1
|
Meng J, Pan P, Guo G, Chen A, Meng X, Liu H. Transient CSF1R inhibition ameliorates behavioral deficits in Cntnap2 knockout and valproic acid-exposed mouse models of autism. J Neuroinflammation 2024; 21:262. [PMID: 39425203 PMCID: PMC11487716 DOI: 10.1186/s12974-024-03259-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Microglial abnormality and heterogeneity are observed in autism spectrum disorder (ASD) patients and animal models of ASD. Microglial depletion by colony stimulating factor 1-receptor (CSF1R) inhibition has been proved to improve autism-like behaviors in maternal immune activation mouse offspring. However, it is unclear whether CSF1R inhibition has extensive effectiveness and pharmacological heterogeneity in treating autism models caused by genetic and environmental risk factors. Here, we report pharmacological functions and cellular mechanisms of PLX5622, a small-molecule CSF1R inhibitor, in treating Cntnap2 knockout and valproic acid (VPA)-exposed autism model mice. For the Cntnap2 knockout mice, PLX5622 can improve their social ability and reciprocal social behavior, slow down their hyperactivity in open field and repetitive grooming behavior, and enhance their nesting ability. For the VPA model mice, PLX5622 can enhance their social ability and social novelty, and alleviate their anxiety behavior, repetitive and stereotyped autism-like behaviors such as grooming and marble burying. At the cellular level, PLX5622 restores the morphology and/or number of microglia in the somatosensory cortex, striatum, and hippocampal CA1 regions of the two models. Specially, PLX5622 corrects neurophysiological abnormalities in the striatum of the Cntnap2 knockout mice, and in the somatosensory cortex, striatum, and hippocampal CA1 regions of the VPA model mice. Incidentally, microglial dynamic changes in the VPA model mice are also reported. Our study demonstrates that microglial depletion and repopulation by transient CSF1R inhibition is effective, and however, has differential pharmacological functions and cellular mechanisms in rescuing behavioral deficits in the two autism models.
Collapse
Affiliation(s)
- Jiao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Pengming Pan
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Gengshuo Guo
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Anqi Chen
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
| | - Heli Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
- Autism Research Center, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
2
|
Zhang HL, Hu S, Qu ST, Lv MD, Wang JJ, Liu XT, Yao JH, Ding YY, Xu GY. Inhibition of NKCC1 Ameliorates Anxiety and Autistic Behaviors Induced by Maternal Immune Activation in Mice. Curr Issues Mol Biol 2024; 46:1851-1864. [PMID: 38534737 DOI: 10.3390/cimb46030121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is thought to result from susceptibility genotypes and environmental risk factors. The offspring of women who experience pregnancy infection have an increased risk for autism. Maternal immune activation (MIA) in pregnant animals produces offspring with autistic behaviors, making MIA a useful model for autism. However, how MIA causes autistic behaviors in offspring is not fully understood. Here, we show that NKCC1 is critical for mediating autistic behaviors in MIA offspring. We confirmed that MIA induced by poly(I:C) infection during pregnancy leads to autistic behaviors in offspring. We further demonstrated that MIA offspring showed significant microglia activation, excessive dendritic spines, and narrow postsynaptic density (PSD) in their prefrontal cortex (PFC). Then, we discovered that these abnormalities may be caused by overexpression of NKCC1 in MIA offspring's PFCs. Finally, we ameliorated the autistic behaviors using PFC microinjection of NKCC1 inhibitor bumetanide (BTN) in MIA offspring. Our findings may shed new light on the pathological mechanisms for autism caused by pregnancy infection.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Shufen Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Shu-Ting Qu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Meng-Dan Lv
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Jun-Jun Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Xin-Ting Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Jia-He Yao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Yi-Yan Ding
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Suzhou Medical College of Soochow University, Medical Center of Soochow University, Suzhou 215123, China
| |
Collapse
|
3
|
Guerrin CG, Prasad K, Vazquez-Matias DA, Zheng J, Franquesa-Mullerat M, Barazzuol L, Doorduin J, de Vries EF. Prenatal infection and adolescent social adversity affect microglia, synaptic density, and behavior in male rats. Neurobiol Stress 2023; 27:100580. [PMID: 37920548 PMCID: PMC10618826 DOI: 10.1016/j.ynstr.2023.100580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
Maternal infection during pregnancy and childhood social trauma have been associated with neurodevelopmental and affective disorders, such as schizophrenia, autism spectrum disorders, bipolar disorder and depression. These disorders are characterized by changes in microglial cells, which play a notable role in synaptic pruning, and synaptic deficits. Here, we investigated the effect of prenatal infection and social adversity during adolescence - either alone or in combination - on behavior, microglia, and synaptic density. Male offspring of pregnant rats injected with poly I:C, mimicking prenatal infection, were exposed to repeated social defeat during adolescence. We found that maternal infection during pregnancy prevented the reduction in social behavior and increase in anxiety induced by social adversity during adolescence. Furthermore, maternal infection and social adversity, alone or in combination, induced hyperlocomotion in adulthood. Longitudinal in vivo imaging with [11C]PBR28 positron emission tomography revealed that prenatal infection alone and social adversity during adolescence alone induced a transient increase in translocator protein TSPO density, an indicator of glial reactivity, whereas their combination induced a long-lasting increase that remained until adulthood. Furthermore, only the combination of prenatal infection and social adversity during adolescence induced an increase in microglial cell density in the frontal cortex. Prenatal infection increased proinflammatory cytokine IL-1β protein levels in hippocampus and social adversity reduced anti-inflammatory cytokine IL-10 protein levels in hippocampus during adulthood. This reduction in IL-10 was prevented if rats were previously exposed to prenatal infection. Adult offspring exposed to prenatal infection or adolescent social adversity had a higher synaptic density in the frontal cortex, but not hippocampus, as evaluated by synaptophysin density. Interestingly, such an increase in synaptic density was not observed in rats exposed to the combination of prenatal infection and social adversity, perhaps due to the long-lasting increase in microglial density, which may lead to an increase in microglial synaptic pruning. These findings suggest that changes in microglia activity and cytokine release induced by prenatal infection and social adversity during adolescence may be related to a reduced synaptic pruning, resulting in a higher synaptic density and behavioral changes in adulthood.
Collapse
Affiliation(s)
- Cyprien G.J. Guerrin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Kavya Prasad
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Daniel A. Vazquez-Matias
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Jing Zheng
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Maria Franquesa-Mullerat
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Erik F.J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| |
Collapse
|
4
|
Edemann-Callesen H, Bernhardt N, Hlusicka EB, Hintz F, Habelt B, Winter R, Neubert I, Pelz M, Filla A, Soto-Montenegro ML, Winter C, Hadar R. Supplement Treatment with NAC and Omega-3 Polyunsaturated Fatty Acids during Pregnancy Partially Prevents Schizophrenia-Related Outcomes in the Poly I:C Rat Model. Antioxidants (Basel) 2023; 12:antiox12051068. [PMID: 37237933 DOI: 10.3390/antiox12051068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Heightened levels of inflammation and oxidative stress are thought to be involved in the pathophysiology of schizophrenia. We aimed to assess whether intake of anti-inflammatory and anti-oxidant drugs during pregnancy prevents later schizophrenia-related outcomes in a neurodevelopmental rat model of this disorder. METHODS Pregnant Wistar rats were injected with polyriboinosinic-polyribocytidilic acid (Poly I:C) or saline and subsequently treated with either N-acetyl cysteine (NAC) or omega-3 polyunsaturated fatty acids (PUFAs) until delivery. Controls rats received no treatment. In the offspring, neuroinflammation and anti-oxidant enzyme activity were assessed on postnatal day (PND) 21, 33, 48, and 90. Behavioral testing was performed at PND 90, followed by post-mortem neurochemical assessment and ex vivo MRI. RESULTS The supplement treatment led to a quicker restoration of the wellbeing of dams. In the adolescent Poly I:C offspring, the supplement treatment prevented an increase in microglial activity and partially prevented a deregulation in the anti-oxidant defense system. In the adult Poly I:C offspring, supplement treatment partially prevented dopamine deficits, which was paralleled by some changes in behavior. Exposure to omega-3 PUFAs prevented the enlargement of lateral ventricles. CONCLUSION Intake of over-the-counter supplements may assist in especially targeting the inflammatory response related to schizophrenia pathophysiology, aiding in diminishing later disease severity in the offspring.
Collapse
Affiliation(s)
- Henriette Edemann-Callesen
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Nadine Bernhardt
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Elizabeth Barroeta Hlusicka
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Franziska Hintz
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Bettina Habelt
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
- Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany
| | - Rebecca Winter
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Isabell Neubert
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Meike Pelz
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Alexandra Filla
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Maria Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Grupo de Investigación de Alto Rendimiento en Fisiopatología y Farmacología del Sistema Digestivo (NeuGut-URJC), Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
| | - Christine Winter
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ravit Hadar
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
5
|
Rodrigues-Neves AC, Ambrósio AF, Gomes CA. Microglia sequelae: brain signature of innate immunity in schizophrenia. Transl Psychiatry 2022; 12:493. [PMID: 36443303 PMCID: PMC9705537 DOI: 10.1038/s41398-022-02197-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Schizophrenia is a psychiatric disorder with significant impact on individuals and society. The current pharmacologic treatment, which principally alleviates psychosis, is focused on neurotransmitters modulation, relying on drugs with severe side effects and ineffectiveness in a significant percentage of cases. Therefore, and due to difficulties inherent to diagnosis and treatment, it is vital to reassess alternative cellular and molecular drug targets. Distinct risk factors - genetic, developmental, epigenetic, and environmental - have been associated with disease onset and progression, giving rise to the proposal of different pathophysiological mechanisms and putative pharmacological targets. Immunity is involved and, particularly microglia - innate immune cells of the central nervous system, critically involved in brain development - have captured attention as cellular players. Microglia undergo marked morphologic and functional alterations in the human disease, as well as in animal models of schizophrenia, as reported in several original papers. We cluster the main findings of clinical studies by groups of patients: (1) at ultra-high risk of psychosis, (2) with a first episode of psychosis or recent-onset schizophrenia, and (3) with chronic schizophrenia; in translational studies, we highlight the time window of appearance of particular microglia alterations in the most well studied animal model in the field (maternal immune activation). The organization of clinical and translational findings based on schizophrenia-associated microglia changes in different phases of the disease course may help defining a temporal pattern of microglia changes and may drive the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- A. Catarina Rodrigues-Neves
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - António. F. Ambrósio
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Catarina A. Gomes
- grid.8051.c0000 0000 9511 4342Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal ,grid.8051.c0000 0000 9511 4342Univ Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| |
Collapse
|
6
|
Huang ST, Chen BB, Song ZJ, Tang HL, Hua R, Zhang YM. Unraveling the role of Epac1-SOCS3 signaling in the development of neonatal-CRD-induced visceral hypersensitivity in rats. CNS Neurosci Ther 2022; 28:1393-1408. [PMID: 35702948 PMCID: PMC9344090 DOI: 10.1111/cns.13880] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/28/2022] Open
Abstract
Aims Visceral hypersensitivity in irritable bowel syndrome (IBS) is widespread, but effective therapies for it remain elusive. As a canonical anti‐inflammatory protein, suppressor of cytokine signaling 3 (SOCS3) reportedly relays exchange protein 1 directly activated by cAMP (Epac1) signaling and inhibits the intracellular response to inflammatory cytokines. Despite the inhibitory effect of SOCS3 on the pro‐inflammatory response and neuroinflammation in PVN, the systematic investigation of Epac1‐SOCS3 signaling involved in visceral hypersensitivity remains unknown. This study aimed to explore Epac1‐SOCS3 signaling in the activity of hypothalamic paraventricular nucleus (PVN) corticotropin‐releasing factor (CRF) neurons and visceral hypersensitivity in adult rats experiencing neonatal colorectal distension (CRD). Methods Rats were subjected to neonatal CRD to simulate visceral hypersensitivity to investigate the effect of Epac1‐SOCS3 signaling on PVN CRF neurons. The expression and activity of Epac1 and SOCS3 in nociceptive hypersensitivity were determined by western blot, RT‐PCR, immunofluorescence, radioimmunoassay, electrophysiology, and pharmacology. Results In neonatal‐CRD‐induced visceral hypersensitivity model, Epac1 and SOCS3 expressions were downregulated and IL‐6 levels elevated in PVN. However, infusion of Epac agonist 8‐pCPT in PVN reduced CRF neuronal firing rates, and overexpression of SOCS3 in PVN by AAV‐SOCS3 inhibited the activation of PVN neurons, reduced visceral hypersensitivity, and precluded pain precipitation. Intervention with IL‐6 neutralizing antibody also alleviated the visceral hypersensitivity. In naïve rats, Epac antagonist ESI‐09 in PVN increased CRF neuronal firing. Consistently, genetic knockdown of Epac1 or SOCS3 in PVN potentiated the firing rate of CRF neurons, functionality of HPA axis, and sensitivity of visceral nociception. Moreover, pharmacological intervention with exogenous IL‐6 into PVN simulated the visceral hypersensitivity. Conclusions Inactivation of Epac1‐SOCS3 pathway contributed to the neuroinflammation accompanied by the sensitization of CRF neurons in PVN, precipitating visceral hypersensitivity and pain in rats experiencing neonatal CRD.
Collapse
Affiliation(s)
- Si-Ting Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.,NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
| | - Bin-Bin Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.,NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
| | - Zhi-Jing Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.,NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
| | - Hui-Li Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.,NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
| | - Rong Hua
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yong-Mei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.,NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
| |
Collapse
|
7
|
Yu D, Li T, Delpech JC, Zhu B, Kishore P, Koshi T, Luo R, Pratt KJ, Popova G, Nowakowski TJ, Villeda SA, Piao X. Microglial GPR56 is the molecular target of maternal immune activation-induced parvalbumin-positive interneuron deficits. SCIENCE ADVANCES 2022; 8:eabm2545. [PMID: 35544642 PMCID: PMC9075805 DOI: 10.1126/sciadv.abm2545] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/23/2022] [Indexed: 06/15/2023]
Abstract
Parvalbumin-positive (PV+) interneurons play a critical role in maintaining circuit rhythm in the brain, and their reduction is implicated in autism spectrum disorders. Animal studies demonstrate that maternal immune activation (MIA) leads to reduced PV+ interneurons in the somatosensory cortex and autism-like behaviors. However, the underlying molecular mechanisms remain largely unknown. Here, we show that MIA down-regulates microglial Gpr56 expression in fetal brains in an interleukin-17a-dependent manner and that conditional deletion of microglial Gpr56 [Gpr56 conditional knockout (cKO)] mimics MIA-induced PV+ interneuron defects and autism-like behaviors in offspring. We further demonstrate that elevated microglial tumor necrosis factor-α expression is the underlying mechanism by which MIA and Gpr56 cKO impair interneuron generation. Genetically restoring Gpr56 expression in microglia ameliorates PV+ interneuron deficits and autism-like behaviors in MIA offspring. Together, our study demonstrates that microglial GPR56 plays an important role in PV+ interneuron development and serves as a salient target of MIA-induced neurodevelopmental disorders.
Collapse
Affiliation(s)
- Diankun Yu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neuroscience, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Tao Li
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neuroscience, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Jean-Christophe Delpech
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Beika Zhu
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neuroscience, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Priya Kishore
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Tatsuhiro Koshi
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Rong Luo
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Karishma J.B. Pratt
- Department of Anatomy, University of California at San Francisco, San Francisco CA 94143, USA
- Developmental and Stem Cell Biology Graduate Program, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Galina Popova
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
- Department of Anatomy, University of California at San Francisco, San Francisco CA 94143, USA
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Tomasz J. Nowakowski
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neuroscience, University of California at San Francisco, San Francisco, CA 94143, USA
- Department of Anatomy, University of California at San Francisco, San Francisco CA 94143, USA
- Department of Psychiatry, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Saul A. Villeda
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
- Department of Anatomy, University of California at San Francisco, San Francisco CA 94143, USA
- Developmental and Stem Cell Biology Graduate Program, University of California at San Francisco, San Francisco, CA 94143, USA
- Department of Physical Therapy and Rehabilitation Science, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Xianhua Piao
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neuroscience, University of California at San Francisco, San Francisco, CA 94143, USA
- Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Neonatology, Department of Pediatrics, University of California at San Francisco, San Francisco, CA 94158, USA
- Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
8
|
Mousaviyan R, Davoodian N, Alizadeh F, Ghasemi-Kasman M, Mousavi SA, Shaerzadeh F, Kazemi H. Zinc Supplementation During Pregnancy Alleviates Lipopolysaccharide-Induced Glial Activation and Inflammatory Markers Expression in a Rat Model of Maternal Immune Activation. Biol Trace Elem Res 2021; 199:4193-4204. [PMID: 33400154 DOI: 10.1007/s12011-020-02553-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/17/2020] [Indexed: 12/22/2022]
Abstract
Maternal immune activation (MIA) model has been profoundly described as a suitable approach to study the pathophysiological mechanisms of neuropsychiatric disorders, including schizophrenia. Our previous study revealed that prenatal exposure to lipopolysaccharide (LPS) induced working memory impairments in only male offspring. Based on the putative role of prefrontal cortex (PFC) in working memory process, the current study was conducted to examine the long-lasting effect of LPS-induced MIA on several neuroinflammatory mediators in the PFC of adult male pups. We also investigated whether maternal zinc supplementation can alleviate LPS-induced alterations in this region. Pregnant rats received intraperitoneal injections of either LPS (0.5 mg/kg) or saline on gestation days 15/16 and supplemented with ZnSO4 (30 mg/kg) throughout pregnancy. At postnatal day 60, the density of both microglia and astrocyte cells and the expression levels of IL-6, IL-1β, iNOS, TNF-α, NF-κB, and GFAP were evaluated in the PFC of male pups. Although maternal LPS treatment increased microglia and astrocyte density, number of neurons in the PFC of adult offspring remained unchanged. These findings were accompanied by the exacerbated mRNA levels of IL-6, IL-1β, iNOS, TNF-α, NF-κB, and GFAP as well. Conversely, prenatal zinc supplementation alleviated the mentioned alterations induced by LPS. These findings support the idea that the deleterious effects of prenatal LPS exposure could be attenuated by zinc supplementation during pregnancy. It is of interest to suggest early therapeutic intervention as a valuable approach to prevent neurodevelopmental deficits, following maternal infection. Schematic diagram describing the experimental timeline. On gestation days (GD) 15 and 16, pregnant dams were administered with intraperitoneal injections of either LPS (0.5 mg/kg) or vehicle and supplemented with ZnSO4 (30 mg/kg) throughout pregnancy by gavage. The resulting offspring were submitted to qPCR, immunostaining, and morphological analysis at PND 60. Maternal zinc supplementation alleviated increased expression levels of inflammatory mediators and microglia and astrocyte density induced by LPS in the PFC of treated offspring. PND postnatal day, PFC prefrontal cortex.
Collapse
Affiliation(s)
- Ronak Mousaviyan
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Nahid Davoodian
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Faezeh Alizadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Neuroscience Reesearch Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Seyed Abdollah Mousavi
- Pathology Department, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Shaerzadeh
- Department of Neuroscience, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Haniyeh Kazemi
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
9
|
Chen JH, Sun Y, Ju PJ, Wei JB, Li QJ, Winston JH. Estrogen augmented visceral pain and colonic neuron modulation in a double-hit model of prenatal and adult stress. World J Gastroenterol 2021; 27:5060-5075. [PMID: 34497435 PMCID: PMC8384739 DOI: 10.3748/wjg.v27.i30.5060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/19/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic stress during pregnancy may increase visceral hyperalgesia of offspring in a sex-dependent way. Combining adult stress in offspring will increase this sensitivity. Based on the evidence implicating estrogen in exacerbating visceral hypersensitivity in female rodents in preclinical models, we predicted that chronic prenatal stress (CPS) + chronic adult stress (CAS) will maximize visceral hyperalgesia; and that estrogen plays an important role in colonic hyperalgesia.
AIM The aim was to illuminate the role of estrogen in colonic hyperalgesia and its underlying mechanisms.
METHODS We established a CPS plus CAS rodent model in which the balloon was used to distend the colorectum. The single-fiber recording in vivo and patch clamp experiments in vitro were used to monitor the colonic neuron’s activity. The reverse transcription-polymerase chain reaction, western blot, and immunofluorescence were used to study the effects of CPS and CAS on colon primary afferent sensitivity. We used ovariectomy and letrozole to reduce estrogen levels of female rats respectively in order to assess the role of estrogen in female-specific enhanced primary afferent sensitization.
RESULTS Spontaneous activity and single fiber activity were significantly greater in females than in males. The enhanced sensitization in female rats mainly came from low-threshold neurons. CPS significantly increased single-unit afferent fiber activity in L6-S2 dorsal roots in response. Activity was further enhanced by CAS. In addition, the excitability of colon-projecting dorsal root ganglion (DRG) neurons increased in CPS + CAS rats and was associated with a decrease in transient A-type K+ currents. Compared with ovariectomy, treatment with the aromatase inhibitor letrozole significantly reduced estrogen levels in female rats, confirming the gender difference. Moreover, mice treated with letrozole had decreased colonic DRG neuron excitability. The intrathecal infusion of estrogen increased brain-derived neurotrophic factor (BDNF) protein levels and contributed to the response to visceral pain. Western blotting showed that nerve growth factor protein was upregulated in CPS + CAS mice.
CONCLUSION This study adds to the evidence that estrogen-dependent sensitization of primary afferent colon neurons is involved in the development of chronic stress-induced visceral hypersensitivity in female rats.
Collapse
Affiliation(s)
- Jing-Hong Chen
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Ying Sun
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Pei-Jun Ju
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jin-Bao Wei
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qing-Jie Li
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - John H Winston
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| |
Collapse
|
10
|
Wang P, Li M, Zhao A, Ma J. Application of animal experimental models in the research of schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2021; 186:209-227. [PMID: 34155806 DOI: 10.1002/ajmg.b.32863] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 05/04/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022]
Abstract
Schizophrenia is a relatively common but serious mental illness that results in a heavy burden to patients, their families, and society. The disease can be triggered by multiple factors, while the specific pathogenesis remains unclear. The development of effective therapeutic drugs for schizophrenia relies on a comprehensive understanding of the basic biology and pathophysiology of the disease. Therefore, effective animal experimental models play a vital role in the study of schizophrenia. Based on different molecular mechanisms and modeling methods, the currently used experimental animal experimental models of schizophrenia can be divided into four categories that can better simulate the clinical symptoms and the interplay between susceptible genes and the environment: neurodevelopmental, drug-induced, genetic-engineering, and genetic-environmental interaction of animal experimental models. Each of these categories contains multiple subtypes, which has its own advantages and disadvantages and therefore requires careful selection in a research application. The emergence and utilization of these models are promising in the prediction of the risk of schizophrenia at the molecular level, which will shed light on effective and targeted treatment at the genetic level.
Collapse
Affiliation(s)
- Pengjie Wang
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China.,Department of Electron Microscope, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Manling Li
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Gui Yang, Guizhou, China
| | - Aizhen Zhao
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Jie Ma
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China.,Department of Electron Microscope, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| |
Collapse
|
11
|
Guo X, Mao R, Cui L, Wang F, Zhou R, Wang Y, Huang J, Zhu Y, Yao Y, Zhao G, Li Z, Chen J, Wang J, Fang Y. PAID study design on the role of PKC activation in immune/inflammation-related depression: a randomised placebo-controlled trial protocol. Gen Psychiatr 2021; 34:e100440. [PMID: 33912799 PMCID: PMC8030460 DOI: 10.1136/gpsych-2020-100440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/18/2021] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
Background Inflammation that is mediated by microglia activation plays an important role in the pathogenesis of depression. Microglia activation can lead to an increase in the levels of proinflammatory cytokines, including TNF-α, which leads to neuronal apoptosis in the specific neural circuits of some brain regions, abnormal cognition and treatment-resistant depression (TRD). Protein kinase C (PKC) is a key regulator of the microglia activation process. We assume that the abnormality in PKC might result in abnormal microglia activation, neuronal apoptosis, significant changes in emotional and cognitive neural circuits, and TRD. In the current study, we plan to target at the PKC signal pathway to improve the TRD treatment outcome. Methods and analysis This is a 12-week, ongoing, randomised, placebo-controlled trial. Patients with TRD (N=180) were recruited from Shanghai Mental Health Center, Shanghai Jiao Tong University. Healthy control volunteers (N=60) were recruited by advertisement. Patients with TRD were randomly assigned to 'escitalopram+golimumab (TNF-α inhibitor)', 'escitalopram+calcium tablet+vitamin D (PKC activator)' or 'escitalopram+placebo' groups. We define the primary outcome as changes in the 17-item Hamilton Depression Rating Scale (HAMD-17). The secondary outcome is defined as changes in anti-inflammatory effects, cognitive function and quality of life. Discussion This study might be the first randomised, placebo-controlled trial to target at the PKC signal pathway in patients with TRD. Our study might help to propose individualised treatment strategies for depression. Trial registration number The trial protocol is registered with ClinicalTrials.gov under protocol ID 81930033 and ClinicalTrials.gov ID NCT04156425.
Collapse
Affiliation(s)
- Xiaoyun Guo
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Mao
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lvchun Cui
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Wang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rubai Zhou
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Wang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Huang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuncheng Zhu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yamin Yao
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoqing Zhao
- Department of Psychology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zezhi Li
- Department of Neurology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Chen
- Clinical Research Center and Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhui Wang
- College of Life Sciences, Chinese Academy of Sciences, Beijing, China
| | - Yiru Fang
- Clinical Research Center and Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Science, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| |
Collapse
|
12
|
Li X, Fan X, Yuan X, Pang L, Hu S, Wang Y, Huang X, Song X. The Role of Butyric Acid in Treatment Response in Drug-Naïve First Episode Schizophrenia. Front Psychiatry 2021; 12:724664. [PMID: 34497548 PMCID: PMC8421030 DOI: 10.3389/fpsyt.2021.724664] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/28/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Butyric acid, a major short-chain fatty acid (SCFA), has an important role in the microbiota-gut-brain axis and brain function. This study investigated the role of butyric acid in treatment response in drug-naïve first episode schizophrenia. Methods: The study recruited 56 Chinese Han schizophrenia inpatients with normal body weight and 35 healthy controls. Serum levels of butyric acid were measured using Gas Chromatography-Mass Spectrometer (GC-MS) analysis at baseline (for all participants) and 24 weeks after risperidone treatment (for patients). Clinical symptoms were measured using the Positive and Negative Syndrome Scale (PANSS) for patients at both time points. Results: At baseline, there was no significant difference in serum levels of butyric acid between patients and healthy controls (p = 0.206). However, there was a significant increase in serum levels of butyric acid in schizophrenia patients after 24-week risperidone treatment (p = 0.030). The PANSS total and subscale scores were decreased significantly after 24-week risperidone treatment (p's < 0.001). There were positive associations between baseline serum levels of butyric acid and the reduction ratio of the PANSS total and subscale scores after controlling for age, sex, education, and duration of illness (p's < 0.05). Further, there was a positive association between the increase in serum levels of butyric acid and the reduction of the PANSS positive symptoms subscale scores (r = 0.38, p = 0.019) after controlling for potential confounding factors. Conclusions: Increased serum levels of butyric acid might be associated with a favorable treatment response in drug-naïve, first episode schizophrenia. The clinical implications of our findings were discussed.
Collapse
Affiliation(s)
- Xue Li
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China.,Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Xiaoduo Fan
- Psychotic Disorders Program, UMass Memorial Medical Center, University of Massachusetts Medical School, Worcester, MA, United States
| | - Xiuxia Yuan
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China.,Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Lijuan Pang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China.,Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| | - Shaohua Hu
- Center for Neuroscience and Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory of Mental Disorder Management in Zhejiang Province, Brain Research Institute of Zhejiang University, Hangzhou, China
| | - Yunpeng Wang
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Centre for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway
| | - Xufeng Huang
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Xueqin Song
- Department of Psychiatry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Biological Psychiatry International Joint Laboratory of Henan, Zhengzhou University, Zhengzhou, China.,Henan Psychiatric Transformation Research Key Laboratory, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Parker KG, White MG, Cipriani NA. Comparison of Molecular Methods and BRAF Immunohistochemistry (VE1 Clone) for the Detection of BRAF V600E Mutation in Papillary Thyroid Carcinoma: A Meta-Analysis. Head Neck Pathol 2020; 14:1067-1079. [PMID: 32358715 PMCID: PMC7669962 DOI: 10.1007/s12105-020-01166-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/23/2020] [Indexed: 10/24/2022]
Abstract
The evaluation of surgically resected papillary thyroid carcinomas (PTC) by immunohistochemistry (IHC) for BRAF mutation has diagnostic, prognostic and therapeutic implications. The goal of this meta-analysis was to perform a systematic review of studies using the VE1 clone (specific for detection of the BRAF V600E mutation) on formalin-fixed paraffin embedded (FFPE) thyroid surgical resection specimens for primary papillary thyroid carcinoma. The authors' molecular techniques, immunohistochemistry protocols, and scoring methods for VE1 immunostaining were also evaluated. This study included 4079 PTCs representing data from 23 studies. The results extracted from each study were split into two different groups, direct sequencing group or PCR group, based on the molecular "gold standard" method used to compare VE1 IHC staining. In the direct sequencing group, the IHC sensitivity was 100% (95% CI 0.97-1.00) and specificity 84% (95% 0.72-0.91). In the PCR group the sensitivity was 98% (95% CI 0.96-0.99) and specificity 89% (95% CI 0.82-0.94). Although immunohistochemical procedures varied by author, the overall performance of the VE1 clone shows that it is highly sensitive and relatively specific for detecting the BRAF V600E mutation in surgical resection specimens. However, standardization of immunohistochemical procedural method and scoring/interpretation criteria may improve the reliability and reproducibility for the use of VE1 clone for future practice.
Collapse
Affiliation(s)
- Kyle G Parker
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA.
| | - Michael G White
- Department of General Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole A Cipriani
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| |
Collapse
|
14
|
Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity. J Clin Med 2020; 9:jcm9082590. [PMID: 32785127 PMCID: PMC7464885 DOI: 10.3390/jcm9082590] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders with unknown heterogeneous aetiologies. Epidemiological studies have found an association between maternal infection and development of ASD in the offspring, and clinical findings reveal a state of immune dysregulation in the pre- and postnatal period of affected subjects. Maternal immune activation (MIA) has been proposed to mediate this association by altering fetal neurodevelopment and leading to autism. Although animal models have supported a causal link between MIA and development of ASD, their validity needs to be explored. Moreover, considering that only a small proportion of affected offspring develop autism, and that MIA has been implicated in related diseases such as schizophrenia, a key unsolved question is how disease specificity and phenotypic outcome are determined. Here, we have integrated preclinical and clinical evidence, including the use of animal models for establishing causality, to explore the role of maternal infections in ASD. A proposed priming/multi-hit model may offer insights into the clinical heterogeneity of ASD, its convergence with related disorders, and therapeutic strategies.
Collapse
|
15
|
Bulleyaconitine A Inhibits Visceral Nociception and Spinal Synaptic Plasticity through Stimulation of Microglial Release of Dynorphin A. Neural Plast 2020; 2020:1484087. [PMID: 32565774 PMCID: PMC7262664 DOI: 10.1155/2020/1484087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/14/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
Background Visceral pain is one of the most common types of pain and particularly in the abdomen is associated with gastrointestinal diseases. Bulleyaconitine A (BAA), isolated from Aconitum bulleyanum, is prescribed in China to treat chronic pain. The present study is aimed at evaluating the mechanisms underlying BAA visceral antinociception. Methods The rat model of chronic visceral hypersensitivity was set up by colonic perfusion of 2,4,6-trinitrobenzene sulfonic acid (TNBS) on postnatal day 10 with coapplication of heterotypic intermittent chronic stress (HeICS). Results The rat model of chronic visceral hypersensitivity exhibited remarkable abdominal withdrawal responses and mechanical hyperalgesia in hind paws, which were dose-dependently attenuated by single subcutaneous of administration of BAA (30 and 90 μg/kg). Pretreatment with the microglial inhibitor minocycline, dynorphin A antiserum, and κ-opioid receptor antagonist totally blocked BAA-induced visceral antinociception and mechanical antihyperalgesia. Spontaneous excitatory postsynaptic currents (sEPSCs) in spinal dorsal horn lamina II neurons were recorded by using whole-cell patch clamp. Its frequency (but not amplitude) from TNBS-treated rats was remarkably higher than that from naïve rats. BAA (1 μM) significantly reduced the frequency of sEPSCs from TNBS-treated rats but not naïve rats. BAA-inhibited spinal synaptic plasticity was blocked by minocycline, the dynorphin A antiserum, and κ-opioid receptor antagonist. Dynorphin A also inhibited spinal synaptic plasticity in a κ-opioid receptor-dependent manner. Conclusions These results suggest that BAA produces visceral antinociception by stimulating spinal microglial release of dynorphin A, which activates presynaptic κ-opioid receptors in afferent neurons and inhibits spinal synaptic plasticity, highlighting a novel interaction mode between microglia and neurons.
Collapse
|
16
|
Low frequency electroacupuncture alleviates neuropathic pain by activation of spinal microglial IL-10/β-endorphin pathway. Biomed Pharmacother 2020; 125:109898. [DOI: 10.1016/j.biopha.2020.109898] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/01/2020] [Accepted: 01/02/2020] [Indexed: 02/06/2023] Open
|