1
|
Ye F, Wei C, Wu A. The potential mechanism of mitochondrial homeostasis in postoperative neurocognitive disorders: an in-depth review. Ann Med 2024; 56:2411012. [PMID: 39450938 PMCID: PMC11514427 DOI: 10.1080/07853890.2024.2411012] [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: 12/29/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 10/26/2024] Open
Abstract
Postoperative neurocognitive disorders (PND) are the most common neurological disorders following surgery and anaesthesia before and within 12 months after surgery, with a high prevalence in the geriatric population. PND can severely deteriorate the quality of life of patients, especially among the elderly, mainly manifested as memory loss, attention, decline and language comprehension disorders, mostly in elderly patients, with an incidence as high as 31%. Previous studies have also raised the possibility of accelerated cognitive decline and underlying neuropathological processes associated with diseases that affect cognitive performance (e.g. Alzheimer's dementia) for reasons related to anaesthesia and surgery. Currently, most research on PND has focused on various molecular pathways, especially in the geriatric population. The various hypotheses that have been proposed regarding the mechanisms imply peripheral neuroinflammation, oxidative stress, mitochondrial homeostasis, synaptic function, autophagy disorder, blood-brain barrier dysfunction, the microbiota-gut-brain axis and lack of neurotrophic support. However, the underlying pathogenesis and molecular mechanisms of PND have not yet been uncovered. Recent research has focused on mitochondrial homeostasis. In this paper, we present a review of various studies to better understand and characterize the mechanisms of associated cognitive dysfunction. As the biochemical basis of PND becomes more clearly defined, future treatments based on mitochondrial homeostasis modulation can prove to be very promising.
Collapse
Affiliation(s)
- Fan Ye
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Hao X, Li Y, Gao H, Wang Z, Fang B. Inhalation Anesthetics Play a Janus-Faced Role in Self-Renewal and Differentiation of Stem Cells. Biomolecules 2024; 14:1167. [PMID: 39334933 PMCID: PMC11430341 DOI: 10.3390/biom14091167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Inhalation anesthesia stands as a pivotal modality within clinical anesthesia practices. Beyond its primary anesthetic effects, inhaled anesthetics have non-anesthetic effects, exerting bidirectional influences on the physiological state of the body and disease progression. These effects encompass impaired cognitive function, inhibition of embryonic development, influence on tumor progression, and so forth. For many years, inhaled anesthetics were viewed as inhibitors of stem cell fate regulation. However, there is now a growing appreciation that inhaled anesthetics promote stem cell biological functions and thus are now regarded as a double-edged sword affecting stem cell fate. In this review, the effects of inhaled anesthetics on self-renewal and differentiation of neural stem cells (NSCs), embryonic stem cells (ESCs), and cancer stem cells (CSCs) were summarized. The mechanisms of inhaled anesthetics involving cell cycle, metabolism, stemness, and niche of stem cells were also discussed. A comprehensive understanding of these effects will enhance our comprehension of how inhaled anesthetics impact the human body, thus promising breakthroughs in the development of novel strategies for innovative stem cell therapy approaches.
Collapse
Affiliation(s)
- Xiaotong Hao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Hairong Gao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhilin Wang
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
3
|
Huuha AM, Norevik CS, Coombes JS, Røsbjørgen RN, Miguel-dos-Santos R, Moreira JBN, Kobro-Flatmoen A, Scrimgeour N, Tari AR. Effects of Intravenously Administered Plasma from Exercise-Trained Donors on Microglia and Cytokines in a Transgenic Rat Model of Alzheimer's Disease. Brain Plast 2024; 9:21-41. [PMID: 38993579 PMCID: PMC11234670 DOI: 10.3233/bpl-230154] [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] [Accepted: 03/26/2024] [Indexed: 07/13/2024] Open
Abstract
Background Microglia and inflammation play a significant role in Alzheimer's disease (AD). Physical exercise and peripheral signals can influence microglial activity in the brain. Modulating the inflammatory response in the brain may provide therapeutic approaches for AD. Objective To assess the effects of intravenously administered blood plasma from exercise-trained donor rats on cognitive function, microglia, and cytokine levels in an AD rat model at two different pathological stages; an early pre-plaque stage and a later stage closer to the emergence of extracellular plaques. Methods Male transgenic McGill-R-Thy1-APP rats aged 2 and 5 months received 14 injections over 6 weeks: 1) plasma from exercise-trained rats (ExPlas), 2) plasma from sedentary rats (SedPlas), or 3) saline. Cognitive function was evaluated in a novel object recognition task. Microglia count and morphology were analyzed in cornu ammonis, dentate gyrus, entorhinal cortex, and subiculum. Amyloid plaque number and size were assessed in the rats with the later treatment start. A multiplex assay was used to measure 23 cytokines in cornu ammonis. Results In rats treated from 2 months of age, ExPlas and SedPlas increased number and length of microglial branches in cornu ammonis and dentate gyrus compared to saline. Only ExPlas-treated rats exhibited similar changes in subiculum, while entorhinal cortex showed no differences across treatments. Microglia count remained unaffected. In rats treated from 5 months of age, there were no significant differences in microglia count or morphology or the number or size of amyloid plaques in any brain region. Compared to both other treatments in early pre-plaque stage rats, SedPlas increased TNF-α levels. ExPlas upregulated GM-CSF, IL-18, and VEGF, while SedPlas increased IL-10 compared to saline. In later-stage rats, ExPlas upregulated IL-17, and SedPlas upregulated TNF-α compared to saline. There were no effects of treatments on recognition memory. Conclusions Intravenous injections of blood plasma from exercise-trained and sedentary donors differentially modulated microglial morphology and cytokine levels in the AD rat model at an early pre-plaque stage of pathology. Exercised plasma may reduce proinflammatory TNF-α signaling and promote microglial responses to early Aβ accumulation but the lack of treatment effects in the later-stage rats emphasizes the potential importance of treatment timing.
Collapse
Affiliation(s)
- Aleksi M. Huuha
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cecilie Skarstad Norevik
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jeff S. Coombes
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Australia
| | - Ragnhild N. Røsbjørgen
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rodrigo Miguel-dos-Santos
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Physiology, Federal University of Sergipe, Sergipe, Brazil
| | - José Bianco N. Moreira
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Centre for Neural Computation, and Egil and Pauline Braathen and Fred Kavli Centre for Cortical Microcircuits, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Centre for Alzheimer’s Disease, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nathan Scrimgeour
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Atefe R. Tari
- Department of Circulation and Medical Imaging, Cardiac Exercise Research Group (CERG), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
4
|
Bettinetti-Luque M, Trujillo-Estrada L, Garcia-Fuentes E, Andreo-Lopez J, Sanchez-Varo R, Garrido-Sánchez L, Gómez-Mediavilla Á, López MG, Garcia-Caballero M, Gutierrez A, Baglietto-Vargas D. Adipose tissue as a therapeutic target for vascular damage in Alzheimer's disease. Br J Pharmacol 2024; 181:840-878. [PMID: 37706346 DOI: 10.1111/bph.16243] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023] Open
Abstract
Adipose tissue has recently been recognized as an important endocrine organ that plays a crucial role in energy metabolism and in the immune response in many metabolic tissues. With this regard, emerging evidence indicates that an important crosstalk exists between the adipose tissue and the brain. However, the contribution of adipose tissue to the development of age-related diseases, including Alzheimer's disease, remains poorly defined. New studies suggest that the adipose tissue modulates brain function through a range of endogenous biologically active factors known as adipokines, which can cross the blood-brain barrier to reach the target areas in the brain or to regulate the function of the blood-brain barrier. In this review, we discuss the effects of several adipokines on the physiology of the blood-brain barrier, their contribution to the development of Alzheimer's disease and their therapeutic potential. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Miriam Bettinetti-Luque
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Laura Trujillo-Estrada
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Garcia-Fuentes
- Unidad de Gestión Clínica Aparato Digestivo, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
- CIBER de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Juana Andreo-Lopez
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Raquel Sanchez-Varo
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Lourdes Garrido-Sánchez
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
| | - Ángela Gómez-Mediavilla
- Departamento de Farmacología, Facultad de Medicina. Instituto Teófilo Hernando para la I+D de Fármacos, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología, Facultad de Medicina. Instituto Teófilo Hernando para la I+D de Fármacos, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Sanitarias (IIS-IP), Hospital Universitario de la Princesa, Madrid, Spain
| | - Melissa Garcia-Caballero
- Departamento de Biología Molecular y Bioquímica, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
| | - Antonia Gutierrez
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - David Baglietto-Vargas
- Departamento de Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain
- CIBER de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Wu S, Zhou Y. Monoclonal antibody precision therapy targeting inflammation for bipolar disorder: a narrative review. Ther Adv Psychopharmacol 2024; 14:20451253241227772. [PMID: 38322010 PMCID: PMC10846009 DOI: 10.1177/20451253241227772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/07/2024] [Indexed: 02/08/2024] Open
Abstract
Bipolar disorder (BD) is a severe mental disorder with various hypotheses regarding its pathogenesis. This article provides a summary of numerous studies on the variations in inflammatory cytokine levels in patients with BD and the effects of treatment with antipsychotics, mood stabilizers, and antidepressants on these levels. In addition, patients with autoimmune diseases who use anti-inflammatory monoclonal antibodies experience symptoms, such as depression, anxiety, and insomnia. These pieces of evidence suggest a potential association between immune inflammation and BD and offer new possibilities for therapy. Building upon this relationship, the authors propose an innovative approach for treating BD through individualized and precise therapy using anti-inflammatory monoclonal antibody drugs. To support this proposal, the authors compile information on pharmacological effects and relevant studies, including trials of various anti-inflammatory therapeutic monoclonal antibody drugs (e.g. infliximab, tocilizumab, and canakinumab) for the potential treatment of BD and its associated side effects in psychiatry. The authors categorize these anti-inflammatory monoclonal antibody drugs into levels I-IV through a comprehensive analysis of their advantages and disadvantages. Their potential is examined, and the need for further exploration of their pharmaceutical effects is established.
Collapse
Affiliation(s)
- Shijin Wu
- School of Clinical Pharmacy (School of integrative Pharmacy), Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuyang Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, 280 Waihuan East Road, Higher Education Mega Center, Panyu, Guangzhou, Guangdong 510006, China
| |
Collapse
|
6
|
Bai X, Wang B, Cui Y, Tian S, Zhang Y, You L, Chang YZ, Gao G. Hepcidin deficiency impairs hippocampal neurogenesis and mediates brain atrophy and memory decline in mice. J Neuroinflammation 2024; 21:15. [PMID: 38195497 PMCID: PMC10777572 DOI: 10.1186/s12974-023-03008-0] [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: 10/04/2023] [Accepted: 12/27/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Hepcidin is the master regulator of iron homeostasis. Hepcidin downregulation has been demonstrated in the brains of Alzheimer's disease (AD) patients. However, the mechanism underlying the role of hepcidin downregulation in cognitive impairment has not been elucidated. METHODS In the present study, we generated GFAP-Cre-mediated hepcidin conditional knockout mice (HampGFAP cKO) to explore the effect of hepcidin deficiency on hippocampal structure and neurocognition. RESULTS We found that the HampGFAP cKO mice developed AD-like brain atrophy and memory deficits. In particular, the weight of the hippocampus and the number of granule neurons in the dentate gyrus were significantly reduced. Further investigation demonstrated that the morphological change in the hippocampus of HampGFAP cKO mice was attributed to impaired neurogenesis caused by decreased proliferation of neural stem cells. Regarding the molecular mechanism, increased iron content after depletion of hepcidin followed by an elevated level of the inflammatory factor tumor necrosis factor-α accounted for the impairment of hippocampal neurogenesis in HampGFAP cKO mice. These observations were further verified in GFAP promoter-driven hepcidin knockdown mice and in Nestin-Cre-mediated hepcidin conditional knockout mice. CONCLUSIONS The present findings demonstrated a critical role for hepcidin in hippocampal neurogenesis and validated the importance of iron and associated inflammatory cytokines as key modulators of neurodevelopment, providing insights into the potential pathogenesis of cognitive dysfunction and related treatments.
Collapse
Affiliation(s)
- Xue Bai
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Bing Wang
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Yiduo Cui
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Siqi Tian
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Yi Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Linhao You
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China.
| | - Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular BiologyHebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Sciences, Hebei Normal University, Shijiazhuang, 050024, Hebei, China.
| |
Collapse
|
7
|
Wang Q, Zhong Y, Chen N, Chen J. From the immune system to mood disorders especially induced by Toxoplasma gondii: CD4+ T cell as a bridge. Front Cell Infect Microbiol 2023; 13:1078984. [PMID: 37077528 PMCID: PMC10106765 DOI: 10.3389/fcimb.2023.1078984] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
Toxoplasma gondii (T. gondii), a ubiquitous and obligatory intracellular protozoa, not only alters peripheral immune status, but crosses the blood-brain barrier to trigger brain parenchymal injury and central neuroinflammation to establish latent cerebral infection in humans and other vertebrates. Recent findings underscore the strong correlation between alterations in the peripheral and central immune environment and mood disorders. Th17 and Th1 cells are important pro-inflammatory cells that can drive the pathology of mood disorders by promoting neuroinflammation. As opposed to Th17 and Th1, regulatory T cells have inhibitory inflammatory and neuroprotective functions that can ameliorate mood disorders. T. gondii induces neuroinflammation, which can be mediated by CD4+ T cells (such as Tregs, Th17, Th1, and Th2). Though the pathophysiology and treatment of mood disorder have been currently studied, emerging evidence points to unique role of CD4+ T cells in mood disorder, especially those caused by T. gondii infection. In this review, we explore some recent studies that extend our understanding of the relationship between mood disorders and T. gondii.
Collapse
|
8
|
The Dialogue Between Neuroinflammation and Adult Neurogenesis: Mechanisms Involved and Alterations in Neurological Diseases. Mol Neurobiol 2023; 60:923-959. [PMID: 36383328 DOI: 10.1007/s12035-022-03102-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022]
Abstract
Adult neurogenesis occurs mainly in the subgranular zone of the hippocampal dentate gyrus and the subventricular zone of the lateral ventricles. Evidence supports the critical role of adult neurogenesis in various conditions, including cognitive dysfunction, Alzheimer's disease (AD), and Parkinson's disease (PD). Several factors can alter adult neurogenesis, including genetic, epigenetic, age, physical activity, diet, sleep status, sex hormones, and central nervous system (CNS) disorders, exerting either pro-neurogenic or anti-neurogenic effects. Compelling evidence suggests that any insult or injury to the CNS, such as traumatic brain injury (TBI), infectious diseases, or neurodegenerative disorders, can provoke an inflammatory response in the CNS. This inflammation could either promote or inhibit neurogenesis, depending on various factors, such as chronicity and severity of the inflammation and underlying neurological disorders. Notably, neuroinflammation, driven by different immune components such as activated glia, cytokines, chemokines, and reactive oxygen species, can regulate every step of adult neurogenesis, including cell proliferation, differentiation, migration, survival of newborn neurons, maturation, synaptogenesis, and neuritogenesis. Therefore, this review aims to present recent findings regarding the effects of various components of the immune system on adult neurogenesis and to provide a better understanding of the role of neuroinflammation and neurogenesis in the context of neurological disorders, including AD, PD, ischemic stroke (IS), seizure/epilepsy, TBI, sleep deprivation, cognitive impairment, and anxiety- and depressive-like behaviors. For each disorder, some of the most recent therapeutic candidates, such as curcumin, ginseng, astragaloside, boswellic acids, andrographolide, caffeine, royal jelly, estrogen, metformin, and minocycline, have been discussed based on the available preclinical and clinical evidence.
Collapse
|
9
|
Artık A, Öztelcan Gündüz B, Mızrak S, Işık Ü. Increased serum levels of tumour necrosis factor-like weak inducer of apoptosis in children with autism spectrum disorder. INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2022; 69:611-616. [PMID: 37346259 PMCID: PMC10281418 DOI: 10.1080/20473869.2022.2143033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/10/2022] [Accepted: 10/30/2022] [Indexed: 06/23/2023]
Abstract
Introduction: A previous study has evaluated the association between serum tumour necrosis factor-like weak apoptosis inducer (TWEAK) levels and autism spectrum disorder (ASD). In line with this investigation, the present study aimed to measure serum TWEAK levels to determine whether their eventual alteration might have etiopathogenetic significance in children with ASD. Methods: A total of 40 treatment-naive children with ASD and 40 healthy children as controls were included in the present study. The Schedule for Affective Disorders and Schizophrenia for School-Aged Children-Present and Lifetime Version, DSM-5 was used by a psychiatrist to screen the healthy controls for psychiatric disorders after a physical examination by a paediatrician. The clinical severity of the ASD symptoms was assessed by the Childhood Autism Rating Scale. Venous blood samples were collected, and serum TWEAK levels were measured. Results: This study included 80 children in total, with 40 (50.0%) in the patient group and 40 (50.0%) in the healthy control group. Thirty four (85.0%) of the participants in the patient group, and 31 (77.5%) in the healthy control group, were male, and the remainder were female. The distribution of the gender ratio was statistically similar between groups (p = 0.568). The volunteers were between 36 and 59 months old. The average age in the patient group was 46.0 ± 6.5, while that in the healthy control group was 45.2 ± 6.7. The ages were also statistically similar between groups (p = 0.615). The TWEAK values of the patient group were found to be statistically higher than those of the healthy control group (p < 0.001). Discussion: This study examined whether serum TWEAK levels were related to ASD in childhood. Our findings indicate that children with ASD have higher TWEAK levels when compared to other children. The findings further indicate that serum TWEAK levels could be related to ASD etiopathogenesis independent of ASD symptom severity.
Collapse
Affiliation(s)
- Abdülbaki Artık
- Faculty of Medicine, Child and Adolescent Mental Health Department, Uşak University, Ankara, Turkey
| | | | - Soycan Mızrak
- Faculty of Medicine, Department of Medical Biochemistry, Uşak University, Ankara, Turkey
| | - Ümit Işık
- Faculty of Medicine, Child and Adolescent Mental Health Department, Süleyman Demirel University, Ankara, Turkey
| |
Collapse
|
10
|
Imam B, Rahmatinia M, Shahsavani A, Khodagholi F, Hopke PK, Bazazzpour S, Hadei M, Yarahmadi M, Abdollahifar MA, Torkmahalleh MA, Kermani M, Ilkhani S, MirBehbahani SH. Autism-like symptoms by exposure to air pollution and valproic acid-induced in male rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:59263-59286. [PMID: 35384534 DOI: 10.1007/s11356-022-19865-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
Exposure to air pollution during prenatal or neonatal periods is associated with autism spectrum disorder (ASD) according to epidemiology studies. Furthermore, prenatal exposure to valproic acid (VPA) has also been found to be associated with an increased prevalence of ASD. To assess the association between simultaneous exposure to VPA and air pollutants, seven exposure groups of rats were included in current study (PM2.5 and gaseous pollutants exposed - high dose of VPA (PGE-high); PM2.5 and gaseous pollutants exposed - low dose of VPA (PGE-low); gaseous pollutants only exposed - high dose of VPA (GE-high); gaseous pollutants only exposed - low dose of VPA (GE-low); clean air exposed - high dose of VPA (CAE-high); clean air exposed - low dose of VPA (CAE-low) and clean air exposed (CAE)). The pollution-exposed rats were exposed to air pollutants from embryonic day (E0) to postnatal day 42 (PND42). In all the induced groups, decreased oxidative stress biomarkers, decreased oxytocin receptor (OXTR) levels, and increased the expression of interleukin 6 (IL-6), interleukin 1β (IL-1β), and tumor necrosis factor alpha (TNF-α) were found. The volumes of the cerebellum, hippocampus, striatum, and prefrontal decreased in all induced groups in comparison to CAE. Additionally, increased numerical density of glial cells and decreased of numerical density of neurons were found in all induced groups. Results show that simultaneous exposure to air pollution and VPA can cause ASD-related behavioral deficits and air pollution reinforced the mechanism of inducing ASD ̉s in VPA-induced rat model of autism.
Collapse
Affiliation(s)
- Bahran Imam
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rahmatinia
- Student Research Committee, Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Shahsavani
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Air Quality and Climate Change Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Philip K Hopke
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
- Institute for a Sustainable Environment, Clarkson University, Potsdam, NY, 13699, USA
| | - Shahriyar Bazazzpour
- Student Research Committee, Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Hadei
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Yarahmadi
- Environmental and Occupational Health Center, Ministry of Health and Medical Education, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Amouei Torkmahalleh
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan, 010000
| | - Majid Kermani
- Department of Environmental Health Engineering, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Saba Ilkhani
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
11
|
Zuo C, Ma J, Pan Y, Zheng D, Chen C, Ruan N, Su Y, Nan H, Lian Q, Lin H. Isoflurane and Sevoflurane Induce Cognitive Impairment in Neonatal Rats by Inhibiting Neural Stem Cell Development Through Microglial Activation, Neuroinflammation, and Suppression of VEGFR2 Signaling Pathway. Neurotox Res 2022; 40:775-790. [PMID: 35471722 PMCID: PMC9098611 DOI: 10.1007/s12640-022-00511-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/26/2022] [Accepted: 04/12/2022] [Indexed: 12/30/2022]
Abstract
Inhaled anesthetics are known to induce neurotoxicity in the developing brains of rodents, although the mechanisms are not well understood. The aim of this study was to elucidate the molecular mechanisms underlying anesthetics-induced neurodevelopmental toxicity by VEGF receptor 2 (VEGFR2) through the interaction between microglia and neural stem cells (NSCs) in postnatal day 7 (P7) rats. Cognitive function of P7 rats exposed to isoflurane and sevoflurane were assessed using Morris Water Maze and T maze tests. We also evaluated the expression levels of NSC biomarkers (Nestin and Sox2), microglia biomarker (CD11b or or IBA1), pro-inflammatory cytokines (IL-6 and TNF-α), and VEGFR2 using western blotting and immunohistochemistry in the brains of control and anesthesia-treated rats. We found spatial learning and working memory was impaired 2 weeks after anesthetics exposure in rats. Isoflurane induced stronger and more prolonged neurotoxicity than sevoflurane. However, cognitive functions were recovered 6 weeks after anesthesia. Isoflurane and sevoflurane decreased the levels of Nestin, Sox2, and p-VEGFR2, activated microglia, decreased the number of NSCs and reduced neurogenesis and the proliferation of NSCs, and increased the levels of IL-6, TNF-α, and CD11b. Our results suggested that isoflurane and sevoflurane induced cognitive impairment in rats by inhibiting NSC development and neurogenesis via microglial activation, neuroinflammation, and suppression of VEGFR2 signaling pathway.
Collapse
Affiliation(s)
- Chunlong Zuo
- Department of Anesthesiology, The First Affiliated Hospital of AnHui Medical University, Hefei, 230022, PRC, China.,Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PRC, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PRC, China
| | - Junmei Ma
- Department of Anesthesiology, Ningbo Medical Center Lihuili Hospital, Ningbo, 315040, PRC, China
| | - Yizhao Pan
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Shangcaicun, Wenzhou, 325000, PRC, China
| | - Dongxu Zheng
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PRC, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PRC, China
| | - Chunjiang Chen
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PRC, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PRC, China
| | - Naqi Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PRC, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PRC, China
| | - Ying Su
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PRC, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PRC, China
| | - Haihan Nan
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325035, PRC, China
| | - Qingquan Lian
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PRC, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PRC, China.
| | - Han Lin
- Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PRC, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, PRC, China.
| |
Collapse
|
12
|
Early Life Events and Maturation of the Dentate Gyrus: Implications for Neurons and Glial Cells. Int J Mol Sci 2022; 23:ijms23084261. [PMID: 35457079 PMCID: PMC9031216 DOI: 10.3390/ijms23084261] [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: 02/27/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
The dentate gyrus (DG), an important part of the hippocampus, plays a significant role in learning, memory, and emotional behavior. Factors potentially influencing normal development of neurons and glial cells in the DG during its maturation can exert long-lasting effects on brain functions. Early life stress may modify maturation of the DG and induce lifelong alterations in its structure and functioning, underlying brain pathologies in adults. In this paper, maturation of neurons and glial cells (microglia and astrocytes) and the effects of early life events on maturation processes in the DG have been comprehensively reviewed. Early postnatal interventions affecting the DG eventually result in an altered number of granule neurons in the DG, ectopic location of neurons and changes in adult neurogenesis. Adverse events in early life provoke proinflammatory changes in hippocampal glia at cellular and molecular levels immediately after stress exposure. Later, the cellular changes may disappear, though alterations in gene expression pattern persist. Additional stressful events later in life contribute to manifestation of glial changes and behavioral deficits. Alterations in the maturation of neuronal and glial cells induced by early life stress are interdependent and influence the development of neural nets, thus predisposing the brain to the development of cognitive and psychiatric disorders.
Collapse
|
13
|
Chronic IL-10 overproduction disrupts microglia-neuron dialogue similar to aging, resulting in impaired hippocampal neurogenesis and spatial memory. Brain Behav Immun 2022; 101:231-245. [PMID: 34990747 DOI: 10.1016/j.bbi.2021.12.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 11/21/2022] Open
Abstract
The subgranular zone of the dentate gyrus is an adult neurogenic niche where new neurons are continuously generated. A dramatic hippocampal neurogenesis decline occurs with increasing age, contributing to cognitive deficits. The process of neurogenesis is intimately regulated by the microenvironment, with inflammation being considered a strong negative factor for this process. Thus, we hypothesize that the reduction of new neurons in the aged brain could be attributed to the age-related microenvironmental changes towards a pro-inflammatory status. In this work, we evaluated whether an anti-inflammatory microenvironment could counteract the negative effect of age on promoting new hippocampal neurons. Surprisingly, our results show that transgenic animals chronically overexpressing IL-10 by astrocytes present a decreased hippocampal neurogenesis in adulthood. This results from an impairment in the survival of neural newborn cells without differences in cell proliferation. In parallel, hippocampal-dependent spatial learning and memory processes were affected by IL-10 overproduction as assessed by the Morris water maze test. Microglial cells, which are key players in the neurogenesis process, presented a different phenotype in transgenic animals characterized by high activation together with alterations in receptors involved in neuronal communication, such as CD200R and CX3CR1. Interestingly, the changes described in adult transgenic animals were similar to those observed by the effect of normal aging. Thus, our data suggest that chronic IL-10 overproduction mimics the physiological age-related disruption of the microglia-neuron dialogue, resulting in hippocampal neurogenesis decrease and spatial memory impairment.
Collapse
|
14
|
Tumor Necrosis Factor-α: The Next Marker of Stroke. DISEASE MARKERS 2022; 2022:2395269. [PMID: 35265224 PMCID: PMC8898850 DOI: 10.1155/2022/2395269] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/05/2022] [Accepted: 02/19/2022] [Indexed: 02/06/2023]
Abstract
Although there is no shortage of research on the markers for stroke, to our knowledge, there are no clear markers that can meet the needs of clinical prediction and treatment. The inflammatory cascade is a critical process that persists and functions throughout the stroke process, ultimately worsening stroke outcomes and increasing mortality. Numerous inflammatory factors, including tumor necrosis factor (TNF), are involved in this process. These inflammatory factors play a dual role during stroke, and their mechanisms are complex. As one of the representatives, TNF is the primary regulator of the immune system and plays an essential role in the spread of inflammation. In researches done over the last few years, tumor necrosis factor-alpha (TNF-α) has emerged as a potential marker for stroke because of its essential role in stroke. This review summarizes the latest research on TNF-α in stroke and explores its potential as a therapeutic target.
Collapse
|
15
|
Experimental Arthritis Inhibits Adult Hippocampal Neurogenesis in Mice. Cells 2022; 11:cells11050791. [PMID: 35269413 PMCID: PMC8909078 DOI: 10.3390/cells11050791] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Background: Adult-born neurons of the hippocampal dentate gyrus play a role in specific forms of learning, and disturbed neurogenesis seems to contribute to the development of neuropsychiatric disorders, such as major depression. Neuroinflammation inhibits adult neurogenesis, but the effect of peripheral inflammation on this form of neuroplasticity is ambiguous. Objective: Our aim was to investigate the influence of acute and chronic experimental arthritis on adult hippocampal neurogenesis and to elucidate putative regulatory mechanisms. Methods: Arthritis was triggered by subcutaneous injection of complete Freund’s adjuvant (CFA) into the hind paws of adult male mice. The animals were killed either seven days (acute inflammation) or 21 days (chronic inflammation) after the CFA injection. Behavioral tests were used to demonstrate arthritis-related hypersensitivity to painful stimuli. We used in vivo bioluminescence imaging to verify local inflammation. The systemic inflammatory response was assessed by complete blood cell counts and by measurement of the cytokine/chemokine concentrations of TNF-α, IL-1α, IL-4, IL-6, IL-10, KC and MIP-2 in the inflamed hind limbs, peripheral blood and hippocampus to characterize the inflammatory responses in the periphery and in the brain. In the hippocampal dentate gyrus, the total number of newborn neurons was determined with quantitative immunohistochemistry visualizing BrdU- and doublecortin-positive cells. Microglial activation in the dentate gyrus was determined by quantifying the density of Iba1- and CD68-positive cells. Results: Both acute and chronic arthritis resulted in paw edema, mechanical and thermal hyperalgesia. We found phagocytic infiltration and increased levels of TNF-α, IL-4, IL-6, KC and MIP-2 in the inflamed hind paws. Circulating neutrophil granulocytes and IL-6 levels increased in the blood solely during the acute phase. In the dentate gyrus, chronic arthritis reduced the number of doublecortin-positive cells, and we found increased density of CD68-positive macrophages/microglia in both the acute and chronic phases. Cytokine levels, however, were not altered in the hippocampus. Conclusions: Our data suggest that acute peripheral inflammation initiates a cascade of molecular and cellular changes that eventually leads to reduced adult hippocampal neurogenesis, which was detectable only in the chronic inflammatory phase.
Collapse
|
16
|
Craig CF, Filippone RT, Stavely R, Bornstein JC, Apostolopoulos V, Nurgali K. Neuroinflammation as an etiological trigger for depression comorbid with inflammatory bowel disease. J Neuroinflammation 2022; 19:4. [PMID: 34983592 PMCID: PMC8729103 DOI: 10.1186/s12974-021-02354-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with inflammatory bowel disease (IBD) suffer from depression at higher rates than the general population. An etiological trigger of depressive symptoms is theorised to be inflammation within the central nervous system. It is believed that heightened intestinal inflammation and dysfunction of the enteric nervous system (ENS) contribute to impaired intestinal permeability, which facilitates the translocation of intestinal enterotoxins into the blood circulation. Consequently, these may compromise the immunological and physiological functioning of distant non-intestinal tissues such as the brain. In vivo models of colitis provide evidence of increased blood–brain barrier permeability and enhanced central nervous system (CNS) immune activity triggered by intestinal enterotoxins and blood-borne inflammatory mediators. Understanding the immunological, physiological, and structural changes associated with IBD and neuroinflammation may aid in the development of more tailored and suitable pharmaceutical treatment for IBD-associated depression.
Collapse
Affiliation(s)
- Colin F Craig
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhiannon T Filippone
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Joel C Bornstein
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Vasso Apostolopoulos
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia.,Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Heath and Sport, Victoria University, Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, VIC, Australia. .,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia. .,Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia. .,Institute for Health and Sport, Victoria University, Level 4 Research Labs, Western Centre for Health Research and Education, Sunshine Hospital, 176 Furlong Road, St Albans, VIC, 3021, Australia.
| |
Collapse
|
17
|
Sodhi K, Pratt R, Wang X, Lakhani HV, Pillai SS, Zehra M, Wang J, Grover L, Henderson B, Denvir J, Liu J, Pierre S, Nelson T, Shapiro JI. Role of adipocyte Na,K-ATPase oxidant amplification loop in cognitive decline and neurodegeneration. iScience 2021; 24:103262. [PMID: 34755095 PMCID: PMC8564125 DOI: 10.1016/j.isci.2021.103262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 11/21/2022] Open
Abstract
Recent studies suggest that a western diet may contribute to clinical neurodegeneration and dementia. Adipocyte-specific expression of the Na,K-ATPase signaling antagonist, NaKtide, ameliorates the pathophysiological consequences of murine experimental obesity and renal failure. In this study, we found that a western diet produced systemic oxidant stress along with evidence of activation of Na,K-ATPase signaling within both murine brain and peripheral tissues. We also noted this diet caused increases in circulating inflammatory cytokines as well as behavioral, and brain biochemical changes consistent with neurodegeneration. Adipocyte specific NaKtide affected by a doxycycline on/off expression system ameliorated all of these diet effects. These data suggest that a western diet produces cognitive decline and neurodegeneration through augmented Na,K-ATPase signaling and that antagonism of this pathway in adipocytes ameliorates the pathophysiology. If this observation is confirmed in humans, the adipocyte Na,K-ATPase may serve as a clinical target in the therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Komal Sodhi
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Rebecca Pratt
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Xiaoliang Wang
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Hari Vishal Lakhani
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Sneha S. Pillai
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Mishghan Zehra
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jiayan Wang
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Lawrence Grover
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Brandon Henderson
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - James Denvir
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Jiang Liu
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Sandrine Pierre
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Thomas Nelson
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Joseph I. Shapiro
- Departments of Medicine, Surgery, and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| |
Collapse
|
18
|
Masetto Antunes M, Godoy G, Masi LN, Curi R, Barbosa Bazotte R. Prefrontal Cortex and Hippocampus Inflammation in Mice Fed High-Carbohydrate or High-Fat Diets. J Med Food 2021; 25:110-113. [PMID: 34495750 DOI: 10.1089/jmf.2021.0026] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We previously reported that a high-carbohydrate diet (HCD) induced systemic inflammation and higher gene expression of proinflammatory mediators in the liver, skeletal muscle, and brain than a high-fat diet (HFD). However, the differences between the groups were less pronounced in the brain. In this study, we extended the evaluation of inflammation to specific areas of the brain. In this study, we evaluated the gene expression of caspase 2, caspase 3, caspase 9, cyclooxygenase-2 (Cox 2), inducible nitric oxide synthase (iNOS), interleukin (IL), IL-6, IL-1β, IL-10, IL-4, tumor necrosis factor-alpha (TNF-α), integrin subunit alpha m (Itgam), S100 protein (S100), allograft inflammatory factor 1 (Aif1), and glial fibrillary acidic protein (Gfap) in the prefrontal cortex and hippocampus of male Swiss mice that were fed with HCD or HFD for 8 weeks. The HCD group exhibited higher IL-1β expression, whereas the HFD group showed higher TNF-α expression in the prefrontal cortex. In the hippocampus, TNF-α expression was higher in the HFD group. IL-1β and TNF-α are proinflammatory cytokines that have been associated with impaired brain function and numerous brain disorders. Our results indicate that both HCD and HFD promote prefrontal cortex inflammation; however, the hippocampus seems more sensitive to a HFD than HCD.
Collapse
Affiliation(s)
- Marina Masetto Antunes
- Post-Graduation Program in Pharmaceutical Sciences, State University of Maringá, Maringá, Brazil
| | - Guilherme Godoy
- Post-Graduation Program in Pharmaceutical Sciences, State University of Maringá, Maringá, Brazil
| | - Laureane Nunes Masi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil and Immunobiological Production Section, Bioindustrial Center, Butantan Institute, São Paulo, Brazil
| | - Rui Curi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil and Immunobiological Production Section, Bioindustrial Center, Butantan Institute, São Paulo, Brazil
| | - Roberto Barbosa Bazotte
- Post-Graduation Program in Pharmaceutical Sciences, State University of Maringá, Maringá, Brazil.,Department of Pharmacology and Therapeutics, State University of Maringá, Maringá, Brazil
| |
Collapse
|
19
|
You K, Gu H, Yuan Z, Xu X. Tumor Necrosis Factor Alpha Signaling and Organogenesis. Front Cell Dev Biol 2021; 9:727075. [PMID: 34395451 PMCID: PMC8361451 DOI: 10.3389/fcell.2021.727075] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/08/2021] [Indexed: 01/04/2023] Open
Abstract
Tumor necrosis factor alpha (TNF-α) plays important roles in processes such as immunomodulation, fever, inflammatory response, inhibition of tumor formation, and inhibition of viral replication. TNF-α and its receptors are ubiquitously expressed in developing organs and they regulate the survival, proliferation, and apoptosis of embryonic stem cells (ESCs) and progenitor cells. TNF-α is an important inflammatory factor that also regulates the inflammatory response during organogenesis, and its cytotoxic effects can interfere with normal developmental processes, even leading to the onset of diseases. This review summarizes the various roles of TNF-α in organogenesis in terms of its secreting pattern, concentration-dependent activities, and interactions with other signaling pathways. We also explored new potential functions of TNF-α.
Collapse
Affiliation(s)
- Kai You
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuewen Xu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, China.,Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Koo JW, Wohleb ES. How Stress Shapes Neuroimmune Function: Implications for the Neurobiology of Psychiatric Disorders. Biol Psychiatry 2021; 90:74-84. [PMID: 33485589 PMCID: PMC8126571 DOI: 10.1016/j.biopsych.2020.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Chronic stress causes physiological and hormonal adaptations that lead to neurobiological consequences and behavioral and cognitive impairments. In particular, chronic stress has been shown to drive reduced neurogenesis and altered synaptic plasticity in brain regions that regulate mood and motivation. The neurobiological and behavioral effects of stress resemble the pathophysiology and symptoms observed in psychiatric disorders, suggesting that there are similar underlying mechanisms. Accumulating evidence indicates that neuroimmune systems, particularly microglia, have a critical role in regulating the neurobiology of stress. Preclinical models indicate that chronic stress provokes changes in microglia phenotype and increases inflammatory cytokine signaling, which affects neuronal function and leads to synaptic plasticity deficits and impaired neurogenesis. More recent work has shown that microglia can also phagocytose neuronal elements and contribute to structural remodeling of neurons in response to chronic stress. In this review we highlight work by the Duman research group (as well as others) that has revealed how chronic stress shapes neuroimmune function and, in turn, how inflammatory mediators and microglia contribute to the neurobiological effects of chronic stress. We also provide considerations to engage the therapeutic potential of neuroimmune systems, with the goal of improving treatment for psychiatric disorders.
Collapse
Affiliation(s)
- Ja Wook Koo
- Department of Neural Development and Disease, Korea Brain
Research Institute, Daegu, Korea,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk
Institute of Science and Technology (DGIST), Daegu, Korea
| | - Eric S. Wohleb
- Department of Pharmacology & Systems Physiology,
University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of
America,Corresponding author: Eric S. Wohleb, Department
of Pharmacology & Systems Physiology, University of Cincinnati College of
Medicine, 2120 East Galbraith Road, Cincinnati, OH 45237 U.S.A.,
| |
Collapse
|
21
|
Shigemoto-Mogami Y, Sato K. [Central Nervous System Developmental Regulation of Microglia via Cytokines and Chemokines]. YAKUGAKU ZASSHI 2021; 141:359-368. [PMID: 33642504 DOI: 10.1248/yakushi.20-00198-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Microglia are immune cells resident in the central nervous system (CNS). It has been gradually clarified that microglia play various roles at the developmental stage of the CNS. From embryonic to early postnatal age, microglia remove apoptotic cells by phagocytosis and refine the neural circuits by synaptic pruning. In addition, microglia promote the proliferation and differentiation of neural stem cells by releasing physiologically active substances. Our group has focused on the physiological actions of microglia via cytokines and chemokines at the early postnatal developmental stage. We found that a large number of activated microglia accumulate in the early postnatal subventricular zone (SVZ). We demonstrated that the these SVZ microglia facilitate neurogenesis and oligodendrogenesis via inflammatory cytokines including IL-1β, TNFα, IL-6, IFNγ. We have also found that microglia regulate the functional maturation of the blood brain barrier (BBB) and identified the cytokines and chemokines involved in the effects of microglia. These findings indicate that microglia are physiologically more important than ever thought to reveal robust brain functions. Furthermore, the new mode of microglial action may lead to the discovery of drug targets of the incurable CNS diseases.
Collapse
Affiliation(s)
| | - Kaoru Sato
- Division of Pharmacology, National Institute of Health Sciences
| |
Collapse
|
22
|
Rodina AV, Semochkina YP, Vysotskaya OV, Romantsova AN, Strepetov AN, Moskaleva EY. Low dose gamma irradiation pretreatment modulates the sensitivity of CNS to subsequent mixed gamma and neutron irradiation of the mouse head. Int J Radiat Biol 2021; 97:926-942. [PMID: 34043460 DOI: 10.1080/09553002.2021.1928787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 10/21/2022]
Abstract
ABSTRACТPurpose: To explore if the total body γ-irradiation at a dose of 0.1 Gy 7 days prior to acute mixed γ, n-irradiation of the head at the dose of 1 Gy can reduce the harmful effects of neutron irradiation on the hippocampal functions, neuroinflammation and neurogenesis.Materials and methods: Mice were exposed to γ-radiation alone, mixed γ,n-radiation or combined γ-rays and γ,n-radiation 7 days after γ-irradiation. Two months post-irradiation, mice were tested in Open Field and in the Morris water maze. The content of microglia, astrocytes, proliferating cells and cytokines TGF-β, TNF-α, IL-1β, GFAP levels, hippocampal BDNF, NT-3, NT-4, NGF mRNA expression were evaluated.Results: Two months after combined irradiation, we observed impaired hippocampus-dependent cognition, which was not detected in mice exposed to γ,n-irradiation. Combined exposure and γ,n-irradiation led to a significant increase in the level of activated microglia and astrocytes in the brains. The level of pro- and anti-inflammatory cytokines in the brain and hippocampal neurotrophine's genes changed differenly after the combined exposure and γ,n-irradiation. The quantity of DCX-positive cells was reduced after γ,n-irradiation exposer alone, but increased after combined irradiation.Conclusions: Our results indicate radio-adaptive responses in brains of mice that were exposed to low-dose gamma irradiation 7 days prior to acute 1 Gy γ,n-irradiation.
Collapse
Affiliation(s)
- Alla V Rodina
- Kurchatov Complex of NBICS Technologies, NRC Kurchatov Institute, Moscow, Russian Federation
- Chair of Biological Chemistry, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Yulia P Semochkina
- Kurchatov Complex of NBICS Technologies, NRC Kurchatov Institute, Moscow, Russian Federation
| | - Olga V Vysotskaya
- Kurchatov Complex of NBICS Technologies, NRC Kurchatov Institute, Moscow, Russian Federation
| | - Anastasia N Romantsova
- Kurchatov Complex of NBICS Technologies, NRC Kurchatov Institute, Moscow, Russian Federation
| | - Aleksandr N Strepetov
- Kurchatov Nuclear Physics Complex, NRC 'Kurchatov Institute', Moscow, Russian Federation
| | - Elizaveta Y Moskaleva
- Kurchatov Complex of NBICS Technologies, NRC Kurchatov Institute, Moscow, Russian Federation
| |
Collapse
|
23
|
Fu Y, Jia J. Isoliquiritigenin Confers Neuroprotection and Alleviates Amyloid-β42-Induced Neuroinflammation in Microglia by Regulating the Nrf2/NF-κB Signaling. Front Neurosci 2021; 15:638772. [PMID: 33642990 PMCID: PMC7904903 DOI: 10.3389/fnins.2021.638772] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Background Neuroinflammation and oxidative stress are two major pathological characteristics of Alzheimer's disease (AD). Amyloid-β oligomers (AβO), a toxic form of Aβ, promote the neuroinflammation and oxidative stress in the development of AD. Isoliquiritigenin (ISL), a natural flavonoid isolated from the root of liquorice, has been shown to exert inhibitory effects on inflammatory response and oxidative stress. Objectives The main purpose of this study is to assess the influence of ISL on inflammatory response and oxidative stress in BV2 cells stimulated with AβO, and to explore the underlying molecular mechanisms. Methods 3-(4,5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H- tetrazolium bromide (MTT) and lactate dehydrogenase (LDH) cytotoxicity assays were used to assess the toxic or protective effects of ISL. The expression levels of interleukin-1β, interleukin-6, and tumor necrosis factor-α were assessed by quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assays. Morphological changes in BV2 cells were assessed by immunofluorescence method. Nitric oxide (NO) assay kit was used to determinate the NO production. Western blot, qRT-PCR and immunofluorescence were used to explore the underlying molecular mechanisms. Results ISL treatment reduced the production of inflammatory cytokines and NO, and alleviated the morphological changes in BV2 cells induced by AβO. ISL treatment further protected N2a cells from the toxic medium of AβO-stimulated BV2 cells. ISL activated nuclear factor erythroid-2 related factor 2 (Nrf2) signaling and suppressed nuclear factor-κB (NF-κB) signaling in BV2 cells. Conclusion ISL suppresses AβO-induced inflammation and oxidative stress in BV2 cells via the regulation of Nrf2/NF-κB signaling. Therefore, ISL indirectly protects neurons from the damage of toxic conditioned media.
Collapse
Affiliation(s)
- Yue Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric Diseases, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| |
Collapse
|
24
|
Wen SJ, Zheng XM, Liu LF, Li NN, Mao HA, Huang L, Yuan QL. Effects of primary microglia and astrocytes on neural stem cells in in vitro and in vivo models of ischemic stroke. Neural Regen Res 2021; 16:1677-1685. [PMID: 33510055 PMCID: PMC8328755 DOI: 10.4103/1673-5374.306093] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Transplantation of neural stem cells (NSCs) can protect neurons in animal stroke models; however, their low rates of survival and neuronal differentiation limit their clinical application. Glial niches, an important location of neural stem cells, regulate survival, proliferation and differentiation of neural stem cells. However, the effects of activated glial cells on neural stem cells remain unclear. In the present study, we explored the effects of activated astrocytes and microglia on neural stem cells in vitro stroke models. We also investigated the effects of combined transplantation of neural stem cells and glial cells after stroke in rats. In a Transwell co-culture system, primary cultured astrocytes, microglia or mixed glial cells were exposed to glutamate or H2O2 and then seeded in the upper inserts, while primary neural stem cells were seeded in the lower uncoated wells and cultured for 7 days. Our results showed that microglia were conducive to neurosphere formation and had no effects on apoptosis within neurospheres, while astrocytes and mixed glial cells were conducive to neurosphere differentiation and reduced apoptosis within neurospheres, regardless of their pretreatment. In contrast, microglia and astrocytes induced neuronal differentiation of neural stem cells in differentiation medium, regardless of their pretreatment, with an exception of astrocytes pretreated with H2O2. Rat models of ischemic stroke were established by occlusion of the middle cerebral artery. Three days later, 5 × 105 neural stem cells with microglia or astrocytes were injected into the right lateral ventricle. Neural stem cell/astrocyte-treated rats displayed better improvement of neurological deficits than neural stem cell only-treated rats at 4 days after cell transplantation. Moreover, neural stem cell/microglia-, and neural stem cell/astrocyte-treated rats showed a significant decrease in ischemic volume compared with neural stem cell-treated rats. These findings indicate that microglia and astrocytes exert different effects on neural stem cells, and that co-transplantation of neural stem cells and astrocytes is more conducive to the recovery of neurological impairment in rats with ischemic stroke. The study was approved by the Animal Ethics Committee of Tongji University School of Medicine, China (approval No. 2010-TJAA08220401) in 2010.
Collapse
Affiliation(s)
- Sheng-Jun Wen
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xi-Min Zheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li-Fen Liu
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Na-Na Li
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hai-An Mao
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Huang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiong-Lan Yuan
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Alvarez Cooper I, Beecher K, Chehrehasa F, Belmer A, Bartlett SE. Tumour Necrosis Factor in Neuroplasticity, Neurogenesis and Alcohol Use Disorder. Brain Plast 2020; 6:47-66. [PMID: 33680846 PMCID: PMC7903009 DOI: 10.3233/bpl-190095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder is a pervasive and detrimental condition that involves changes in neuroplasticity and neurogenesis. Alcohol activates the neuroimmune system and alters the inflammatory status of the brain. Tumour necrosis factor (TNF) is a well characterised neuroimmune signal but its involvement in alcohol use disorder is unknown. In this review, we discuss the variable findings of TNF's effect on neuroplasticity and neurogenesis. Acute ethanol exposure reduces TNF release while chronic alcohol intake generally increases TNF levels. Evidence suggests TNF potentiates excitatory transmission, promotes anxiety during alcohol withdrawal and is involved in drug use in rodents. An association between craving for alcohol and TNF is apparent during withdrawal in humans. While anti-inflammatory therapies show efficacy in reversing neurogenic deficit after alcohol exposure, there is no evidence for TNF's essential involvement in alcohol's effect on neurogenesis. Overall, defining TNF's role in alcohol use disorder is complicated by poor understanding of its variable effects on synaptic transmission and neurogenesis. While TNF may be of relevance during withdrawal, the neuroimmune system likely acts through a larger group of inflammatory cytokines to alter neuroplasticity and neurogenesis. Understanding the individual relevance of TNF in alcohol use disorder awaits a more comprehensive understanding of TNF's effects within the brain.
Collapse
Affiliation(s)
- Ignatius Alvarez Cooper
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
| | - Kate Beecher
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
| | - Arnauld Belmer
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Selena E. Bartlett
- Institute of Health and Biomedical Innovation, Translational Research Institute, Brisbane, Australia
- School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
26
|
Atorvastatin ameliorates viral burden and neural stem/progenitor cell (NSPC) death in an experimental model of Japanese encephalitis. J Biosci 2020. [DOI: 10.1007/s12038-020-00052-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
27
|
Abd-Allah NA, Ibrahim OM, Elmalt HA, Shehata MA, Hamed RA, Elsaadouni NM, Hawary B. Thioredoxin level and inflammatory markers in children with autism spectrum disorders. MIDDLE EAST CURRENT PSYCHIATRY 2020. [DOI: 10.1186/s43045-020-00021-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Autism Spectrum Disorders (ASD) are a group of neurodevelopmental disabilities with unknown etiology. Recent studies suggest the contribution of immune dysfunction and oxidative stress in its pathophysiology. The present study aimed to investigate the serum level of thioredoxin (Trx), as a marker of oxidative stress and some inflammatory cytokines, and to evaluate their role in children with ASD.
Results
Concentrations of Trx, IL-1β, IL-8, and TNF-α were significantly higher in children with ASD compared with matched controls. There were no association between cytokine levels and the severity of clinical manifestations, according to CARS classification of severity.
Conclusion
The present study provides support for the idea that physiological abnormalities, such as oxidative stress and immune dysfunction, may contribute in the pathophysiology of ASD.
Collapse
|
28
|
Dietary Protein Source Influences Brain Inflammation and Memory in a Male Senescence-Accelerated Mouse Model of Dementia. Mol Neurobiol 2020; 58:1312-1329. [PMID: 33169333 DOI: 10.1007/s12035-020-02191-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/29/2020] [Indexed: 12/21/2022]
Abstract
Dementia is a pathological condition characterized by a decline in memory, as well as in other cognitive and social functions. The cellular and molecular mechanisms of brain damage in dementia are not completely understood; however, neuroinflammation is involved. Evidence suggests that chronic inflammation may impair cognitive performance and that dietary protein source may differentially influence this process. Dietary protein source has previously been shown to modify systemic inflammation in mouse models. Thus, we aimed to investigate the effect of chronic dietary protein source substitution in an ageing and dementia male mouse model, the senescence-accelerated mouse-prone 8 (SAMP8) model. We observed that dietary protein source differentially modified memory as shown by inhibitory avoidance testing at 4 months of age. Also, dietary protein source differentially modified neuroinflammation and gliosis in male SAMP8 mice. Our results suggest that chronic dietary protein source substitution may influence brain ageing and memory-related mechanisms in male SAMP8 mice. Moreover, the choice of dietary protein source in mouse diets for experimental purposes may need to be carefully considered when interpreting results.
Collapse
|
29
|
Araki T, Ikegaya Y, Koyama R. The effects of microglia‐ and astrocyte‐derived factors on neurogenesis in health and disease. Eur J Neurosci 2020; 54:5880-5901. [PMID: 32920880 PMCID: PMC8451940 DOI: 10.1111/ejn.14969] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022]
Abstract
Hippocampal neurogenesis continues throughout life and has been suggested to play an essential role in maintaining spatial cognitive function under physiological conditions. An increasing amount of evidence has indicated that adult neurogenesis is tightly controlled by environmental conditions in the neurogenic niche, which consists of multiple types of cells including microglia and astrocytes. Microglia maintain the environment of neurogenic niche through their phagocytic capacity and interaction with neurons via fractalkine‐CX3CR1 signaling. In addition, microglia release growth factors such as brain‐derived neurotrophic factor (BDNF) and cytokines such as tumor necrosis factor (TNF)‐α to support the development of adult born neurons. Astrocytes also manipulate neurogenesis by releasing various soluble factors including adenosine triphosphate and lactate. Whereas, under pathological conditions such as Alzheimer's disease, depression, and epilepsy, microglia and astrocytes play a leading role in inflammation and are involved in attenuating the normal process of neurogenesis. The modulation of glial functions on neurogenesis in these brain diseases are attracting attention as a new therapeutic target. This review describes how these glial cells play a role in adult hippocampal neurogenesis in both health and disease, especially focusing glia‐derived factors.
Collapse
Affiliation(s)
- Tasuku Araki
- Laboratory of Chemical Pharmacology Graduate School of Pharmaceutical Sciences The University of Tokyo Tokyo Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology Graduate School of Pharmaceutical Sciences The University of Tokyo Tokyo Japan
- Center for Information and Neural Networks Suita City Osaka Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology Graduate School of Pharmaceutical Sciences The University of Tokyo Tokyo Japan
| |
Collapse
|
30
|
Guarnieri G, Sarchielli E, Comeglio P, Herrera-Puerta E, Piaceri I, Nacmias B, Benelli M, Kelsey G, Maggi M, Gallina P, Vannelli GB, Morelli A. Tumor Necrosis Factor α Influences Phenotypic Plasticity and Promotes Epigenetic Changes in Human Basal Forebrain Cholinergic Neuroblasts. Int J Mol Sci 2020; 21:E6128. [PMID: 32854421 PMCID: PMC7504606 DOI: 10.3390/ijms21176128] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023] Open
Abstract
TNFα is the main proinflammatory cytokine implicated in the pathogenesis of neurodegenerative disorders, but it also modulates physiological functions in both the developing and adult brain. In this study, we investigated a potential direct role of TNFα in determining phenotypic changes of a recently established cellular model of human basal forebrain cholinergic neuroblasts isolated from the nucleus basalis of Meynert (hfNBMs). Exposing hfNBMs to TNFα reduced the expression of immature markers, such as nestin and β-tubulin III, and inhibited primary cilium formation. On the contrary, TNFα increased the expression of TNFα receptor TNFR2 and the mature neuron marker MAP2, also promoting neurite elongation. Moreover, TNFα affected nerve growth factor receptor expression. We also found that TNFα induced the expression of DNA-methylation enzymes and, accordingly, downregulated genes involved in neuronal development through epigenetic mechanisms, as demonstrated by methylome analysis. In summary, TNFα showed a dual role on hfNBMs phenotypic plasticity, exerting a negative influence on neurogenesis despite a positive effect on differentiation, through mechanisms that remain to be elucidated. Our results help to clarify the complexity of TNFα effects in human neurons and suggest that manipulation of TNFα signaling could provide a potential therapeutic approach against neurodegenerative disorders.
Collapse
Affiliation(s)
- Giulia Guarnieri
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| | - Erica Sarchielli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| | - Paolo Comeglio
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | | | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy; (I.P.); (B.N.)
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy; (I.P.); (B.N.)
| | - Matteo Benelli
- Bioinformatics Unit, Hospital of Prato, Azienda USL Toscana Centro, 50122 Prato, Italy;
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge CB22 3AT, UK;
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 1TN, UK
| | - Mario Maggi
- Endocrinology Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy;
| | - Pasquale Gallina
- Neurosurgical Unit, Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy;
| | - Gabriella Barbara Vannelli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| | - Annamaria Morelli
- Section of Human Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.S.); (G.B.V.)
| |
Collapse
|
31
|
Bonafina A, Paratcha G, Ledda F. Deciphering New Players in the Neurogenic Adult Hippocampal Niche. Front Cell Dev Biol 2020; 8:548. [PMID: 32714932 PMCID: PMC7346873 DOI: 10.3389/fcell.2020.00548] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/10/2020] [Indexed: 12/23/2022] Open
Abstract
In the mammalian adult hippocampus, new neurons are continuously generated throughout life in the subgranular zone of the dentate gyrus. Increasing evidence point out the contribution of adult-born hippocampal granule cells (GCs) to cognitive processes such as learning and memory, indicating the relevance of understanding the molecular mechanisms that control the development of these new neurons in the preexisting hippocampal circuits. Cell proliferation and functional integration of adult-born GCs is a process highly regulated by different intrinsic and extrinsic factors. In this review, we discuss recent advances related with cellular components and extrinsic signals of the hippocampal neurogenic niche that support and modulate neurogenesis under physiological conditions.
Collapse
Affiliation(s)
- Antonela Bonafina
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Gustavo Paratcha
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Fernanda Ledda
- Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
32
|
Barichello T, Giridharan VV, Bhatti G, Sayana P, Doifode T, Macedo D, Quevedo J. Inflammation as a Mechanism of Bipolar Disorder Neuroprogression. Curr Top Behav Neurosci 2020; 48:215-237. [PMID: 33040314 DOI: 10.1007/7854_2020_173] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bipolar disorder (BD) is a severe, debilitating psychiatric condition with onset in adolescence or young adulthood and often follows a relapsing and remitting course throughout life. The concept of neuroprogression in BD refers to the progressive path with an identifiable trajectory that takes place with recurrent mood episodes, which eventually leads to cognitive, functional, and clinical deterioration in the course of BD. Understanding the biological basis of neuroprogression helps to explain the subset of BD patients who experience worsening of their disorder over time. Additionally, the study of the neurobiological mechanisms underpinning neuroprogression will help BD staging based on systems biology. Replicated epidemiological studies have suggested inflammatory mechanisms as primary contributors to the neuroprogression of mood disorders. It is known that dysregulated inflammatory/immune pathways are often associated with BD pathophysiology. Hence, in this chapter, we focus on the evidence for the involvement of inflammation and immune regulated pathways in the neurobiological consequences of BD neuroprogression. Herein we put forth the evidence of immune markers from autoimmune disorders, chronic infections, and gut-brain axis that lead to BD neuroprogression. Further, we highlighted the peripheral and central inflammatory components measured along with BD progression.
Collapse
Affiliation(s)
- Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA. .,Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil. .,Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Vijayasree Vayalanellore Giridharan
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Gursimrat Bhatti
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Pavani Sayana
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Tejaswini Doifode
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research, and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirao Preto, Brazil
| | - Joao Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.,Laboratory of Neurosciences, Graduate Program in Health Sciences, University of Southern Santa Catarina-UNESC, Criciúma, SC, Brazil.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| |
Collapse
|
33
|
Yli-Karjanmaa M, Larsen KS, Fenger CD, Kristensen LK, Martin NA, Jensen PT, Breton A, Nathanson L, Nielsen PV, Lund MC, Carlsen SL, Gramsbergen JB, Finsen B, Stubbe J, Frich LH, Stolp H, Brambilla R, Anthony DC, Meyer M, Lambertsen KL. TNF deficiency causes alterations in the spatial organization of neurogenic zones and alters the number of microglia and neurons in the cerebral cortex. Brain Behav Immun 2019; 82:279-297. [PMID: 31505254 DOI: 10.1016/j.bbi.2019.08.195] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/21/2019] [Accepted: 08/29/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Although tumor necrosis factor (TNF) inhibitors are used to treat chronic inflammatory diseases, there is little information about how long-term inhibition of TNF affects the homeostatic functions that TNF maintains in the intact CNS. MATERIALS AND METHODS To assess whether developmental TNF deficiency causes alterations in the naïve CNS, we estimated the number of proliferating cells, microglia, and neurons in the developing neocortex of E13.5, P7 and adult TNF knock out (TNF-/-) mice and wildtype (WT) littermates. We also measured changes in gene and protein expression and monoamine levels in adult WT and TNF-/- mice. To evaluate long-term effects of TNF inhibitors, we treated healthy adult C57BL/6 mice with either saline, the selective soluble TNF inhibitor XPro1595, or the nonselective TNF inhibitor etanercept. We estimated changes in cell number and protein expression after two months of treatment. We assessed the effects of TNF deficiency on cognition by testing adult WT and TNF-/- mice and mice treated with saline, XPro1595, or etanercept with specific behavioral tasks. RESULTS TNF deficiency decreased the number of proliferating cells and microglia and increased the number of neurons. At the same time, TNF deficiency decreased the expression of WNT signaling-related proteins, specifically Collagen Triple Helix Repeat Containing 1 (CTHRC1) and Frizzled receptor 6 (FZD6). In contrast to XPro1595, long-term inhibition of TNF with etanercept in adult C57BL/6 mice decreased the number of BrdU+ cells in the granule cell layer of the dentate gyrus. Etanercept, but not XPro1595, also impaired spatial learning and memory in the Barnes maze memory test. CONCLUSION TNF deficiency impacts the organization of neurogenic zones and alters the cell composition in brain. Long-term inhibition of TNF with the nonselective TNF inhibitor etanercept, but not the soluble TNF inhibitor XPro1595, decreases neurogenesis in the adult mouse hippocampus and impairs learning and memory after two months of treatment.
Collapse
Affiliation(s)
- Minna Yli-Karjanmaa
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Kathrine Solevad Larsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Christina Dühring Fenger
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lotte Kellemann Kristensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nellie Anne Martin
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Peter Toft Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Lubov Nathanson
- Institute for Neuro Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Pernille Vinther Nielsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Minna Christiansen Lund
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Stephanie Lindeman Carlsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Jan Bert Gramsbergen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Bente Finsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jane Stubbe
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lars Henrik Frich
- Orthopedic Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Helen Stolp
- Department of Pharmacology, University of Oxford, Oxford, UK; Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Roberta Brambilla
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark; The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Daniel Clive Anthony
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Pharmacology, University of Oxford, Oxford, UK; BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Neurology, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
34
|
Hypertension and Its Impact on Stroke Recovery: From a Vascular to a Parenchymal Overview. Neural Plast 2019; 2019:6843895. [PMID: 31737062 PMCID: PMC6815533 DOI: 10.1155/2019/6843895] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Hypertension is the first modifiable vascular risk factor accounting for 10.4 million deaths worldwide; it is strongly and independently associated with the risk of stroke and is related to worse prognosis. In addition, hypertension seems to be a key player in the implementation of vascular cognitive impairment. Long-term hypertension, complicated or not by the occurrence of ischemic stroke, is often reviewed on its vascular side, and parenchymal consequences are put aside. Here, we sought to review the impact of isolated hypertension or hypertension associated to stroke on brain atrophy, neuron connectivity and neurogenesis, and phenotype modification of microglia and astrocytes. Finally, we discuss the impact of antihypertensive therapies on cell responses to hypertension and functional recovery. This attractive topic remains a focus of continued investigation and stresses the relevance of including this vascular risk factor in preclinical investigations of stroke outcome.
Collapse
|
35
|
Van der Veeken L, Van der Merwe J, Devroe S, Inversetti A, Galgano A, Bleeser T, Meeusen R, Rex S, Deprest J. Maternal surgery during pregnancy has a transient adverse effect on the developing fetal rabbit brain. Am J Obstet Gynecol 2019; 221:355.e1-355.e19. [PMID: 31336075 DOI: 10.1016/j.ajog.2019.07.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/22/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Recently, the US Food and Drug Administration called for cautious use of anesthetic drugs during pregnancy. In 0.2-2% of pregnancies, nonobstetric surgery is being performed. The consequences of anesthesia during pregnancy on fetal development remain unclear, and preclinical studies in relevant animal models may help to elucidate them. OBJECTIVE To assess the effect of maternal anesthesia and surgery during pregnancy on the developing fetal brain, using a rabbit model. MATERIALS AND METHODS This is a randomized, sham-controlled study in time-mated pregnant does at 28 days of gestation (term = 31 days), which corresponds to the end of the second trimester in humans. Anesthesia was induced in 14 does (155 pups) with propofol and maintained with 4 vol% (equivalent to 1 minimum alveolar concentration) sevoflurane for 2 hours, and a laparotomy with minimal organ manipulation was performed (surgery group). Maternal vital signs (blood pressure, heart rate, peripheral and cerebral oxygen saturation, temperature, end-tidal CO2, pH, lactate) were continuously monitored. Sham controls consisted of 7 does (74 pups) undergoing invasive hemodynamic monitoring for 2 hours without sedation. At term, does underwent cesarean delivery under ketamine-medetomidine sedation and local anesthesia. Pups either underwent motor and sensory neurologic testing followed by euthanasia at day 1 or daily neurodevelopment testing for 2 weeks and extensive neurologic assessment at 5 and 7 weeks (open field and object recognition test, T-maze, and radial-arm maze). Brains were harvested for histologic assessment of neuron density and synaptophysin expression. RESULTS Blood gases and vital parameters were stable in both groups. On postnatal day 1, surgery pups had significant lower motor (25 ± 1 vs 23 ± 3; P = .004) and sensory (16 ± 2 vs 15 ± 2; P = .005) neurobehavioral scores and lower brain-to-body weight ratios (3.7% ± 0.6% vs 3.4% ± 0.6%; P = .001). This was accompanied by lower neuron density in multiple brain regions (eg, hippocampus 2617 ± 410 vs 2053 ± 492 neurons/mm2; P = .004) with lower proliferation rates and less synaptophysin expression. Furthermore, surgery pups had delayed motor development during the first week of life, for example with hopping appearing later (6 ± 5 vs 12 ± 3 days; P = .011). Yet, by 7 weeks of age, neurobehavioral impairment was limited to a reduced digging behavior, and no differences in neuron density or synaptophysin expression were seen. CONCLUSION In rabbits, 2 hours of maternal general anesthesia and laparotomy, with minimal organ and no fetal manipulation, had a measurable impact on neonatal neurologic function and brain morphology. Pups had a slower motoric neurodevelopment, but by 7 weeks the effect became almost undetectable.
Collapse
Affiliation(s)
- Lennart Van der Veeken
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Belgium; Clinical Department Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Johannes Van der Merwe
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Belgium; Clinical Department Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Sarah Devroe
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium; Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Belgium
| | - Annalisa Inversetti
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Belgium
| | - Angela Galgano
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Belgium
| | - Tom Bleeser
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Belgium; Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Roselien Meeusen
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| | - Steffen Rex
- Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium; Department of Cardiovascular Sciences, Group Biomedical Sciences, KU Leuven, Belgium
| | - Jan Deprest
- Department of Development and Regeneration, Cluster Woman and Child, Group Biomedical Sciences, KU Leuven, Belgium; Clinical Department Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium; Institute for Women's Health, University College London, London, UK.
| |
Collapse
|
36
|
Tomiga Y, Yoshimura S, Ra SG, Takahashi Y, Goto R, Kugimoto I, Uehara Y, Kawanaka K, Higaki Y. Anxiety-like behaviors and hippocampal nNOS in response to diet-induced obesity combined with exercise. J Physiol Sci 2019; 69:711-722. [PMID: 31124076 PMCID: PMC10717450 DOI: 10.1007/s12576-019-00686-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/14/2019] [Indexed: 01/22/2023]
Abstract
A high-fat diet (HFD) and overweight status can induce hippocampal dysfunction, leading to depression and anxiety. Exercise has beneficial effects on emotional behaviors. We previously reported that exercise training rescues HFD-induced excess hippocampal neuronal nitric oxide synthase (nNOS) expression, which is a key regulator of anxiety. Here, we investigated anxiety-like behaviors and hippocampal nNOS expression in response to HFD combined with exercise. Mice were assigned to standard diet, HFD, or HFD with exercise groups for 12 weeks. We found that exercise during the final 6 weeks of the HFD regime improved 12 weeks of HFD-induced defecation, accompanied by rescue of excess nNOS expression. However, anxiety indicators in the elevated plus maze were unchanged. These effects were not apparent after only 1 week of exercise. In conclusion, 6 weeks of exercise training reduced HFD-related anxiety according to one of our measures (defecation), and reversed changes in the hippocampal nNOS/NO pathway.
Collapse
Affiliation(s)
- Yuki Tomiga
- Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Saki Yoshimura
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Song-Gyu Ra
- Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports and Health Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yuri Takahashi
- Faculty of Sports and Health Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Rina Goto
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Ikumi Kugimoto
- Graduate School of Sports and Health Science, Fukuoka University, Fukuoka, Japan
| | - Yoshinari Uehara
- Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports and Health Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Kentaro Kawanaka
- Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports and Health Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Yasuki Higaki
- Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.
- Faculty of Sports and Health Science, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| |
Collapse
|
37
|
Egorova E, Starinets A, Tyrtyshnaia A, Ponomarenko A, Manzhulo I. Hippocampal Neurogenesis in Conditions of Chronic Stress Induced by Sciatic Nerve Injury in the Rat. Cells Tissues Organs 2019; 207:58-68. [PMID: 31284284 DOI: 10.1159/000501236] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
The dentate gyrus of the hippocampus is the primary location of adult neurogenesis, which is affected by a variety of external and internal factors, including activity of surrounding glial cells. This study concerns alterations in hippocampal neurogenesis and changes in activity of both proinflammatory and neuroprotective microglia/macrophages after sciatic nerve injury in the rat. Here, we demonstrated that the chronic pain induced by a peripheral nerve injury manifests in the hippocampus by a decrease in proliferation (PCNA+) and neurogenesis (DCX+), an increase in proinflammatory cytokines (CD86+), and a reduction in neuroprotective (CD163+) microglia/macrophages. We suggest that a pathological increase microglia/macrophage activity is the cause of neurogenesis suppression observed in chronic neuropathic pain.
Collapse
Affiliation(s)
- Evgeniia Egorova
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation.,Far Eastern Federal University, Vladivostok, Russian Federation
| | - Anna Starinets
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation.,Far Eastern Federal University, Vladivostok, Russian Federation
| | - Anna Tyrtyshnaia
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation.,Far Eastern Federal University, Vladivostok, Russian Federation
| | - Arina Ponomarenko
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation.,Far Eastern Federal University, Vladivostok, Russian Federation
| | - Igor Manzhulo
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation, .,Far Eastern Federal University, Vladivostok, Russian Federation,
| |
Collapse
|
38
|
Inhibition of TNF-α-induced neuronal apoptosis by antidepressants acting through the lysophosphatidic acid receptor LPA1. Apoptosis 2019; 24:478-498. [DOI: 10.1007/s10495-019-01530-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Zhou Y, Peng W, Wang J, Zhou W, Zhou Y, Ying B. Plasma levels of IL-1Ra are associated with schizophrenia. Psychiatry Clin Neurosci 2019; 73:109-115. [PMID: 30375100 DOI: 10.1111/pcn.12794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/20/2018] [Accepted: 10/24/2018] [Indexed: 02/05/2023]
Abstract
AIM Although peripheral low-grade inflammation and brain-derived neurotrophic factor (BDNF) levels have been implicated in schizophrenia (SCZ), the interactions between them remain to be fully revealed. We aimed to compare BDNF and cytokines in patients with SCZ and healthy controls (HC). Additionally, we aimed to investigate the association between peripheral levels of cytokines and BDNF in patients with SCZ. METHODS Plasma levels of BDNF, interferon gamma, interleukin (IL)-10, IL-12, IL-1, IL-6, IL-8, tumor necrosis factor alpha, macrophage migration inhibitory factor, IL-1 receptor antagonist (IL-1Ra), and CD40 Ligand were compared in 45 SCZ patients and 38 HC using Luminex technology. RESULTS Compared to HC, patients had significantly higher IL-1Ra levels (P = 0.031). We found a strong positive association between BDNF and CD40 Ligand in the patient group (rho = 0.858, P < 0.001) as well as in the HC group (rho = 0.822, P < 0.001), respectively. Furthermore, there was a negative association between BDNF and tumor necrosis factor alpha in patients (rho = -0.429, P = 0.030) as well as in HC (rho = -0.649, P < 0.001). CONCLUSION These results suggest that the cytokine IL-1Ra may play a role in SCZ pathophysiology. Additionally, the interaction between cytokines and BDNF levels further indicated the diverse actions of these cytokines.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wu Peng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - WenJing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - YanHong Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - BinWu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
40
|
Cai B, Seong KJ, Bae SW, Kook MS, Chun C, Lee JH, Choi WS, Jung JY, Kim WJ. Water-Soluble Arginyl–Diosgenin Analog Attenuates Hippocampal Neurogenesis Impairment Through Blocking Microglial Activation Underlying NF-κB and JNK MAPK Signaling in Adult Mice Challenged by LPS. Mol Neurobiol 2019; 56:6218-6238. [DOI: 10.1007/s12035-019-1496-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022]
|
41
|
Gharbaran R, Somenarain L. Putative Cellular and Molecular Roles of Zika Virus in Fetal and Pediatric Neuropathologies. Pediatr Dev Pathol 2019; 22:5-21. [PMID: 30149771 DOI: 10.1177/1093526618790742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although the World Health Organization declared an end to the recent Zika virus (ZIKV) outbreak and its association with adverse fetal and pediatric outcome, on November 18, 2016, the virus still remains a severe public health threat. Laboratory experiments thus far supported the suspicions that ZIKV is a teratogenic agent. Evidence indicated that ZIKV infection cripples the host cells' innate immune responses, allowing productive replication and potential dissemination of the virus. In addition, studies suggest potential transplacental passage of the virus and subsequent selective targeting of neural progenitor cells (NPCs). Depletion of NPCs by ZIKV is associated with restricted brain growth. And while microcephaly can result from infection at any gestational stages, the risk is greater during the first trimester. Although a number of recent studies revealed some of specific molecular and cellular roles of ZIKV proteins of this mosquito-borne flavivirus, the mechanisms by which it produces it suspected pathophysiological effects are not completely understood. Thus, this review highlights the cellular and molecular evidence that implicate ZIKV in fetal and pediatric neuropathologies.
Collapse
Affiliation(s)
- Rajendra Gharbaran
- 1 Department of Biological Sciences, Bronx Community College, The City University of New York, Bronx, New York
| | - Latchman Somenarain
- 1 Department of Biological Sciences, Bronx Community College, The City University of New York, Bronx, New York
| |
Collapse
|
42
|
Netto MB, de Oliveira Junior AN, Goldim M, Mathias K, Fileti ME, da Rosa N, Laurentino AO, de Farias BX, Costa AB, Rezin GT, Fortunato JJ, Giustina AD, Barichello T, Dal-Pizzol F, Petronilho F. Oxidative stress and mitochondrial dysfunction contributes to postoperative cognitive dysfunction in elderly rats. Brain Behav Immun 2018; 73:661-669. [PMID: 30041011 DOI: 10.1016/j.bbi.2018.07.016] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/13/2018] [Accepted: 07/20/2018] [Indexed: 11/25/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is defined by cognitive impairment determined by neuropsychological tests from before to after surgery. Several mechanisms have been proposed in this bidirectional communication between the immune system and the brain after surgery. We aimed at understanding the mechanisms underlying POCD elderly rats in an experimental tibial fracture model. Elderly male Wistar rats were subjected to tibial fracture (TF) model. Control (sham) and fracture (TF) groups were followed to determine nitrite/nitrate concentration; oxidative damage to lipids and proteins; the activity of antioxidant enzymes (superoxide dismutase-SOD and catalase-CAT), mitochondrial respiratory chain enzymes, and creatine kinase (CK); and BDNF levels in the hippocampus and prefrontal cortex (at 24 h and at seven days) and cognitive function through habituation to the open field task and novel object recognition task (only at seven days). TF group presented increased concentration of nitrite/nitrate, hippocampal lipid peroxidation at seven days, protein oxidative damage in the prefrontal cortex and hippocampus at 24 h, decreased antioxidant activity in both structures on the first postoperative day and compromised function of the mitochondrial respiratory chain complexes as well as the CK enzyme. In addition, the levels of BDNF were reduced and memory function was impaired in the TF group. In conclusion, elderly rats submitted to an experimental model of tibial fracture displayed memory impairment accompanied by an increase in oxidative stress, mitochondrial dysfunction and reduced neurotrophin level.
Collapse
Affiliation(s)
- Martins Back Netto
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Aloir Neri de Oliveira Junior
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Mariana Goldim
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Khiany Mathias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Maria Eduarda Fileti
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Naiana da Rosa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Ana Olivia Laurentino
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Bianca Xavier de Farias
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Ana Beatriz Costa
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Jucelia Jeremias Fortunato
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Amanda Della Giustina
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil; Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Fabricia Petronilho
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarão, SC, Brazil.
| |
Collapse
|
43
|
Huang MH, Chen MH, Tu PC, Bai YM, Su TP, Yang BH, Liu RS, Li CT. Elevated tumor necrosis factor-alpha receptor subtype 1 and the association with abnormal brain function in treatment-resistant depression. J Affect Disord 2018; 235:250-256. [PMID: 29660639 DOI: 10.1016/j.jad.2018.04.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/06/2018] [Accepted: 04/04/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) patients have shown elevated plasma levels of pro-inflammatory biomarkers compared to healthy controls. We hypothesized increased serum tumor necrosis factor-alpha receptor subtype 1 (TNF-α R1) is more associated with impaired brain function in patients with treatment-resistant depression (TRD) than those without TRD. METHODS 34 MDD patients and 34 healthy subjects were recruited and we separated MDD patients to TRD group (n = 20) and non-TRD (n = 14) group. Pro-inflammatory cytokines were assessed by enzyme-linked immunosorbent assays. A standardized uptake values (SUV) of glucose metabolism measured by 18F-FDG positron-emission-tomography (PET) was applied to all subjects for subsequent region-of- interest analyses and whole-brain voxel-wise analyses. 18F-FDG-PET measures glucose uptake into astrocytes in response to glutamate release from neuronal cells, and was thus used as a proxy measure to quantify glutamatergic neurotransmission in the human brain. RESULTS Post-hoc analysis revealed that TRD group had higher serum concentrations of TNF-α R1 compared to healthy control or non-TRD group. In the MDD group, higher serum concentrations of TNF-α R1 significantly correlated with decreased SUV in anterior cingulate cortex (ACC) and bilateral caudate nucleus. The ROI analysis further supported the negative correlations of plasma TNF-α R1 and SUV in the ACC and caudate nucleus. Such correlation is more consistent in TRD group than in non-TRD and HC groups. LIMITATION Glutamate neurotransmission and the effect of chronic stress on glutamate release in the brain were not measured directly. CONCLUSIONS Increased TNF-α R1 was associated with impaired glutamatergic neurotransmission of caudate nucleus and ACC in MDD patients, particularly in the TRD.
Collapse
Affiliation(s)
- Mao-Hsuan Huang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Pei-Chi Tu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Ya Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, 112, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan; Department of Psychiatry, Cheng-Hsin General Hospital, Taiwan 112
| | - Bang-Hung Yang
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ren-Shyan Liu
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Ta Li
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, 112, Taiwan; Division of Psychiatry, Faculty of Medicine, National Yang-Ming University, Taipei, 112, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
44
|
Huang YQ, Wu C, He XF, Wu D, He X, Liang FY, Dai GY, Pei Z, Xu GQ, Lan Y. Effects of Voluntary Wheel-Running Types on Hippocampal Neurogenesis and Spatial Cognition in Middle-Aged Mice. Front Cell Neurosci 2018; 12:177. [PMID: 29997480 PMCID: PMC6028571 DOI: 10.3389/fncel.2018.00177] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/05/2018] [Indexed: 12/03/2022] Open
Abstract
While increasing evidence demonstrated that voluntary wheel running promotes cognitive function, little is known on how different types of voluntary wheel running affect cognitive function in elderly populations. We investigated the effects of various voluntary wheel-running types on adult hippocampal neurogenesis and spatial cognition in middle-aged mice. Male C57BL6 and Thy1-green fluorescent protein (GFP) transgenic mice (13 months) were equally assigned to one of the following groups: (1) T1: no voluntary wheel running; (2) T2: intermittent voluntary wheel running; and (3) T3: continuous voluntary wheel running. The Thy1-GFP transgenic mice were used to specifically label granule cells, since Thy-1 is a promoter for neuronal expression. Behavioral evaluations suggested that intermittent voluntary wheel running improved Morris water maze performance in middle-aged mice. The number of BrdU-positive cells was significantly higher in both intermittent and continuous voluntary wheel running compared with no voluntary wheel running. However, only intermittent voluntary wheel running facilitated the newborn cells to differentiate into granule cells, while newborn cells tended to differentiate into astrocytes and repopulation of microglia was also enhanced in the continuous voluntary wheel-running group. These results indicated that intermittent voluntary exercise may be more beneficial for enhancing spatial memory. Effective improvement of hippocampal neurogenesis was also caused by intermittent voluntary wheel running in middle-aged mice.
Collapse
Affiliation(s)
- Yi-Qing Huang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cheng Wu
- Department of Rehabilitation Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Fei He
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dan Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xia He
- Department of Integrated Traditional and Western Medicine, Sichuan Bayi Rehabilitation Center, Chengdu, China
| | - Feng-Yin Liang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guang-Yan Dai
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Pei
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guang-Qing Xu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Rehabilitation Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yue Lan
- Department of Rehabilitation Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
45
|
Sandvig I, Augestad IL, Håberg AK, Sandvig A. Neuroplasticity in stroke recovery. The role of microglia in engaging and modifying synapses and networks. Eur J Neurosci 2018; 47:1414-1428. [PMID: 29786167 DOI: 10.1111/ejn.13959] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 02/06/2023]
Abstract
Neuroplasticity after ischaemic injury involves both spontaneous rewiring of neural networks and circuits as well as functional responses in neurogenic niches. These events involve complex interactions with activated microglia, which evolve in a dynamic manner over time. Although the exact mechanisms underlying these interactions remain poorly understood, increasing experimental evidence suggests a determining role of pro- and anti-inflammatory microglial activation profiles in shaping both synaptogenesis and neurogenesis. While the inflammatory response of microglia was thought to be detrimental, a more complex profile of the role of microglia in tissue remodelling is emerging. Experimental evidence suggests that microglia in response to injury can rapidly modify neuronal activity and modulate synaptic function, as well as be beneficial for the proliferation and integration of neural progenitor cells (NPCs) from endogenous neurogenic niches into functional networks thereby supporting stroke recovery. The manner in which microglia contribute towards sculpting neural synapses and networks, both in terms of activity-dependent and homeostatic plasticity, suggests that microglia-mediated pro- and/or anti-inflammatory activity may significantly contribute towards spontaneous neuronal plasticity after ischaemic lesions. In this review, we first introduce some of the key cellular and molecular mechanisms underlying neuroplasticity in stroke and then proceed to discuss the crosstalk between microglia and endogenous neuroplasticity in response to brain ischaemia with special focus on the engagement of synapses and neural networks and their implications for grey matter integrity and function in stroke repair.
Collapse
Affiliation(s)
- Ioanna Sandvig
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ingrid Lovise Augestad
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Asta Kristine Håberg
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Radiology and Nuclear Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Axel Sandvig
- Faculty of Medicine and Health Sciences, Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Neurology, St Olav's Hospital, Trondheim University Hospital, Trondheim, Norway.,Department of Pharmacology and Clinical Neurosciences, Division of Neuro, Head and Neck, Umeå University Hospital, Umeå, Sweden
| |
Collapse
|
46
|
Zhu SZ, Szeto V, Bao MH, Sun HS, Feng ZP. Pharmacological approaches promoting stem cell-based therapy following ischemic stroke insults. Acta Pharmacol Sin 2018; 39:695-712. [PMID: 29671416 DOI: 10.1038/aps.2018.23] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/13/2018] [Indexed: 02/06/2023] Open
Abstract
Stroke can lead to long-term neurological deficits. Adult neurogenesis, the continuous generation of newborn neurons in distinct regions of the brain throughout life, has been considered as one of the appoaches to restore the neurological function following ischemic stroke. However, ischemia-induced spontaneous neurogenesis is not suffcient, thus cell-based therapy, including infusing exogenous stem cells or stimulating endogenous stem cells to help repair of injured brain, has been studied in numerous animal experiments and some pilot clinical trials. While the effects of cell-based therapy on neurological function during recovery remains unproven in randomized controlled trials, pharmacological agents have been administrated to assist the cell-based therapy. In this review, we summarized the limitations of ischemia-induced neurogenesis and stem-cell transplantation, as well as the potential proneuroregenerative effects of drugs that may enhance efficacy of cell-based therapies. Specifically, we discussed drugs that enhance proliferation, migration, differentiation, survival and function connectivity of newborn neurons, which may restore neurobehavioral function and improve outcomes in stroke patients.
Collapse
|
47
|
Kim M, Jung K, Kim IS, Lee IS, Ko Y, Shin JE, Park KI. TNF-α induces human neural progenitor cell survival after oxygen-glucose deprivation by activating the NF-κB pathway. Exp Mol Med 2018; 50:1-14. [PMID: 29622770 PMCID: PMC5938012 DOI: 10.1038/s12276-018-0033-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 11/14/2017] [Accepted: 12/05/2017] [Indexed: 11/21/2022] Open
Abstract
Neural progenitor cell (NPC) transplantation has been shown to be beneficial in the ischemic brain. However, the low survival rate of transplanted NPCs in an ischemic microenvironment limits their therapeutic effects. Tumor necrosis factor-alpha (TNF-α) is one of the proinflammatory cytokines involved in the pathogenesis of various injuries. On the other hand, several studies have shown that TNF-α influences the proliferation, survival, and differentiation of NPCs. Our study investigated the effect of TNF-α pretreatment on human NPCs (hNPCs) under ischemia-related conditions in vitro. hNPCs harvested from fetal brain tissue were pretreated with TNF-α before being subjected to oxygen–glucose deprivation (OGD) to mimic ischemia in vitro. TNF-α pretreatment improved the viability and reduced the apoptosis of hNPCs after OGD. At the molecular level, TNF-α markedly increased the level of NF-κB signaling in hNPCs, and an NF-κB pathway inhibitor, BAY11-7082, completely reversed the protective effects of TNF-α on hNPCs. These results suggest that TNF-α improves hNPC survival by activating the NF-κB pathway. In addition, TNF-α significantly enhanced the expression of cellular inhibitor of apoptosis 2 (cIAP2). Use of a lentivirus-mediated short hairpin RNA targeting cIAP2 mRNA demonstrated that cIAP2 protected against OGD-induced cytotoxicity in hNPCs. Our study of intracellular NF-κB signaling revealed that inhibition of NF-κB activity abolished the TNF-α-mediated upregulation of cIAP2 in hNPCs and blocked TNF-α-induced cytoprotection against OGD. Therefore, this study suggests that TNF-α pretreatment, which protects hNPCs from OGD-induced apoptosis by activating the NF-κB pathway, provides a safe and simple approach to improve the viability of transplanted hNPCs in cerebral ischemia. A potent “survival signal” for brain stem cells could enable effective regenerative therapies for stroke patients. Neural progenitor cells (NPCs) can develop into functional neurons and supportive glial cells, and researchers are tantalized by the prospect of using NPCs to repair damaged brain tissue. NPCs generally fail to flourish after transplantation, but a team led by Kook In Park at Yonsei University College of Medicine, South Korea, have found a signaling factor that helps these cells to survive and divide. Tumor necrosis factor-α (TNF-α) is associated with inflammation, but also protects neurons after a stroke. The researchers showed that pretreatment with TNF-α preserved NPCs exposed to starvation and oxygen-deprivation conditions in cell culture by activating critical cell survival pathways. These findings suggest that TNF-α may enable NPCs to survive long enough to repair post-stroke neurological damage.
Collapse
Affiliation(s)
- Miri Kim
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kwangsoo Jung
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Il-Sun Kim
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Il-Shin Lee
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Younhee Ko
- Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin, 17035, Korea
| | - Jeong Eun Shin
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Kook In Park
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Korea. .,Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Korea. .,Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, 03722, Korea.
| |
Collapse
|
48
|
The use of quetiapine in the treatment of major depressive disorder: Evidence from clinical and experimental studies. Neurosci Biobehav Rev 2018; 86:36-50. [DOI: 10.1016/j.neubiorev.2017.12.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/24/2017] [Accepted: 12/24/2017] [Indexed: 12/19/2022]
|
49
|
Neurotrophins and neuroinflammation in fetuses exposed to maternal depression and anxiety disorders during pregnancy: a comparative study on cord blood. Arch Womens Ment Health 2018; 21:105-111. [PMID: 28884439 DOI: 10.1007/s00737-017-0774-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/24/2017] [Indexed: 12/27/2022]
Abstract
In recent years, there have been changes in the approach to maternal psychiatric disorders and their effects on the fetus, with the focus redirected to the search for biological markers. Neurotrophic factors and inflammatory processes have received particular attention in the past few years. According to the Structured Clinical Interview for Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV), the study sample (n = 136) consisted of three groups: mothers with major depressive disorder (MDD group, n = 25), mothers with anxiety disorder (AD group, n = 18), and mothers without any psychiatric disorders (not diagnosed (ND) group, n = 93). During the delivery/cesarean section, a blood sample was obtained from the umbilical cord. Serum concentrations of BDNF, NT-3, FGF2, TNF-α, and neopterin were determined by enzyme-linked immunosorbent assay (ELISA), according to the manufacturer's procedure. Clinical and biochemical characteristics were assessed. We did not find a significant difference among the three study groups with regard to BDNF, NT-3, and TNF-α levels. The ANOVA test indicated statistically significant differences in FGF2 levels and neopterin between the study groups. The newborns of mothers with AD had significantly higher FGF2 levels and significantly higher neopterin levels when compared with those of mothers with MDD and healthy mothers. The present study sheds light on the effects of higher FGF2 and neopterin levels in fetuses exposed to AD. Our results should be replicated through further prospective studies with a larger sample size.
Collapse
|
50
|
Wang Z, He X, Fan X. Postnatal administration of memantine rescues TNF-α-induced decreased hippocampal precursor proliferation. Neurosci Lett 2018; 662:173-180. [DOI: 10.1016/j.neulet.2017.10.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/19/2017] [Accepted: 10/13/2017] [Indexed: 01/16/2023]
|