1
|
McMath AL, Aguilar-Lopez M, Cannavale CN, Khan NA, Donovan SM. A systematic review on the impact of gastrointestinal microbiota composition and function on cognition in healthy infants and children. Front Neurosci 2023; 17:1171970. [PMID: 37389363 PMCID: PMC10306408 DOI: 10.3389/fnins.2023.1171970] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
Evidence from animal models or children with neurodevelopmental disorders has implicated the gut microbiome (GM) in neurocognitive development. However, even subclinical impairement of cognition can have negative consequences, as cognition serves as the foundation for skills necessary to succeed in school, vocation and socially. The present study aims to identify gut microbiome characteristics or changes in gut microbiome characteristics that consistently associate with cognitive outcomes in healthy, neurotypical infants and children. Of the 1,520 articles identified in the search, 23 were included in qualitative synthesis after applying exclusion criteria. Most studies were cross-sectional and focused on behavior or motor and language skills. Bifidobacterium, Bacteroides, Clostridia, Prevotella, and Roseburia were related to these aspects of cognition across several studies. While these results support the role of GM in cognitive development, higher quality studies focused on more complex cognition are needed to understand the extent to which the GM contributes to cognitive development.
Collapse
Affiliation(s)
- Arden L. McMath
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Miriam Aguilar-Lopez
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Corinne N. Cannavale
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Naiman A. Khan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois Urbana-Champaign, Champaign, IL, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, United States
| | - Sharon M. Donovan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
2
|
Abbah J, Vacher CM, Goldstein EZ, Li Z, Kundu S, Talbot B, Bhattacharya S, Hashimoto-Torii K, Wang L, Banerjee P, Scafidi J, Smith NA, Chew LJ, Gallo V. Oxidative Stress-Induced Damage to the Developing Hippocampus Is Mediated by GSK3β. J Neurosci 2022; 42:4812-4827. [PMID: 35589394 PMCID: PMC9188427 DOI: 10.1523/jneurosci.2389-21.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/10/2022] [Accepted: 04/17/2022] [Indexed: 11/30/2022] Open
Abstract
Neonatal brain injury renders the developing brain vulnerable to oxidative stress, leading to cognitive deficit. However, oxidative stress-induced damage to hippocampal circuits and the mechanisms underlying long-term changes in memory and learning are poorly understood. We used high oxygen tension or hyperoxia (HO) in neonatal mice of both sexes to investigate the role of oxidative stress in hippocampal damage. Perinatal HO induces reactive oxygen species and cell death, together with reduced interneuron maturation, inhibitory postsynaptic currents, and dentate progenitor proliferation. Postinjury interneuron stimulation surprisingly improved inhibitory activity and memory tasks, indicating reversibility. With decreased hippocampal levels of Wnt signaling components and somatostatin, HO aberrantly activated glycogen synthase kinase 3 β activity. Pharmacological inhibition or ablation of interneuron glycogen synthase kinase 3 β during HO challenge restored progenitor cell proliferation, interneuron development, inhibitory/excitatory balance, as well as hippocampal-dependent behavior. Biochemical targeting of interneuron function may benefit learning deficits caused by oxidative damage.SIGNIFICANCE STATEMENT Premature infants are especially vulnerable to oxidative stress, as their antioxidant defenses are underdeveloped. Indeed, high oxygen tension is associated with poor neurologic outcomes. Because of its sustained postnatal development and role in learning and memory, the hippocampus is especially vulnerable to oxidative damage in premature infants. However, the role of oxidative stress in the developing hippocampus has yet to be explored. With ever-rising rates of neonatal brain injury and no universally viable approach to maximize functional recovery, a better understanding of the mechanisms underlying neonatal brain injury is needed. Addressing this need, this study uses perinatal hyperoxia to study cognitive deficits, pathophysiology, and molecular mechanisms of oxidative damage in the developing hippocampus.
Collapse
Affiliation(s)
- Joseph Abbah
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Claire-Marie Vacher
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Evan Z Goldstein
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Zhen Li
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Srikanya Kundu
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Brooke Talbot
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Surajit Bhattacharya
- Center for Genetic Medicine, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Li Wang
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Payal Banerjee
- Bioinformatics Core, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Joseph Scafidi
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Nathan A Smith
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Li-Jin Chew
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010
| |
Collapse
|
3
|
Mandour DA, Tolba AM, El-Bestawy EM. Maternal exposure to the environmental pollutant "BDE-47" impairs the postnatal development of rat cerebellar cortex by modulating neuronal proliferation, synaptogenesis, NGF and BDNF pathways. Histol Histopathol 2022; 37:555-573. [PMID: 35191013 DOI: 10.14670/hh-18-441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
UNLABELLED 2,2',4,4'-Tetrabromodiphenyl ether (BDE-47) is an environmental contaminant that crosses the blood placental barrier and interferes with the homeostasis of fetal thyroid hormones. AIM OF WORK This study was designed to investigate the perinatal effect of BDE-47 exposure on the postnatal development of the rat cerebellar cortex. MATERIALS AND METHODS This study was carried out on 20 pregnant rats and 36 of their offspring. The pregnant rats were divided equally into control and BDE-47 treated mother groups; supplemented orally with BDE-47 (0.2 mg/kg/day from day 8 of gestation until the day of weaning). The offspring of both mother groups were subdivided, according to their developmental age, into three subgroups; PND14, PND21and PND42. SerumT3, T4 and TSH were assessed for dams and their offspring. Testing the motor coordination of the offspring via the rotarod test was conducted. Sections of the cerebellar cortex from offspring subgroups were stained with hematoxylin and eosin alongside immunohistochemical reactions and optical density of nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), proliferating cell nuclear antigen (PCNA) and synaptophysin (SYN). Also, the thickness of different layers of the cerebellar cortex was histomorphometrically measured. RESULTS BDE-47 treated mothers and their offspring subgroups showed a significant decrease in the serum free T3, T4 and increased TSH. The BDE-47 offspring displayed incoordination of the motor activity together with disturbed cytoarchitecture of the cerebellar cortical layers, and impaired migration of its germinative neuronal zones, particularly on PND14 and PND21. Moreover, these offspring displayed a decrease of the immune-expression and optical density of NGF, BDNF in the cerebellar cortical layers with impaired proliferation, and synaptogenesis. CONCLUSION Maternal exposure to BDE-47 during pregnancy and lactation effectuated a potential deleterious retarding effect on the postnatal development of the rat cerebellar cortex mostly via modulating neuronal proliferation, synaptogenesis, NGF and BDNF pathways secondary to its hypothyroid effect.
Collapse
Affiliation(s)
- Dalia A Mandour
- Department of Human Anatomy and Embryology, Faculty of medicine, Zagazig University, Zagazig, Egypt
| | - Asmaa M Tolba
- Department of Human Anatomy and Embryology, Faculty of medicine, Zagazig University, Zagazig, Egypt.
| | - Emtethal M El-Bestawy
- Department of Human Anatomy and Embryology, Faculty of medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
4
|
Sieghart W, Chiou LC, Ernst M, Fabjan J, M Savić M, Lee MT. α6-Containing GABA A Receptors: Functional Roles and Therapeutic Potentials. Pharmacol Rev 2022; 74:238-270. [PMID: 35017178 DOI: 10.1124/pharmrev.121.000293] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
GABAA receptors containing the α6 subunit are highly expressed in cerebellar granule cells and less abundantly in many other neuronal and peripheral tissues. Here, we for the first time summarize their importance for the functions of the cerebellum and the nervous system. The cerebellum is not only involved in motor control but also in cognitive, emotional, and social behaviors. α6βγ2 GABAA receptors located at cerebellar Golgi cell/granule cell synapses enhance the precision of inputs required for cerebellar timing of motor activity and are thus involved in cognitive processing and adequate responses to our environment. Extrasynaptic α6βδ GABAA receptors regulate the amount of information entering the cerebellum by their tonic inhibition of granule cells, and their optimal functioning enhances input filtering or contrast. The complex roles of the cerebellum in multiple brain functions can be compromised by genetic or neurodevelopmental causes that lead to a hypofunction of cerebellar α6-containing GABAA receptors. Animal models mimicking neuropsychiatric phenotypes suggest that compounds selectively activating or positively modulating cerebellar α6-containing GABAA receptors can alleviate essential tremor and motor disturbances in Angelman and Down syndrome as well as impaired prepulse inhibition in neuropsychiatric disorders and reduce migraine and trigeminal-related pain via α6-containing GABAA receptors in trigeminal ganglia. Genetic studies in humans suggest an association of the human GABAA receptor α6 subunit gene with stress-associated disorders. Animal studies support this conclusion. Neuroimaging and post-mortem studies in humans further support an involvement of α6-containing GABAA receptors in various neuropsychiatric disorders, pointing to a broad therapeutic potential of drugs modulating α6-containing GABAA receptors. SIGNIFICANCE STATEMENT: α6-Containing GABAA receptors are abundantly expressed in cerebellar granule cells, but their pathophysiological roles are widely unknown, and they are thus out of the mainstream of GABAA receptor research. Anatomical and electrophysiological evidence indicates that these receptors have a crucial function in neuronal circuits of the cerebellum and the nervous system, and experimental, genetic, post-mortem, and pharmacological studies indicate that selective modulation of these receptors offers therapeutic prospects for a variety of neuropsychiatric disorders and for stress and its consequences.
Collapse
Affiliation(s)
- Werner Sieghart
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Lih-Chu Chiou
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Margot Ernst
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Jure Fabjan
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Miroslav M Savić
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| | - Ming Tatt Lee
- Center for Brain Research, Department of Molecular Neurosciences (W.S.), and Center for Brain Research, Department of Pathobiology of the Nervous System (M.E., J.F.), Medical University Vienna, Vienna, Austria; Graduate Institute of Pharmacology (L.-C.C., M.T.L.), and Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan (L.-C.C., M.T.L.); Faculty of Pharmacy, Department of Pharmacology, University of Belgrade, Belgrade, Serbia (M.M.S.); Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia (M.T.L.); and Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan (L.-C.C.)
| |
Collapse
|
5
|
August PM, Klein CP, Grings M, Sagini JP, Rodrigues PIDL, Stocher DP, Stone V, Silva YD, Couto PRG, Salomon TB, Benfato MDS, Leipnitz G, Matté C. Maternal polyphenol intake impairs cerebellar redox homeostasis in newborn rats. Nutr Neurosci 2021; 25:2066-2076. [PMID: 34076555 DOI: 10.1080/1028415x.2021.1933330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Polyphenols are compounds found in plants that have been extensively studied due to the health benefits of its consumption in adulthood. Meanwhile, recent evidence suggests that polyphenol consumption during pregnancy may not be safe for the fetus. OBJECTIVE The goal of this study was to evaluate the effect of naringenin supplementation during pregnancy on brain redox homeostasis and mitochondrial activity of the newborn rat. METHODS Adult female Wistar rats were divided into two groups: (1) vehicle (1 mL/Kg p.o.) or (2) naringenin (50 mg/Kg p.o.). Naringenin was administered once a day during pregnancy. The offspring were euthanized on postnatal day 7, as well the dams, and brain regions were dissected. RESULTS The offspring cerebellum was the most affected region, presenting increased activity of the mitochondrial electron transport system, allied to increased reactive species levels, lipid peroxidation, and glutathione concentration. The nitric oxide levels suffered structure-dependent alteration, with decreased levels in the pups' cerebellum and increased in the hippocampus. The offspring parietal cortex was not affected, as well as the parameters evaluated in the dams' brains. CONCLUSION Maternal consumption of naringenin alters offspring cerebellar redox homeostasis, which could be related to adverse effects on the motor and cognitive development in the descendants.
Collapse
Affiliation(s)
- Pauline Maciel August
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Caroline Peres Klein
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mateus Grings
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - João Pedro Sagini
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Daniela Pereira Stocher
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Vinicius Stone
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Yasmini Dandara Silva
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pablo Ribeiro Gonçalves Couto
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tiago Boeira Salomon
- Programa de Pós-graduação em Biologia Molecular e Celular, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mara da Silveira Benfato
- Programa de Pós-graduação em Biologia Molecular e Celular, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristiane Matté
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências Biológicas: Fisiologia, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
6
|
Ren BX, Huen I, Wu ZJ, Wang H, Duan MY, Guenther I, Bhanu Prakash KN, Tang FR. Early postnatal irradiation-induced age-dependent changes in adult mouse brain: MRI based characterization. BMC Neurosci 2021; 22:28. [PMID: 33882822 PMCID: PMC8061041 DOI: 10.1186/s12868-021-00635-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
Background Brain radiation exposure, in particular, radiotherapy, can induce cognitive impairment in patients, with significant effects persisting for the rest of their life. However, the main mechanisms leading to this adverse event remain largely unknown. A study of radiation-induced injury to multiple brain regions, focused on the hippocampus, may shed light on neuroanatomic bases of neurocognitive impairments in patients. Hence, we irradiated BALB/c mice (male and female) at postnatal day 3 (P3), day 10 (P10), and day 21 (P21) and investigated the long-term radiation effect on brain MRI changes and hippocampal neurogenesis. Results We found characteristic brain volume reductions in the hippocampus, olfactory bulbs, the cerebellar hemisphere, cerebellar white matter (WM) and cerebellar vermis WM, cingulate, occipital and frontal cortices, cerebellar flocculonodular WM, parietal region, endopiriform claustrum, and entorhinal cortex after irradiation with 5 Gy at P3. Irradiation at P10 induced significant volume reduction in the cerebellum, parietal region, cingulate region, and olfactory bulbs, whereas the reduction of the volume in the entorhinal, parietal, insular, and frontal cortices was demonstrated after irradiation at P21. Immunohistochemical study with cell division marker Ki67 and immature marker doublecortin (DCX) indicated the reduced cell division and genesis of new neurons in the subgranular zone of the dentate gyrus in the hippocampus after irradiation at all three postnatal days, but the reduction of total granule cells in the stratum granulosun was found after irradiation at P3 and P10. Conclusions The early life radiation exposure during different developmental stages induces varied brain pathophysiological changes which may be related to the development of neurological and neuropsychological disorders later in life.
Collapse
Affiliation(s)
- Bo Xu Ren
- Department of Medical Imaging, School of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
| | - Isaac Huen
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), Singapore, 138667, Singapore
| | - Zi Jun Wu
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Wang
- Radiation Physiology Laboratory, Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, 1 CREATE Way #04-01, Singapore, 138602, Singapore
| | - Meng Yun Duan
- Department of Medical Imaging, School of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023, Hubei, China
| | - Ilonka Guenther
- Comparative Medicine, Centre for Life Sciences (CeLS), National University of Singapore, #05-02, 28 Medical Drive, Singapore, 117456, Singapore
| | - K N Bhanu Prakash
- Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), Singapore, 138667, Singapore.
| | - Feng Ru Tang
- Radiation Physiology Laboratory, Nuclear Research and Safety Initiative, National University of Singapore, CREATE Tower, 1 CREATE Way #04-01, Singapore, 138602, Singapore.
| |
Collapse
|
7
|
Modi ME, Sahin M. A unified circuit for social behavior. Neurobiol Learn Mem 2019; 165:106920. [PMID: 30149055 PMCID: PMC6387844 DOI: 10.1016/j.nlm.2018.08.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/04/2018] [Accepted: 08/14/2018] [Indexed: 12/23/2022]
Abstract
Recent advances in circuit manipulation technologies have enabled the association of distinct neural circuits with complex social behaviors. The brain areas identified through historical anatomical characterizations as mediators of sexual and parental behaviors can now be functionally linked to adult social behaviors within a unified circuit. In vivo electrophysiology, optogenetics and chemogenetics have been used to follow the processing of social sensory stimuli from perception by the olfactory system to valence detection by the amygdala and mesolimbic dopamine system to integration by the cerebral and cerebellar cortices under modulation of hypothalamic neuropeptides. Further, these techniques have been able to identify the distinct functional changes induced by social as opposed to non-social stimuli. Together this evidence suggests that there is a distinct, functionally coupled circuit that is selectively activated by social stimuli. A unified social circuit provides a new framework against which synaptopathic autism related mutations can be considered and novel pharmacotherapeutic strategies can be developed.
Collapse
Affiliation(s)
- Meera E Modi
- Translational Neuroscience Center, F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, United States
| | - Mustafa Sahin
- Translational Neuroscience Center, F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, United States.
| |
Collapse
|
8
|
Tate KM, Munson JM. Assessing drug response in engineered brain microenvironments. Brain Res Bull 2019; 150:21-34. [PMID: 31054318 PMCID: PMC6754984 DOI: 10.1016/j.brainresbull.2019.04.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/26/2019] [Accepted: 04/21/2019] [Indexed: 12/11/2022]
Abstract
Tissue engineered systems are important models for the testing and discovery of therapeutics against a number of diseases. The use of these models in vitro can expand both our understanding of the mechanisms behind disease and allow for higher throughput and personalized modeling of therapeutic response. Over the past decade there has been an explosion of models of neurological disorders that can be used in vitro to study new therapies against devastating neurodegenerative, neurodevelopmental, and neuro-oncological disease. These models span several types of engineered microenvironments which are produced using microfluidic devices, microtissue technology and/or the incorporation of biomaterial scaffolds to model neurological conditions such as; Alzheimer's disease, idiopathic autism, Parkinson's disease, Zika-induced microcephaly and neoplasms. Using engineered brain microenvironments, therapeutics can be tested in more physiologically relevant ways leading to new knowledge of the underlying causes and interactions occurring at the tissue level. However, much is still left to learn and model within these systems to make them truly valuable in the discovery and testing of novel therapies. Here we review the current state of the art of engineered brain microenvironments being used specifically to screen and test new therapeutic strategies and discuss the current benefits and limitations that still exist.
Collapse
Affiliation(s)
- Kinsley M Tate
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jennifer M Munson
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.
| |
Collapse
|
9
|
Valdés-Hernández PA, Bae J, Song Y, Sumiyoshi A, Aubert-Vázquez E, Riera JJ. Validating Non-invasive EEG Source Imaging Using Optimal Electrode Configurations on a Representative Rat Head Model. Brain Topogr 2019; 32:599-624. [PMID: 27026168 DOI: 10.1007/s10548-016-0484-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 03/05/2016] [Indexed: 12/20/2022]
Abstract
The curtain of technical limitations impeding rat multichannel non-invasive electroencephalography (EEG) has risen. Given the importance of this preclinical model, development and validation of EEG source imaging (ESI) is essential. We investigate the validity of well-known human ESI methodologies in rats which individual tissue geometries have been approximated by those extracted from an MRI template, leading also to imprecision in electrode localizations. With the half and fifth sensitivity volumes we determine both the theoretical minimum electrode separation for non-redundant scalp EEG measurements and the electrode sensitivity resolution, which vary over the scalp because of the head geometry. According to our results, electrodes should be at least ~3 to 3.5 mm apart for an optimal configuration. The sensitivity resolution is generally worse for electrodes at the boundaries of the scalp measured region, though, by analogy with human montages, concentrates the sensitivity enough to localize sources. Cramér-Rao lower bounds of source localization errors indicate it is theoretically possible to achieve ESI accuracy at the level of anatomical structures, such as the stimulus-specific somatosensory areas, using the template. More validation for this approximation is provided through the comparison between the template and the individual lead field matrices, for several rats. Finally, using well-accepted inverse methods, we demonstrate that somatosensory ESI is not only expected but also allows exploring unknown phenomena related to global sensory integration. Inheriting the advantages and pitfalls of human ESI, rat ESI will boost the understanding of brain pathophysiological mechanisms and the evaluation of ESI methodologies, new pharmacological treatments and ESI-based biomarkers.
Collapse
Affiliation(s)
- Pedro A Valdés-Hernández
- Neuroimaging Department, Cuban Neuroscience Center, Havana, Cuba
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Jihye Bae
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA
| | - Yinchen Song
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA
| | - Akira Sumiyoshi
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | - Jorge J Riera
- Department of Biomedical Engineering, Florida International University, Miami, FL, USA.
| |
Collapse
|
10
|
Toledo A, Lang F, Doengi M, Morrison H, Stein V, Baader SL. Merlin modulates process outgrowth and synaptogenesis in the cerebellum. Brain Struct Funct 2019; 224:2121-2142. [PMID: 31165301 DOI: 10.1007/s00429-019-01897-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
Neurofibromatosis type 2 (NF2) patients are prone to develop glial-derived tumors in the peripheral and central nervous system (CNS). The Nf2 gene product -Merlin is not only expressed in glia, but also in neurons of the CNS, where its function still remains elusive. Here, we show that cerebellar Purkinje cells (PCs) of isoform-specific Merlin-deficient mice were innervated by smaller vGluT2-positive clusters at presynaptic terminals than those of wild-type mice. This was paralleled by a reduction in frequency and amplitude of miniature excitatory postsynaptic currents (mEPSC). On the contrary, in conditional transgenic mice in which Merlin expression was specifically ablated in PCs (L7Cre;Nf2fl/fl), we found enlarged vGluT2-positive clusters in their presynaptic buttons together with increased amplitudes of miniature postsynaptic currents. The presynaptic terminals of these PCs innervating neurons of the deep cerebellar nuclei were also enlarged. When exploring mice with Merlin-deficient granule cells (GCs) (Math1Cre;Nf2fl/fl), we found cerebellar extracts to contain higher amounts of vGluT1 present in parallel fiber terminals. In parallel, mEPSC frequency was increased in Math1Cre;Nf2fl/fl mice. On the contrary, VGluT2 clusters in cerebellar glomeruli composed of NF2-deficient presynaptic Mossy fiber terminals and NF2-deficient postsynaptic GC were reduced in size as shown for isoform-specific knockout mice. These changes in Math1Cre;Nf2fl/fl-deficient mice were paralleled by an increased activation of Rac1-Cofilin signaling which is known to impact on cytoskeletal reorganization and synapse formation. Consistent with the observed synaptic alterations in these transgenic mice, we observed altered ultrasonic vocalization, which is known to rely on proper cerebellar function. No gross morphological changes or motor coordination deficits were observed in any of these transgenic mice. We therefore conclude that Merlin does not regulate overall cerebellar development, but impacts on pre- and post-synaptic terminal organization.
Collapse
Affiliation(s)
- A Toledo
- Institute of Anatomy, Anatomy and Cell Biology, Bonn University, 53115, Bonn, Germany
| | - F Lang
- Institute of Anatomy, Anatomy and Cell Biology, Bonn University, 53115, Bonn, Germany
| | - M Doengi
- Institute of Physiology II, Bonn University, 53115, Bonn, Germany
| | - H Morrison
- Leibniz Institute for Age Research, Fritz Lipmann Institute, 07745, Jena, Germany
| | - V Stein
- Institute of Physiology II, Bonn University, 53115, Bonn, Germany
| | - S L Baader
- Institute of Anatomy, Anatomy and Cell Biology, Bonn University, 53115, Bonn, Germany.
| |
Collapse
|
11
|
Miquel M, Nicola SM, Gil-Miravet I, Guarque-Chabrera J, Sanchez-Hernandez A. A Working Hypothesis for the Role of the Cerebellum in Impulsivity and Compulsivity. Front Behav Neurosci 2019; 13:99. [PMID: 31133834 PMCID: PMC6513968 DOI: 10.3389/fnbeh.2019.00099] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/23/2019] [Indexed: 12/27/2022] Open
Abstract
Growing evidence associates cerebellar abnormalities with several neuropsychiatric disorders in which compulsive symptomatology and impulsivity are part of the disease pattern. Symptomatology of autism, addiction, obsessive-compulsive (OCD), and attention deficit/hyperactivity (ADHD) disorders transcends the sphere of motor dysfunction and essentially entails integrative processes under control of prefrontal-thalamic-cerebellar loops. Patients with brain lesions affecting the cortico-striatum thalamic circuitry and the cerebellum indeed exhibit compulsive symptoms. Specifically, lesions of the posterior cerebellar vermis cause affective dysregulation and deficits in executive function. These deficits may be due to impairment of one of the main functions of the cerebellum, implementation of forward internal models of the environment. Actions that are independent of internal models may not be guided by predictive relationships or a mental representation of the goal. In this review article, we explain how this deficit might affect executive functions. Additionally, regionalized cerebellar lesions have been demonstrated to impair other brain functions such as the emergence of habits and behavioral inhibition, which are also altered in compulsive disorders. Similar to the infralimbic cortex, clinical studies and research in animal models suggest that the cerebellum is not required for learning goal-directed behaviors, but it is critical for habit formation. Despite this accumulating data, the role of the cerebellum in compulsive symptomatology and impulsivity is still a matter of discussion. Overall, findings point to a modulatory function of the cerebellum in terminating or initiating actions through regulation of the prefrontal cortices. Specifically, the cerebellum may be crucial for restraining ongoing actions when environmental conditions change by adjusting prefrontal activity in response to the new external and internal stimuli, thereby promoting flexible behavioral control. We elaborate on this explanatory framework and propose a working hypothesis for the involvement of the cerebellum in compulsive and impulsive endophenotypes.
Collapse
Affiliation(s)
- Marta Miquel
- Área de Psicobiología, School of Health Science, Universitat Jaume I, Castellón de la Plana, Spain
| | - Saleem M Nicola
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Isis Gil-Miravet
- Área de Psicobiología, School of Health Science, Universitat Jaume I, Castellón de la Plana, Spain
| | - Julian Guarque-Chabrera
- Área de Psicobiología, School of Health Science, Universitat Jaume I, Castellón de la Plana, Spain
| | - Aitor Sanchez-Hernandez
- Área de Psicobiología, School of Health Science, Universitat Jaume I, Castellón de la Plana, Spain
| |
Collapse
|
12
|
Regional and sex-dependent alterations in Purkinje cell density in the valproate mouse model of autism. Neuroreport 2019; 30:82-88. [PMID: 30461560 DOI: 10.1097/wnr.0000000000001164] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neuropathological and neuroimaging studies indicate a decrease in Purkinje cell (PC) density in the cerebellum of autistic patients and rodent models of autism. Autism is far more prevalent in males than females, and sex-specific properties of PCs have been reported recently. We investigated the differential sensitivity of PCs in the valproate acid (VPA) mouse model of autism by estimating the linear density of PCs immununolabelled with calbindin in the cerebellum of males and females. Whereas prenatal VPA treatment surprisingly increased PC linear density in both sexes 13 days after birth (P13), it significantly reduced the linear density of PCs in the cerebellum of 40-day-old (P40) males, but not females. In males, PC loss was more pronounced in the posterior part of the cerebellum and was significant in the VIth, VIIth, IXth and paramedian lobules. In females, PC loss was restricted to the paramedian lobule. These results suggest that this sex-specific sensitivity of PCs to VPA may contribute towards the motor disturbances and behavioural abnormalities observed in autism.
Collapse
|
13
|
Deshmukh A, Leichner J, Bae J, Song Y, Valdés-Hernández PA, Lin WC, Riera JJ. Histological Characterization of the Irritative Zones in Focal Cortical Dysplasia Using a Preclinical Rat Model. Front Cell Neurosci 2018; 12:52. [PMID: 29867355 PMCID: PMC5968101 DOI: 10.3389/fncel.2018.00052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/15/2018] [Indexed: 12/19/2022] Open
Abstract
Current clinical practice in focal epilepsy involves brain source imaging (BSI) to localize brain areas where from interictal epileptiform discharges (IEDs) emerge. These areas, named irritative zones, have been useful to define candidate seizures-onset zones during pre-surgical workup. Since human histological data are mostly available from final resected zones, systematic studies characterizing pathophysiological mechanisms and abnormal molecular/cellular substrates in irritative zones—independent of them being epileptogenic—are challenging. Combining BSI and histological analysis from all types of irritative zones is only possible through the use of preclinical animal models. Here, we recorded 32-channel spontaneous electroencephalographic data from rats that have focal cortical dysplasia (FCD) and chronic seizures. BSI for different IED subtypes was performed using the methodology presented in Bae et al. (2015). Post-mortem brain sections containing irritative zones were stained to quantify anatomical, functional, and inflammatory biomarkers specific for epileptogenesis, and the results were compared with those obtained using the contralateral healthy brain tissue. We found abnormal anatomical structures in all irritative zones (i.e., larger neuronal processes, glioreactivity, and vascular cuffing) and larger expressions for neurotransmission (i.e., NR2B) and inflammation (i.e., ILβ1, TNFα and HMGB1). We conclude that irritative zones in this rat preclinical model of FCD comprise abnormal tissues disregarding whether they are actually involved in icto-genesis or not. We hypothesize that seizure perpetuation happens gradually; hence, our results could support the use of IED-based BSI for the early diagnosis and preventive treatment of potential epileptic foci. Further verifications in humans are yet needed.
Collapse
Affiliation(s)
- Abhay Deshmukh
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Jared Leichner
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Jihye Bae
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Yinchen Song
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Pedro A Valdés-Hernández
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Wei-Chiang Lin
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Jorge J Riera
- Neuronal Mass Dynamics Laboratory, Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| |
Collapse
|
14
|
Luk B, Veeraragavan S, Engevik M, Balderas M, Major A, Runge J, Luna RA, Versalovic J. Postnatal colonization with human "infant-type" Bifidobacterium species alters behavior of adult gnotobiotic mice. PLoS One 2018; 13:e0196510. [PMID: 29763437 PMCID: PMC5953436 DOI: 10.1371/journal.pone.0196510] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
Accumulating studies have defined a role for the intestinal microbiota in modulation of host behavior. Research using gnotobiotic mice emphasizes that early microbial colonization with a complex microbiota (conventionalization) can rescue some of the behavioral abnormalities observed in mice that grow to adulthood completely devoid of bacteria (germ-free mice). However, the human infant and adult microbiomes vary greatly, and effects of the neonatal microbiome on neurodevelopment are currently not well understood. Microbe-mediated modulation of neural circuit patterning in the brain during neurodevelopment may have significant long-term implications that we are only beginning to appreciate. Modulation of the host central nervous system by the early-life microbiota is predicted to have pervasive and lasting effects on brain function and behavior. We sought to replicate this early microbe-host interaction by colonizing gnotobiotic mice at the neonatal stage with a simplified model of the human infant gut microbiota. This model consortium consisted of four “infant-type” Bifidobacterium species known to be commensal members of the human infant microbiota present in high abundance during postnatal development. Germ-free mice and mice neonatally-colonized with a complex, conventional murine microbiota were used for comparison. Motor and non-motor behaviors of the mice were tested at 6–7 weeks of age, and colonization patterns were characterized by 16S ribosomal RNA gene sequencing. Adult germ-free mice were observed to have abnormal memory, sociability, anxiety-like behaviors, and motor performance. Conventionalization at the neonatal stage rescued these behavioral abnormalities, and mice colonized with Bifidobacterium spp. also exhibited important behavioral differences relative to the germ-free controls. The ability of Bifidobacterium spp. to improve the recognition memory of both male and female germ-free mice was a prominent finding. Together, these data demonstrate that the early-life gut microbiome, and human “infant-type” Bifidobacterium species, affect adult behavior in a strongly sex-dependent manner, and can selectively recapitulate the results observed when mice are colonized with a complex microbiota.
Collapse
Affiliation(s)
- Berkley Luk
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Surabi Veeraragavan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Melinda Engevik
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Miriam Balderas
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Angela Major
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Jessica Runge
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Ruth Ann Luna
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas, United States of America
| | - James Versalovic
- Department of Pathology, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
15
|
Du Y, Fryer SL, Fu Z, Lin D, Sui J, Chen J, Damaraju E, Mennigen E, Stuart B, Loewy RL, Mathalon DH, Calhoun VD. Dynamic functional connectivity impairments in early schizophrenia and clinical high-risk for psychosis. Neuroimage 2017; 180:632-645. [PMID: 29038030 DOI: 10.1016/j.neuroimage.2017.10.022] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 01/14/2023] Open
Abstract
Individuals at clinical high-risk (CHR) for psychosis are characterized by attenuated psychotic symptoms. Only a minority of CHR individuals convert to full-blown psychosis. Therefore, there is a strong interest in identifying neurobiological abnormalities underlying the psychosis risk syndrome. Dynamic functional connectivity (DFC) captures time-varying connectivity over short time scales, and has the potential to reveal complex brain functional organization. Based on resting-state functional magnetic resonance imaging (fMRI) data from 70 healthy controls (HCs), 53 CHR individuals, and 58 early illness schizophrenia (ESZ) patients, we applied a novel group information guided ICA (GIG-ICA) to estimate inherent connectivity states from DFC, and then investigated group differences. We found that ESZ patients showed more aberrant connectivities and greater alterations than CHR individuals. Results also suggested that disease-related connectivity states occurred in CHR and ESZ groups. Regarding the dominant state with the highest contribution to dynamic connectivity, ESZ patients exhibited greater impairments than CHR individuals primarily in the cerebellum, frontal cortex, thalamus and temporal cortex, while CHR and ESZ populations shared common aberrances mainly in the supplementary motor area, parahippocampal gyrus and postcentral cortex. CHR-specific changes were also found in the connections between the superior frontal gyrus and calcarine cortex in the dominant state. Our findings suggest that CHR individuals generally show an intermediate functional connectivity pattern between HCs and SZ patients but also have unique connectivity alterations.
Collapse
Affiliation(s)
- Yuhui Du
- The Mind Research Network, Albuquerque, NM, USA; School of Computer & Information Technology, Shanxi University, Taiyuan, China.
| | - Susanna L Fryer
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, USA; The Mental Health Service, San Francisco VA Healthcare System, San Francisco, CA, USA
| | - Zening Fu
- The Mind Research Network, Albuquerque, NM, USA
| | | | - Jing Sui
- The Mind Research Network, Albuquerque, NM, USA; Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jiayu Chen
- The Mind Research Network, Albuquerque, NM, USA
| | | | - Eva Mennigen
- The Mind Research Network, Albuquerque, NM, USA; Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM, USA
| | - Barbara Stuart
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, USA
| | - Rachel L Loewy
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, USA
| | - Daniel H Mathalon
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, USA; The Mental Health Service, San Francisco VA Healthcare System, San Francisco, CA, USA
| | - Vince D Calhoun
- The Mind Research Network, Albuquerque, NM, USA; Department of Electrical and Computer Engineering, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
16
|
Koning IV, Tielemans MJ, Hoebeek FE, Ecury-Goossen GM, Reiss IKM, Steegers-Theunissen RPM, Dudink J. Impacts on prenatal development of the human cerebellum: a systematic review. J Matern Fetal Neonatal Med 2016; 30:2461-2468. [PMID: 27806674 DOI: 10.1080/14767058.2016.1253060] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE The cerebellum is essential for normal neurodevelopment and is particularly susceptible for intra-uterine disruptions. Although some causal prenatal exposures have been identified, the origin of neurodevelopmental disorders remains mostly unclear. Therefore, a systematic literature search was conducted to provide an overview of parental environmental exposures and intrinsic factors influencing prenatal cerebellar growth and development in humans. MATERIALS AND METHODS The literature search was limited to human studies in the English language and was conducted in Embase, Medline, Cochrane, Web of Science, Pubmed and GoogleScholar. Eligible studies were selected by three independent reviewers and study quality was scored by two independent reviewers. RESULTS The search yielded 3872 articles. We found 15 eligible studies reporting associations between cerebellar development and maternal smoking (4), use of alcohol (3), in vitro fertilization mediums (1), mercury (1), mifepristone (2), aminopropionitriles (1), ethnicity (2) and cortisol levels (1). No studies reported on paternal factors. CONCLUSIONS Current literature on associations between parental environmental exposures, intrinsic factors and human cerebellar development is scarce. Yet, this systematic review provided an essential overview of human studies demonstrating the vulnerability of the cerebellum to the intra-uterine environment.
Collapse
Affiliation(s)
- Irene V Koning
- a Department of Obstetrics and Gynecology , Erasmus MC University Medical Center , Rotterdam , The Netherlands.,b Department of Pediatrics , Subdivision of Neonatology, Sophia Children's Hospital , Rotterdam , The Netherlands
| | | | - Freek E Hoebeek
- d Department of Neuroscience , Erasmus MC University Medical Center , Rotterdam , The Netherlands , and
| | - Ginette M Ecury-Goossen
- b Department of Pediatrics , Subdivision of Neonatology, Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Irwin K M Reiss
- b Department of Pediatrics , Subdivision of Neonatology, Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Regine P M Steegers-Theunissen
- a Department of Obstetrics and Gynecology , Erasmus MC University Medical Center , Rotterdam , The Netherlands.,b Department of Pediatrics , Subdivision of Neonatology, Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Jeroen Dudink
- b Department of Pediatrics , Subdivision of Neonatology, Sophia Children's Hospital , Rotterdam , The Netherlands.,e Department of Neonatology , Wilhelmina Children's Hospital, University Medical Center Utrecht , Utrecht , The Netherlands
| |
Collapse
|
17
|
The Purkinje cell as a model of synaptogenesis and synaptic specificity. Brain Res Bull 2016; 129:12-17. [PMID: 27721030 DOI: 10.1016/j.brainresbull.2016.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/28/2016] [Accepted: 10/05/2016] [Indexed: 01/03/2023]
Abstract
Since the groundbreaking work of Ramon y Cajal, the cerebellar Purkinje cell has always represented an ideal model for studying the organization, development and function of synaptic circuits. Purkinje cells receive distinct types of glutamatergic and GABAergic synapses, each characterized by exquisite sub-cellular and molecular specificity. The formation and refinement of these connections results from a temporally-regulated sequence of events that involves molecular interactions between distinct sets of secreted and surface proteins, as well as activity-dependent competition between converging inputs. Insights into the mechanisms controlling synaptic specificity in Purkinje cells may help understand synapse development also in other brain regions and disclose circuit abnormalities that underlie neurodevelopmental disorders.
Collapse
|
18
|
Alexander MS, Gasperini MJ, Tsai PT, Gibbs DE, Spinazzola JM, Marshall JL, Feyder MJ, Pletcher MT, Chekler ELP, Morris CA, Sahin M, Harms JF, Schmidt CJ, Kleiman RJ, Kunkel LM. Reversal of neurobehavioral social deficits in dystrophic mice using inhibitors of phosphodiesterases PDE5A and PDE9A. Transl Psychiatry 2016; 6:e901. [PMID: 27676442 PMCID: PMC5048211 DOI: 10.1038/tp.2016.174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022] Open
Abstract
Duchenne muscular dystrophy is caused by mutations in the DYSTROPHIN gene. Although primarily associated with muscle wasting, a significant portion of patients (approximately 25%) are also diagnosed with autism spectrum disorder. We describe social behavioral deficits in dystrophin-deficient mice and present evidence of cerebellar deficits in cGMP production. We demonstrate therapeutic potential for selective inhibitors of the cGMP-specific PDE5A and PDE9A enzymes to restore social behaviors in dystrophin-deficient mice.
Collapse
Affiliation(s)
- M S Alexander
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
- The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - M J Gasperini
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - P T Tsai
- The F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - D E Gibbs
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - J M Spinazzola
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
| | - J L Marshall
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - M J Feyder
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - M T Pletcher
- Rare Disease Research Unit, Pfizer, Cambridge, MA, USA
| | - E L P Chekler
- Rare Disease Research Unit, Pfizer, Cambridge, MA, USA
| | - C A Morris
- Rare Disease Research Unit, Pfizer, Cambridge, MA, USA
| | - M Sahin
- The F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - J F Harms
- Neuroscience Research Unit, Pfizer Global Research and Development, Cambridge, MA, USA
| | - C J Schmidt
- Neuroscience Research Unit, Pfizer Global Research and Development, Cambridge, MA, USA
| | - R J Kleiman
- The F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - L M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Departments of Pediatrics and Genetics, Harvard Medical School, Boston, MA, USA
- The Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- The Manton Center for Orphan Diseases, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
19
|
Del Rio-Bermudez C, Plumeau AM, Sattler NJ, Sokoloff G, Blumberg MS. Spontaneous activity and functional connectivity in the developing cerebellorubral system. J Neurophysiol 2016; 116:1316-27. [PMID: 27385801 DOI: 10.1152/jn.00461.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
The development of the cerebellar system depends in part on the emergence of functional connectivity in its input and output pathways. Characterization of spontaneous activity within these pathways provides insight into their functional status in early development. In the present study we recorded extracellular activity from the interpositus nucleus (IP) and its primary downstream target, the red nucleus (RN), in unanesthetized rats at postnatal days 8 (P8) and P12, a period of dramatic change in cerebellar circuitry. The two structures exhibited state-dependent activity patterns and age-related changes in rhythmicity and overall firing rate. Importantly, sensory feedback (i.e., reafference) from myoclonic twitches (spontaneous, self-generated movements that are produced exclusively during active sleep) drove neural activity in the IP and RN at both ages. Additionally, anatomic tracing confirmed the presence of cerebellorubral connections as early as P8. Finally, inactivation of the IP and adjacent nuclei using the GABAA receptor agonist muscimol caused a substantial decrease in neural activity in the contralateral RN at both ages, as well as the disappearance of rhythmicity; twitch-related activity in the RN, however, was preserved after IP inactivation, indicating that twitch-related reafference activates the two structures in parallel. Overall, the present findings point to the contributions of sleep-related spontaneous activity to the development of cerebellar networks.
Collapse
Affiliation(s)
| | - Alan M Plumeau
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
| | - Nicholas J Sattler
- Department of Psychological & Brain Sciences, University of Iowa, Iowa City, Iowa
| | - Greta Sokoloff
- Department of Psychological & Brain Sciences, University of Iowa, Iowa City, Iowa; DeLTA Center, University of Iowa, Iowa City, Iowa
| | - Mark S Blumberg
- Department of Psychological & Brain Sciences, University of Iowa, Iowa City, Iowa; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa; Department of Biology, University of Iowa, Iowa City, Iowa; and DeLTA Center, University of Iowa, Iowa City, Iowa
| |
Collapse
|
20
|
Slotkin TA, Skavicus S, Seidler FJ. Prenatal drug exposures sensitize noradrenergic circuits to subsequent disruption by chlorpyrifos. Toxicology 2015; 338:8-16. [PMID: 26419632 DOI: 10.1016/j.tox.2015.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 09/11/2015] [Accepted: 09/24/2015] [Indexed: 11/20/2022]
Abstract
We examined whether nicotine or dexamethasone, common prenatal drug exposures, sensitize the developing brain to chlorpyrifos. We gave nicotine to pregnant rats throughout gestation at a dose (3mg/kg/day) producing plasma levels typical of smokers; offspring were then given chlorpyrifos on postnatal days 1-4, at a dose (1mg/kg) that produces minimally-detectable inhibition of brain cholinesterase activity. In a parallel study, we administered dexamethasone to pregnant rats on gestational days 17-19 at a standard therapeutic dose (0.2mg/kg) used in the management of preterm labor, followed by postnatal chlorpyrifos. We evaluated cerebellar noradrenergic projections, a known target for each agent, and contrasted the effects with those in the cerebral cortex. Either drug augmented the effect of chlorpyrifos, evidenced by deficits in cerebellar β-adrenergic receptors; the receptor effects were not due to increased systemic toxicity or cholinesterase inhibition, nor to altered chlorpyrifos pharmacokinetics. Further, the deficits were not secondary adaptations to presynaptic hyperinnervation/hyperactivity, as there were significant deficits in presynaptic norepinephrine levels that would serve to augment the functional consequence of receptor deficits. The pretreatments also altered development of cerebrocortical noradrenergic circuits, but with a different overall pattern, reflecting the dissimilar developmental stages of the regions at the time of exposure. However, in each case the net effects represented a change in the developmental trajectory of noradrenergic circuits, rather than simply a continuation of an initial injury. Our results point to the ability of prenatal drug exposure to create a subpopulation with heightened vulnerability to environmental neurotoxicants.
Collapse
Affiliation(s)
- Theodore A Slotkin
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Samantha Skavicus
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Frederic J Seidler
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
21
|
Feedforward and feedback motor control abnormalities implicate cerebellar dysfunctions in autism spectrum disorder. J Neurosci 2015; 35:2015-25. [PMID: 25653359 DOI: 10.1523/jneurosci.2731-14.2015] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Sensorimotor abnormalities are common in autism spectrum disorder (ASD) and among the earliest manifestations of the disorder. They have been studied far less than the social-communication and cognitive deficits that define ASD, but a mechanistic understanding of sensorimotor abnormalities in ASD may provide key insights into the neural underpinnings of the disorder. In this human study, we examined rapid, precision grip force contractions to determine whether feedforward mechanisms supporting initial motor output before sensory feedback can be processed are disrupted in ASD. Sustained force contractions also were examined to determine whether reactive adjustments to ongoing motor behavior based on visual feedback are altered. Sustained force was studied across multiple force levels and visual gains to assess motor and visuomotor mechanisms, respectively. Primary force contractions of individuals with ASD showed greater peak rate of force increases and large transient overshoots. Individuals with ASD also showed increased sustained force variability that scaled with force level and was more severe when visual gain was highly amplified or highly degraded. When sustaining a constant force level, their reactive adjustments were more periodic than controls, and they showed increased reliance on slower feedback mechanisms. Feedforward and feedback mechanism alterations each were associated with more severe social-communication impairments in ASD. These findings implicate anterior cerebellar circuits involved in feedforward motor control and posterior cerebellar circuits involved in transforming visual feedback into precise motor adjustments in ASD.
Collapse
|
22
|
Marzban H, Del Bigio MR, Alizadeh J, Ghavami S, Zachariah RM, Rastegar M. Cellular commitment in the developing cerebellum. Front Cell Neurosci 2015; 8:450. [PMID: 25628535 PMCID: PMC4290586 DOI: 10.3389/fncel.2014.00450] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 12/12/2014] [Indexed: 12/11/2022] Open
Abstract
The mammalian cerebellum is located in the posterior cranial fossa and is critical for motor coordination and non-motor functions including cognitive and emotional processes. The anatomical structure of cerebellum is distinct with a three-layered cortex. During development, neurogenesis and fate decisions of cerebellar primordium cells are orchestrated through tightly controlled molecular events involving multiple genetic pathways. In this review, we will highlight the anatomical structure of human and mouse cerebellum, the cellular composition of developing cerebellum, and the underlying gene expression programs involved in cell fate commitments in the cerebellum. A critical evaluation of the cell death literature suggests that apoptosis occurs in ~5% of cerebellar cells, most shortly after mitosis. Apoptosis and cellular autophagy likely play significant roles in cerebellar development, we provide a comprehensive discussion of their role in cerebellar development and organization. We also address the possible function of unfolded protein response in regulation of cerebellar neurogenesis. We discuss recent advancements in understanding the epigenetic signature of cerebellar compartments and possible connections between DNA methylation, microRNAs and cerebellar neurodegeneration. Finally, we discuss genetic diseases associated with cerebellar dysfunction and their role in the aging cerebellum.
Collapse
Affiliation(s)
- Hassan Marzban
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada
| | - Marc R Del Bigio
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada ; Department of Pathology, University of Manitoba Winnipeg, MB, Canada
| | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada
| | - Robby M Zachariah
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg, MB, Canada ; Regenerative Medicine Program, University of Manitoba Winnipeg, MB, Canada
| | - Mojgan Rastegar
- Department of Biochemistry and Medical Genetics, University of Manitoba Winnipeg, MB, Canada ; Regenerative Medicine Program, University of Manitoba Winnipeg, MB, Canada
| |
Collapse
|
23
|
Parker KL, Narayanan NS, Andreasen NC. The therapeutic potential of the cerebellum in schizophrenia. Front Syst Neurosci 2014; 8:163. [PMID: 25309350 PMCID: PMC4163988 DOI: 10.3389/fnsys.2014.00163] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 08/22/2014] [Indexed: 12/19/2022] Open
Abstract
The cognitive role of the cerebellum is critically tied to its distributed connections throughout the brain. Accumulating evidence from anatomical, structural and functional imaging, and lesion studies advocate a cognitive network involving indirect connections between the cerebellum and non-motor areas in the prefrontal cortex. Cerebellar stimulation dynamically influences activity in several regions of the frontal cortex and effectively improves cognition in schizophrenia. In this manuscript, we summarize current literature on the cingulocerebellar circuit and we introduce a method to interrogate this circuit combining opotogenetics, neuropharmacology, and electrophysiology in awake-behaving animals while minimizing incidental stimulation of neighboring cerebellar nuclei. We propose the novel hypothesis that optogenetic cerebellar stimulation can restore aberrant frontal activity and rescue impaired cognition in schizophrenia. We focus on how a known cognitive region in the frontal cortex, the anterior cingulate, is influenced by the cerebellum. This circuit is of particular interest because it has been confirmed using tracing studies, neuroimaging reveals its role in cognitive tasks, it is conserved from rodents to humans, and diseases such as schizophrenia and autism appear in its aberrancy. Novel tract tracing results presented here provide support for how these two areas communicate. The primary pathway involves a disynaptic connection between the cerebellar dentate nuclei (DN) and the anterior cingulate cortex. Secondarily, the pathway from cerebellar fastigial nuclei (FN) to the ventral tegmental area, which supplies dopamine to the prefrontal cortex, may play a role as schizophrenia characteristically involves dopamine deficiencies. We hope that the hypothesis described here will inspire new therapeutic strategies targeting currently untreatable cognitive impairments in schizophrenia.
Collapse
|