1
|
Lemos FS, Prins CA, Martinez AMB, Carpi-Santos R, Neumann AS, Meireles-da-Costa N, Luisetto R, de Mello-Coelho V, Oliveira FL. UHT Cow's Milk Supplementation Affects Cell Niches and Functions of the Gut-Brain Axis in BALB/c Mice. Biomedicines 2024; 12:2448. [PMID: 39595015 PMCID: PMC11591918 DOI: 10.3390/biomedicines12112448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Cow's milk is a bioactive cocktail with essential nutritional factors that is widely consumed during early childhood development. However, it has been associated with allergic responses and immune cell activation. Here, we investigate whether cow's milk consumption regulates gut-brain axis functions and affects patterns of behaviors in BALB/c mice, previously described by present low sociability, significant stereotypes, and restricted interest features. The major objectives consist of to investigate cow's milk supplementation as possible triggers interfering with cellular niches of the gut-brain axis and behavioral patterns. METHODS Male BALB/c at 6 weeks were randomly divided into two groups, one supplemented with cow's milk processed at ultra-high temperature (UHT) and another group receiving water (controls) three times per day (200 μL per dose) for one week. RESULTS Milk consumption disturbed histological compartments of the small intestine, including niches of KI67+-proliferating cells and CD138+ Ig-secreting plasma cells. In the liver, milk intake was associated with pro-inflammatory responses, oxidative stress, and atypical glycogen distribution. Milk-supplemented mice showed significant increase in granulocytes (CD11b+SSChigh cells) and CD4+ T cells in the blood. These mice also had neuroinflammatory signals, including an enhanced number of cortical Iba-1+ microglial cells in the brain and significant cerebellar expression of nitric oxide synthase 2 by Purkinje cells. These phenotypes and tissue disorders in milk-supplemented mice were associated with atypical behaviors, including low sociability, high restricted interest, and severe stereotypies. Moreover, synaptic niches were also disturbed after milk consumption, and Shank-3+ and Drebrin+ post-synaptic cells were significantly reduced in the brain of these mice. CONCLUSIONS Together, these data suggest that milk consumption interfered with the gut-brain axis in BALB/c mice and increased atypical behaviors, at least in part, linked to synapse dysfunctions, neuroinflammation, and oxidative stress regulation.
Collapse
Affiliation(s)
- Felipe S. Lemos
- Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-360, Brazil;
| | - Caio A. Prins
- Department of Pathology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (C.A.P.); (A.M.B.M.)
| | - Ana M. B. Martinez
- Department of Pathology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (C.A.P.); (A.M.B.M.)
| | - Raul Carpi-Santos
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| | - Arthur S. Neumann
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| | - Nathalia Meireles-da-Costa
- Molecular Carcinogenesis Program, Brazilian National Cancer Institute, Rio de Janeiro 20230-130, Brazil;
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35124 Padova, Italy;
| | - Valeria de Mello-Coelho
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| | - Felipe L. Oliveira
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (R.C.-S.); (A.S.N.); (V.d.M.-C.)
| |
Collapse
|
2
|
Qian F, He R, Du X, Wei Y, Zhou Z, Fan J, He Y. Microglia and Astrocytes Responses Contribute to Alleviating Inflammatory Damage by Repetitive Transcranial Magnetic Stimulation in Rats with Traumatic Brain Injury. Neurochem Res 2024; 49:2636-2651. [PMID: 38909329 DOI: 10.1007/s11064-024-04197-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a therapeutic strategy that shows promise in ameliorating the clinical sequelae following traumatic brain injury (TBI). These improvements are associated with neuroplastic changes in neurons and their synaptic connections. However, it has been hypothesized that rTMS may also modulate microglia and astrocytes, potentially potentiating their neuroprotective capabilities. This study aims to investigate the effects of high-frequency rTMS on microglia and astrocytes that may contribute to its neuroprotective effects. Feeney's weight-dropping method was used to establish rat models of moderate TBI. To evaluate the neuroprotective effect of high frequency rTMS on rats by observing the synaptic ultrastructure and the level of neuron apoptosis. The levels of several important inflammation-related proteins within microglia and astrocytes were assessed through immunofluorescence staining and western blot. Our findings demonstrate that injured neurons can be rescued through the modulation of microglia and astrocytes by rTMS. This modulation plays a key role in preserving the synaptic ultrastructure and inhibiting neuronal apoptosis. Among microglia, we observed that rTMS inhibited the levels of proinflammatory factors (CD16, IL-6 and TNF-α) and promoted the levels of anti-inflammatory factors (CD206, IL-10 and TNF-β). rTMS also reduced the levels of pyroptosis within microglia and pyroptosis-related proteins (NLRP3, Caspase-1, GSDMD, IL-1β and IL-18). Moreover, rTMS downregulated P75NTR expression and up-regulated IL33 expression in astrocytes. These findings suggest that regulation of microglia and astrocytes is the mechanism through which rTMS attenuates neuronal inflammatory damage after moderate TBI.
Collapse
Affiliation(s)
- FangFang Qian
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - RenHong He
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - XiaoHui Du
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Yi Wei
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Zhou Zhou
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - JianZhong Fan
- Department of Rehabilitation Medicine, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| | - YouHua He
- Department of Comprehensive Medical Treatment Ward, Guangdong Province, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Li Z, Wu X, Li H, Bi C, Zhang C, Sun Y, Yan Z. Complex interplay of neurodevelopmental disorders (NDDs), fractures, and osteoporosis: a mendelian randomization study. BMC Psychiatry 2024; 24:232. [PMID: 38539137 PMCID: PMC10967110 DOI: 10.1186/s12888-024-05693-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/18/2024] [Indexed: 12/11/2024] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs), such as Attention-Deficit/Hyperactivity Disorder (ADHD), Autism Spectrum Disorder (ASD), and Tourette Syndrome (TS), have been extensively studied for their multifaceted impacts on social and emotional well-being. Recently, there has been growing interest in their potential relationship with fracture risks in adulthood. This study aims to explore the associations between these disorders and fracture rates, in order to facilitate better prevention and treatment. METHODS Employing a novel approach, this study utilized Mendelian randomization (MR) analysis to investigate the complex interplay between ADHD, ASD, TS, and fractures. The MR framework, leveraging extensive genomic datasets, facilitated a systematic examination of potential causal relationships and genetic predispositions. RESULTS The findings unveil intriguing bidirectional causal links between ADHD, ASD, and specific types of fractures. Notably, ADHD is identified as a risk factor for fractures, with pronounced associations in various anatomical regions, including the skull, trunk, and lower limbs. Conversely, individuals with specific fractures, notably those affecting the femur and lumbar spine, exhibit an increased genetic predisposition to ADHD and ASD. In this research, no correlation was found between TS and fractures, or osteoporosis.These results provide a genetic perspective on the complex relationships between NDDs and fractures, emphasizing the importance of early diagnosis, intervention, and a holistic approach to healthcare. CONCLUSION This research sheds new light on the intricate connections between NDDs and fractures, offering valuable insights into potential risk factors and causal links. The bidirectional causal relationships between ADHD, ASD, and specific fractures highlight the need for comprehensive clinical approaches that consider both NDDs and physical well-being.
Collapse
Affiliation(s)
- Zefang Li
- Department of The First Clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xueqiang Wu
- Department of Health Science, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Hanzheng Li
- Department of The First Clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cong Bi
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Can Zhang
- School of Biomedical Sciences, Shandong First Medical University, Jinan, China
| | - Yiqing Sun
- Department of The First Clinical medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhaojun Yan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
4
|
Ong RCS, Beros JL, Fuller K, Wood FM, Melton PE, Rodger J, Fear MW, Barrett L, Stevenson AW, Tang AD. Non-severe thermal burn injuries induce long-lasting downregulation of gene expression in cortical excitatory neurons and microglia. Front Mol Neurosci 2024; 17:1368905. [PMID: 38476460 PMCID: PMC10927825 DOI: 10.3389/fnmol.2024.1368905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Burn injuries are devastating traumas, often leading to life-long consequences that extend beyond the observable burn scar. In the context of the nervous system, burn injury patients commonly develop chronic neurological disorders and have been suggested to have impaired motor cortex function, but the long-lasting impact on neurons and glia in the brain is unknown. Using a mouse model of non-severe burn injury, excitatory and inhibitory neurons in the primary motor cortex were labelled with fluorescent proteins using adeno-associated viruses (AAVs). A total of 5 weeks following the burn injury, virus labelled excitatory and inhibitory neurons were isolated using fluorescence-activated cell sorting (FACS). In addition, microglia and astrocytes from the remaining cortical tissue caudal to the motor cortex were immunolabelled and isolated with FACS. Whole transcriptome RNA-sequencing was used to identify any long-lasting changes to gene expression in the different cell types. RNA-seq analysis showed changes to the expression of a small number of genes with known functions in excitatory neurons and microglia, but not in inhibitory neurons or astrocytes. Specifically, genes related to GABA-A receptors in excitatory neurons and several cellular functions in microglia were found to be downregulated in burn injured mice. These findings suggest that non-severe burn injuries lead to long lasting transcriptomic changes in the brain, but only in specific cell types. Our findings provide a broad overview of the long-lasting impact of burn injuries on the central nervous system which may help identify potential therapeutic targets to prevent neurological dysfunction in burn patients.
Collapse
Affiliation(s)
- Rebecca C. S. Ong
- Experimental and Regenerative Neuroscience, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Sciences, Nedlands, WA, Australia
| | - Jamie L. Beros
- Experimental and Regenerative Neuroscience, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Sciences, Nedlands, WA, Australia
| | - Kathy Fuller
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Fiona M. Wood
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- Burn Injury Research Unit, The University of Western Australia, Crawley, WA, Australia
- Burns Service of Western Australia, WA Department of Health, Murdoch, WA, Australia
- Paediatric Burn Care, Telethon Kids Institute, Nedlands, WA, Australia
| | - Phillip E. Melton
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- School of Global and Population Health, The University of Western Australia, Crawley, WA, Australia
| | - Jennifer Rodger
- Experimental and Regenerative Neuroscience, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Sciences, Nedlands, WA, Australia
| | - Mark W. Fear
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Lucy Barrett
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- Burn Injury Research Unit, The University of Western Australia, Crawley, WA, Australia
- Burns Service of Western Australia, WA Department of Health, Murdoch, WA, Australia
| | - Andrew W. Stevenson
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
- Burn Injury Research Unit, The University of Western Australia, Crawley, WA, Australia
| | - Alexander D. Tang
- Experimental and Regenerative Neuroscience, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Sciences, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
5
|
Lamanna J, Meldolesi J. Autism Spectrum Disorder: Brain Areas Involved, Neurobiological Mechanisms, Diagnoses and Therapies. Int J Mol Sci 2024; 25:2423. [PMID: 38397100 PMCID: PMC10889781 DOI: 10.3390/ijms25042423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Autism spectrum disorder (ASD), affecting over 2% of the pre-school children population, includes an important fraction of the conditions accounting for the heterogeneity of autism. The disease was discovered 75 years ago, and the present review, based on critical evaluations of the recognized ASD studies from the beginning of 1990, has been further developed by the comparative analyses of the research and clinical reports, which have grown progressively in recent years up to late 2023. The tools necessary for the identification of the ASD disease and its related clinical pathologies are genetic and epigenetic mutations affected by the specific interaction with transcription factors and chromatin remodeling processes occurring within specific complexes of brain neurons. Most often, the ensuing effects induce the inhibition/excitation of synaptic structures sustained primarily, at dendritic fibers, by alterations of flat and spine response sites. These effects are relevant because synapses, established by specific interactions of neurons with glial cells, operate as early and key targets of ASD. The pathology of children is often suspected by parents and communities and then confirmed by ensuing experiences. The final diagnoses of children and mature patients are then completed by the combination of neuropsychological (cognitive) tests and electro-/magneto-encephalography studies developed in specialized centers. ASD comorbidities, induced by processes such as anxieties, depressions, hyperactivities, and sleep defects, interact with and reinforce other brain diseases, especially schizophrenia. Advanced therapies, prescribed to children and adult patients for the control of ASD symptoms and disease, are based on the combination of well-known brain drugs with classical tools of neurologic and psychiatric practice. Overall, this review reports and discusses the advanced knowledge about the biological and medical properties of ASD.
Collapse
Affiliation(s)
- Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), 20132 Milan, Italy;
- Faculty of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Jacopo Meldolesi
- IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy
- CNR Institute of Neuroscience, Milano-Bicocca University, 20854 Vedano al Lambro, Italy
| |
Collapse
|
6
|
Vingan I, Phatarpekar S, Tung VSK, Hernández AI, Evgrafov OV, Alarcon JM. Spatially Resolved Transcriptomic Signatures of Hippocampal Subregions and Arc-Expressing Ensembles in Active Place Avoidance Memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573225. [PMID: 38260257 PMCID: PMC10802250 DOI: 10.1101/2023.12.30.573225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The rodent hippocampus is a spatially organized neuronal network that supports the formation of spatial and episodic memories. We conducted bulk RNA sequencing and spatial transcriptomics experiments to measure gene expression changes in the dorsal hippocampus following the recall of active place avoidance (APA) memory. Through bulk RNA sequencing, we examined the gene expression changes following memory recall across the functionally distinct subregions of the dorsal hippocampus. We found that recall induced differentially expressed genes (DEGs) in the CA1 and CA3 hippocampal subregions were enriched with genes involved in synaptic transmission and synaptic plasticity, while DEGs in the dentate gyrus (DG) were enriched with genes involved in energy balance and ribosomal function. Through spatial transcriptomics, we examined gene expression changes following memory recall across an array of spots encompassing putative memory-associated neuronal ensembles marked by the expression of the IEGs Arc, Egr1, and c-Jun. Within samples from both trained and untrained mice, the subpopulations of spatial transcriptomic spots marked by these IEGs were transcriptomically and spatially distinct from one another. DEGs detected between Arc+ and Arc- spots exclusively in the trained mouse were enriched in several memory-related gene ontology terms, including "regulation of synaptic plasticity" and "memory." Our results suggest that APA memory recall is supported by regionalized transcriptomic profiles separating the CA1 and CA3 from the DG, transcriptionally and spatially distinct IEG expressing spatial transcriptomic spots, and biological processes related to synaptic plasticity as a defining the difference between Arc+ and Arc- spatial transcriptomic spots.
Collapse
Affiliation(s)
- Isaac Vingan
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Shwetha Phatarpekar
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Victoria Sook Keng Tung
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - A. Iván Hernández
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| | - Oleg V. Evgrafov
- Institute of Genomics in Health, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- School of Graduates Studies, Program in Molecular and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Juan Marcos Alarcon
- School of Graduates Studies, Program in Neural and Behavioral Sciences, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Pathology, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
- The Robert F. Furchgott Center for Neural & Behavioral Science, State University of New York, Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
7
|
Shao Y, Cai Y, Chen T, Hao K, Luo B, Wang X, Guo W, Su X, Lv L, Yang Y, Li W. Impaired erythropoietin-producing hepatocellular B receptors signaling in the prefrontal cortex and hippocampus following maternal immune activation in male rats. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12863. [PMID: 37575018 PMCID: PMC10733575 DOI: 10.1111/gbb.12863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023]
Abstract
An environmental risk factor for schizophrenia (SZ) is maternal infection, which exerts longstanding effects on the neurodevelopment of offspring. Accumulating evidence suggests that synaptic disturbances may contribute to the pathology of the disease, but the underlying molecular mechanisms remain poorly understood. Erythropoietin-producing hepatocellular B (EphB) receptor signaling plays an important role in synaptic plasticity by regulating the formation and maturation of dendritic spines and regulating excitatory neurotransmission. We examined whether EphB receptors and downstream associated proteins are susceptible to environmental risk factors implicated in the etiology of synaptic disturbances in SZ. Using an established rodent model, which closely imitates the characteristics of SZ, we observed the behavioral performance and synaptic structure of male offspring in adolescence and early adulthood. We then analyzed the expression of EphB receptors and associated proteins in the prefrontal cortex and hippocampus. Maternal immune activation offspring showed significantly progressive cognitive impairment and pre-pulse inhibition deficits together with an increase in the expression of EphB2 receptors and NMDA receptor subunits. We also found changes in EphB receptor downstream signaling, in particular, a decrease in phospho-cofilin levels which may explain the reduced dendritic spine density. Besides, we found that the AMPA glutamate, another glutamate ionic receptor associated with cofilin, decreased significantly in maternal immune activation offspring. Thus, alterations in EphB signaling induced by immune activation during pregnancy may underlie disruptions in synaptic plasticity and function in the prefrontal cortex and hippocampus associated with behavioral and cognitive impairment. These findings may provide insight into the mechanisms underlying SZ.
Collapse
Affiliation(s)
- Yiqian Shao
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Yaqi Cai
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Tengfei Chen
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Keke Hao
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Binbin Luo
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Xiujuan Wang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
| | - Weiyun Guo
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Stem Cell and Biological Treatment Engineering Research Center of Henan, College of Life Science and TechnologyXinxiang Medical UniversityXinxiangChina
| | - Xi Su
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| | - Luxian Lv
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| | - Yongfeng Yang
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| | - Wenqiang Li
- Henan Mental HospitalThe Second Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
- Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of HenanXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental DisorderXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
8
|
Woelfle S, Pedro MT, Wagner J, Schön M, Boeckers TM. Expression profiles of the autism-related SHANK proteins in the human brain. BMC Biol 2023; 21:254. [PMID: 37953224 PMCID: PMC10641957 DOI: 10.1186/s12915-023-01712-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/25/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND SHANKs are major scaffolding proteins at postsynaptic densities (PSDs) in the central nervous system. Mutations in all three family members have been associated with neurodevelopmental disorders such as autism spectrum disorders (ASDs). Despite the pathophysiological importance of SHANK2 and SHANK3 mutations in humans, research on the expression of these proteins is mostly based on rodent model organisms. RESULTS In the present study, cellular and neuropil SHANK2 expression was analyzed by immunofluorescence (IF) staining of post mortem human brain tissue from four male individuals (19 brain regions). Mouse brains were analyzed in comparison to evaluate the degree of phylogenetic conservation. Furthermore, SHANK2 and SHANK3 isoform patterns were compared in human and mouse brain lysates. While isoform expression and subcellular distribution were largely conserved, differences in neuropil levels of SHANK2 were found by IF staining: Maximum expression was concordantly measured in the cerebellum; however, higher SHANK2 expression was detected in the human brainstem and thalamus when compared to mice. One of the lowest SHANK2 levels was found in the human amygdala, a moderately expressing region in mouse. Quantification of SHANK3 IF in mouse brains unveiled a distribution comparable to humans. CONCLUSIONS In summary, these data show that the overall expression pattern of SHANK is largely conserved in defined brain regions; however, differences do exist, which need to be considered in the translation of rodent studies. The summarized expression patterns of SHANK2 and SHANK3 should serve as a reference for future studies.
Collapse
Affiliation(s)
- Sarah Woelfle
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Maria T Pedro
- Department of Neurosurgery, Ulm University, Campus Günzburg, Lindenallee 2, 89312, Günzburg, Germany
| | - Jan Wagner
- Department of Neurology, Ulm University and Universitäts- and Rehabilitationskliniken Ulm, 89081, Ulm, Germany
| | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
- Deutsches Zentrum Für Neurodegenerative Erkrankungen, DZNE, Ulm Site, 89081, Ulm, Germany.
| |
Collapse
|
9
|
Rajkumar S, Böckers TM, Catanese A. Fast and efficient synaptosome isolation and post-synaptic density enrichment from hiPSC-motor neurons by biochemical sub-cellular fractionation. STAR Protoc 2023; 4:102061. [PMID: 36853677 PMCID: PMC9898788 DOI: 10.1016/j.xpro.2023.102061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/13/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
We describe here a time-efficient, in-house protocol for synaptosome isolation and enrichment of the post-synaptic density (PSD) from hiPSC-derived motor neurons. By using biochemical sub-cellular fractionation, the crude synaptosome is first isolated from the cytosol and is then further separated into the synaptic cytosol and the enriched PSD fraction. The protocol can also potentially be adapted to other hiPSC-derived neuronal types, with necessary changes made to cell seeding density and buffer volumes.
Collapse
Affiliation(s)
- Sandeep Rajkumar
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany.
| | - Tobias M Böckers
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm Site, 89081 Ulm, Germany
| | - Alberto Catanese
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm Site, 89081 Ulm, Germany.
| |
Collapse
|
10
|
Bauer HF, Delling JP, Bockmann J, Boeckers TM, Schön M. Development of sex- and genotype-specific behavioral phenotypes in a Shank3 mouse model for neurodevelopmental disorders. Front Behav Neurosci 2023; 16:1051175. [PMID: 36699652 PMCID: PMC9868822 DOI: 10.3389/fnbeh.2022.1051175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Individuals with a SHANK3-related neurodevelopmental disorder, also termed Phelan-McDermid syndrome or abbreviated as PMS, exhibit significant global developmental delay, language impairment, and muscular hypotonia. Also common are repetitive behaviors and altered social interactions, in line with a diagnosis of autism spectrum disorders. This study investigated the developmental aspect of autism-related behaviors and other phenotypes in a Shank3-transgenic mouse model. The animals underwent two sets of identical behavioral experiments, spanning motor skills, social and repetitive behavior, and cognition: baseline began at 5 weeks of age, corresponding to human adolescence, and the follow-up was initiated when aged 13 weeks, resembling early adulthood in humans. Interestingly, the animals displayed relatively stable phenotypes. Moreover, motor coordination and endurance were impaired, while muscle strength was unchanged. Surprisingly, the animals displayed only minor impairments in social behavior, but pronounced stereotypic and repetitive behaviors. Some behavioral tests indicated increased avoidance and anxiety. While spatial learning and memory were unchanged, knockout animals displayed slightly impaired cognitive flexibility. Female animals had similar abnormalities as males in the paradigms testing avoidance, anxiety, and cognition, but were less pathological in motor function and repetitive behavior. In all test paradigms, heterozygous Shank3 knockout animals had either no abnormal or a milder phenotype. Accurate characterization of animal models for genetic diseases is a prerequisite for understanding the pathophysiology. This is subsequently the basis for finding suitable and, ideally, translational biomarkers for therapeutic approaches and, thereby reducing the number of animals needed for preclinical trials.
Collapse
Affiliation(s)
- Helen Friedericke Bauer
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany,International Graduate School in Molecular Medicine Ulm, Ulm University, Ulm, Germany
| | | | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Tobias M. Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm Site, Ulm, Germany,*Correspondence: Tobias M. Boeckers,
| | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany,Michael Schön,
| |
Collapse
|
11
|
Atanasova E, Arévalo AP, Graf I, Zhang R, Bockmann J, Lutz AK, Boeckers TM. Immune activation during pregnancy exacerbates ASD-related alterations in Shank3-deficient mice. Mol Autism 2023; 14:1. [PMID: 36604742 PMCID: PMC9814193 DOI: 10.1186/s13229-022-00532-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/11/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is mainly characterized by deficits in social interaction and communication and repetitive behaviors. Known causes of ASD are mutations of certain risk genes like the postsynaptic protein SHANK3 and environmental factors including prenatal infections. METHODS To analyze the gene-environment interplay in ASD, we combined the Shank3Δ11-/- ASD mouse model with maternal immune activation (MIA) via an intraperitoneal injection of polyinosinic/polycytidylic acid (Poly I:C) on gestational day 12.5. The offspring of the injected dams was further analyzed for autistic-like behaviors and comorbidities followed by biochemical experiments with a focus on synaptic analysis. RESULTS We show that the two-hit mice exhibit excessive grooming and deficits in social behavior more prominently than the Shank3Δ11-/- mice. Interestingly, these behavioral changes were accompanied by an unexpected upregulation of postsynaptic density (PSD) proteins at excitatory synapses in striatum, hippocampus and prefrontal cortex. LIMITATIONS We found several PSD proteins to be increased in the two-hit mice; however, we can only speculate about possible pathways behind the worsening of the autistic phenotype in those mice. CONCLUSIONS With this study, we demonstrate that there is an interplay between genetic susceptibility and environmental factors defining the severity of ASD symptoms. Moreover, we show that a general misbalance of PSD proteins at excitatory synapses is linked to ASD symptoms, making this two-hit model a promising tool for the investigation of the complex pathophysiology of neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | - Ines Graf
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Rong Zhang
- Neuroscience Research Institute, Health Science Centre, Peking University, Peking, China
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm Site, Ulm, Germany.
| |
Collapse
|
12
|
Grabrucker AM. Non-Genetic Modifiers of Synaptic Plasticity and Neurotransmission in the Central Nervous System (CNS) in Health and Disease. Int J Mol Sci 2022; 23:ijms232012135. [PMID: 36292989 PMCID: PMC9603206 DOI: 10.3390/ijms232012135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Andreas M. Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland;
- Bernal Institute, University of Limerick, V94PH61 Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94PH61 Limerick, Ireland
| |
Collapse
|