AD. The lipid invasion model argues that a key role of the BBB is protecting the brain from external lipid access. It shows how the BBB can be damaged by excess Aβ, as well as by most other known risk factors for AD, including aging, apolipoprotein E4 (APOE4), and lifestyle factors such as hypertension, smoking, obesity, diabetes, chronic sleep deprivation, stress, and head injury. The lipid invasion model gives a new rationale for what we already know about AD, explaining its many associated risk factors and neuropathologies, including some that are less well-accounted for in other explanations of AD. It offers new insights and suggests new ways to prevent, detect, and treat this destructive disease and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan D’Arcy Rudge
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
44
|
Ghosh C, Myers R, O'Connor C, Williams S, Liu X, Hossain M, Nemeth M, Najm IM. Cortical Dysplasia in Rats Provokes Neurovascular Alterations, GLUT1 Dysfunction, and Metabolic Disturbances That Are Sustained Post-Seizure Induction. Mol Neurobiol 2022; 59:2389-2406. [PMID: 35084654 PMCID: PMC9018620 DOI: 10.1007/s12035-021-02624-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/26/2021] [Indexed: 10/19/2022]
Abstract
Focal cortical dysplasia (FCD) is associated with blood-brain barrier (BBB) dysfunction in patients with difficult-to-treat epilepsy. However, the underlying cellular and molecular factors in cortical dysplasia (CD) associated with progressive neurovascular challenges during the pro-epileptic phase, post-seizure, and during epileptogenesis remain unclear. We studied the BBB function in a rat model of congenital (in utero radiation-induced, first hit) CD and longitudinally examined the cortical brain tissues at baseline and the progressive neurovascular alterations, glucose transporter-1 (GLUT1) expression, and glucose metabolic activity at 2, 15, and 30 days following a second hit using pentylenetetrazole-induced seizure. Our study revealed through immunoblotting, immunohistochemistry, and biochemical analysis that (1) altered vascular density and prolongation of BBB albumin leakages in CD rats continued through 30 days post-seizure; (2) CD brain tissues showed elevated matrix metalloproteinase-9 levels at 2 days post-seizure and microglial overactivation through 30 days post-seizure; (3) BBB tight junction protein and GLUT1 levels were decreased and neuronal monocarboxylate transporter-2 (MCT2) and mammalian target of rapamycin (mTOR) levels were increased in the CD rat brain: (4) ATPase activity is elevated and a low glucose/high lactate imbalance exists in CD rats; and (5) the mTOR pathway is activated and MCT2 levels are elevated in the presence of high lactate during glucose starvation in vitro. Together, this study suggests that BBB dysfunction, including decreased GLUT1 expression and metabolic disturbance, may contribute to epileptogenesis in this CD rat model through multiple mechanisms that could be translated to FCD therapy in medically refractory epilepsy.
Collapse
Affiliation(s)
- Chaitali Ghosh
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA. .,Department of Biomedical Engineering and Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Rosemary Myers
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Christina O'Connor
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sherice Williams
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Xuefeng Liu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mohammed Hossain
- Cerebrovascular Research, Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Michael Nemeth
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Imad M Najm
- Charles Shor Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
45
|
Lee JW, Profant M, Wang C. Metabolic Sex Dimorphism of the Brain at the Gene, Cell, and Tissue Level. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:212-220. [PMID: 35017210 DOI: 10.4049/jimmunol.2100853] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/09/2021] [Indexed: 12/21/2022]
Abstract
The palpable observation in the sex bias of disease prevalence in the CNS has fascinated scientists for several generations. Brain sex dimorphism has been visualized by imaging and analytical tools at the tissue, cellular, and molecular levels. Recent work highlighted the specificity of such sex bias in the brain and its subregions, offering a unique lens through which disease pathogenesis can be investigated. The brain is the largest consumer of energy in the body and provides a unique metabolic environment for diverse lineages of cells. Immune cells are increasingly recognized as an integral part of brain physiology, and their function depends on metabolic homeostasis. This review focuses on metabolic sex dimorphism in brain tissue, resident, and infiltrating immune cells. In this context, we highlight the relevance of recent advances in metabolomics and RNA sequencing technologies at the single cell resolution and the development of novel computational approaches.
Collapse
Affiliation(s)
- Jun Won Lee
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and
| | - Martin Profant
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Chao Wang
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; and .,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Lei H, Hu R, Luo G, Yang T, Shen H, Deng H, Chen C, Zhao H, Liu J. Altered Structural and Functional MRI Connectivity in Type 2 Diabetes Mellitus Related Cognitive Impairment: A Review. Front Hum Neurosci 2022; 15:755017. [PMID: 35069149 PMCID: PMC8770326 DOI: 10.3389/fnhum.2021.755017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/13/2021] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with cognitive impairment in many domains. There are several pieces of evidence that changes in neuronal neuropathies and metabolism have been observed in T2DM. Structural and functional MRI shows that abnormal connections and synchronization occur in T2DM brain circuits and related networks. Neuroplasticity and energy metabolism appear to be principal effector systems, which may be related to amyloid beta (Aβ) deposition, although there is no unified explanation that includes the complex etiology of T2DM with cognitive impairment. Herein, we assume that cognitive impairment in diabetes may lead to abnormalities in neuroplasticity and energy metabolism in the brain, and those reflected to MRI structural connectivity and functional connectivity, respectively.
Collapse
|
47
|
Xu Y, Gao G, Sun X, Liu Q, Li C. ATPase Inhibitory Factor 1 Is Critical for Regulating Sevoflurane-Induced Microglial Inflammatory Responses and Caspase-3 Activation. Front Cell Neurosci 2022; 15:770666. [PMID: 34975409 PMCID: PMC8714895 DOI: 10.3389/fncel.2021.770666] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/02/2021] [Indexed: 02/05/2023] Open
Abstract
Postoperative delirium (POD) is one of the most important complications after surgery with general anesthesia, for which the neurotoxicity of general anesthetics is a high-risk factor. However, the mechanism remains largely unknown, which also hinders the effective treatment of POD. Here, we confirmed that a clinical concentration of the general anesthetic sevoflurane increased the expression of inflammatory factors and activated the caspase-3 by upregulating ATPase inhibitory factor 1 (ATPIF1) expression in microglia. Upregulation of ATPIF1 decreased the synthesis of ATP which is an important signaling molecule secreted by microglia. Extracellular supplementation with ATP attenuated the microglial inflammatory response and caspase-3 activation caused by sevoflurane or overexpression of ATPIF1. Additionally, the microglial inflammatory response further upregulated ATPIF1 expression, resulting in a positive feedback loop. Animal experiments further indicated that intraperitoneal injection of ATP significantly alleviated sevoflurane anesthesia-induced POD-related anxiety behavior and memory damage in mice. This study reveals that ATPIF1, an important protein regulating ATP synthesis, mediates sevoflurane-induced neurotoxicity in microglia. ATP supplementation may be a potential clinical treatment to alleviate sevoflurane-induced POD.
Collapse
Affiliation(s)
- Yaru Xu
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China.,Department of Anesthesiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ge Gao
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoru Sun
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
| | - Qidong Liu
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cheng Li
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Clinical Research Center for Anesthesiology and Perioperative Medicine, Tongji University, Shanghai, China
| |
Collapse
|
48
|
da Silva Zandonadi F, dos Santos EAF, Marques MS, Sussulini A. Metabolomics: A Powerful Tool to Understand the Schizophrenia Biology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1400:105-119. [DOI: 10.1007/978-3-030-97182-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
49
|
Morales-Rosales SL, Santín-Márquez R, Posadas-Rodriguez P, Rincon-Heredia R, Montiel T, Librado-Osorio R, Luna-López A, Rivero-Segura NA, Torres C, Cano-Martínez A, Silva-Palacios A, Cortés-Hernández P, Morán J, Massieu L, Konigsberg M. Senescence in Primary Rat Astrocytes Induces Loss of the Mitochondrial Membrane Potential and Alters Mitochondrial Dynamics in Cortical Neurons. Front Aging Neurosci 2021; 13:766306. [PMID: 34924995 PMCID: PMC8672143 DOI: 10.3389/fnagi.2021.766306] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/27/2021] [Indexed: 01/10/2023] Open
Abstract
The decline in brain function during aging is one of the most critical health problems nowadays. Although senescent astrocytes have been found in old-age brains and neurodegenerative diseases, their impact on the function of other cerebral cell types is unknown. The aim of this study was to evaluate the effect of senescent astrocytes on the mitochondrial function of a neuron. In order to evaluate neuronal susceptibility to a long and constant senescence-associated secretory phenotype (SASP) exposure, we developed a model by using cellular cocultures in transwell plates. Rat primary cortical astrocytes were seeded in transwell inserts and induced to premature senescence with hydrogen peroxide [stress-induced premature senescence (SIPS)]. Independently, primary rat cortical neurons were seeded at the bottom of transwells. After neuronal 6 days in vitro (DIV), the inserts with SIPS-astrocytes were placed in the chamber and cocultured with neurons for 6 more days. The neuronal viability, the redox state [reduced glutathione/oxidized glutathione (GSH/GSSG)], the mitochondrial morphology, and the proteins and membrane potential were determined. Our results showed that the neuronal mitochondria functionality was altered after being cocultured with senescent astrocytes. In vivo, we found that old animals had diminished mitochondrial oxidative phosphorylation (OXPHOS) proteins, redox state, and senescence markers as compared to young rats, suggesting effects of the senescent astrocytes similar to the ones we observed in vitro. Overall, these results indicate that the microenvironment generated by senescent astrocytes can affect neuronal mitochondria and physiology.
Collapse
Affiliation(s)
- Sandra Lizbeth Morales-Rosales
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Roberto Santín-Márquez
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Pedro Posadas-Rodriguez
- Posgrado Biología Experimental, Universidad Autónoma Metropolitana, Mexico City, Mexico.,Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| | - Ruth Rincon-Heredia
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Teresa Montiel
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Raúl Librado-Osorio
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City, Mexico
| | - Armando Luna-López
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City, Mexico
| | | | - Claudio Torres
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Agustina Cano-Martínez
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Paulina Cortés-Hernández
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédica de Oriente, Atlixco, Mexico
| | - Julio Morán
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Lourdes Massieu
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mina Konigsberg
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Mexico City, Mexico
| |
Collapse
|
50
|
Maly IV, Morales MJ, Pletnikov MV. Astrocyte Bioenergetics and Major Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:173-227. [PMID: 34888836 DOI: 10.1007/978-3-030-77375-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ongoing research continues to add new elements to the emerging picture of involvement of astrocyte energy metabolism in the pathophysiology of major psychiatric disorders, including schizophrenia, mood disorders, and addictions. This review outlines what is known about the energy metabolism in astrocytes, the most numerous cell type in the brain, and summarizes the recent work on how specific perturbations of astrocyte bioenergetics may contribute to the neuropsychiatric conditions. The role of astrocyte energy metabolism in mental health and disease is reviewed on the organism, organ, and cell level. Data arising from genomic, metabolomic, in vitro, and neurobehavioral studies is critically analyzed to suggest future directions in research and possible metabolism-focused therapeutic interventions.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael J Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA.
| |
Collapse
|