1
|
Selten M, Bernard C, Mukherjee D, Hamid F, Hanusz-Godoy A, Oozeer F, Zimmer C, Marín O. Regulation of PV interneuron plasticity by neuropeptide-encoding genes. Nature 2025:10.1038/s41586-025-08933-z. [PMID: 40307547 DOI: 10.1038/s41586-025-08933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
Neuronal activity must be regulated in a narrow permissive band for the proper operation of neural networks. Changes in synaptic connectivity and network activity-for example, during learning-might disturb this balance, eliciting compensatory mechanisms to maintain network function1-3. In the neocortex, excitatory pyramidal cells and inhibitory interneurons exhibit robust forms of stabilizing plasticity. However, although neuronal plasticity has been thoroughly studied in pyramidal cells4-8, little is known about how interneurons adapt to persistent changes in their activity. Here we describe a critical cellular process through which cortical parvalbumin-expressing (PV+) interneurons adapt to changes in their activity levels. We found that changes in the activity of individual PV+ interneurons drive bidirectional compensatory adjustments of the number and strength of inhibitory synapses received by these cells, specifically from other PV+ interneurons. High-throughput profiling of ribosome-associated mRNA revealed that increasing the activity of a PV+ interneuron leads to upregulation of two genes encoding multiple secreted neuropeptides: Vgf and Scg2. Functional experiments demonstrated that VGF is critically required for the activity-dependent scaling of inhibitory PV+ synapses onto PV+ interneurons. Our findings reveal an instructive role for neuropeptide-encoding genes in regulating synaptic connections among PV+ interneurons in the adult mouse neocortex.
Collapse
Affiliation(s)
- Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Clémence Bernard
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Diptendu Mukherjee
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Alicia Hanusz-Godoy
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Christoph Zimmer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
2
|
Bonnycastle K, Nawaz MS, Kind PC, Cousin MA. Convergent depression of activity-dependent bulk endocytosis in rodent models of autism spectrum disorder. Mol Autism 2025; 16:26. [PMID: 40241211 PMCID: PMC12004638 DOI: 10.1186/s13229-025-00660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND The key pathological mechanisms underlying autism spectrum disorder (ASD) remain relatively undetermined, potentially due to the heterogenous nature of the condition. Targeted studies of a series of monogenic ASDs have revealed postsynaptic dysfunction as a central conserved mechanism. Presynaptic dysfunction is emerging as an additional disease locus in neurodevelopmental disorders; however, it is unclear whether this dysfunction drives ASD or is an adaptation to the altered brain microenvironment. METHODS To differentiate between these two competing scenarios, we performed a high content analysis of key stages of the synaptic vesicle lifecycle in primary neuronal cultures derived from a series of preclinical rat models of monogenic ASD. These five independent models (Nrxn1+/-, Nlgn3-/y, Syngap+/-, Syngap+/Δ-GAP, Pten+/-) were specifically selected to have perturbations in a diverse palette of genes that were expressed either at the pre- or post-synapse. Synaptic vesicle exocytosis and cargo trafficking were triggered via two discrete trains of activity and monitored using the genetically-encoded reporter synaptophysin-pHluorin. Activity-dependent bulk endocytosis was assessed during intense neuronal activity using the fluid phase marker tetramethylrhodamine-dextran. RESULTS Both synaptic vesicle fusion events and cargo trafficking were unaffected in all models investigated under all stimulation protocols. However, a key convergent phenotype across neurons derived from all five models was revealed, a depression in activity-dependent bulk endocytosis. LIMITATIONS The study is exclusively conducted in primary cultures of hippocampal neurons; therefore, the impact on neurons from other brain regions or altered brain microcircuitry was not assessed. No molecular mechanism has been identified for this depression. CONCLUSION This suggests that depression of activity-dependent bulk endocytosis is a presynaptic homeostatic mechanism to correct for intrinsic dysfunction in ASD neurons.
Collapse
Affiliation(s)
- Katherine Bonnycastle
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Service de Génétique Médicale, Centre Hospitalier Universitaire (CHU) Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Mohammed Sarfaraz Nawaz
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK.
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK.
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, Scotland, EH8 9XD, UK.
| |
Collapse
|
3
|
Zhang X, Mukherjee A, Halassa MM, Chen ZS. Mediodorsal thalamus regulates task uncertainty to enable cognitive flexibility. Nat Commun 2025; 16:2640. [PMID: 40097445 PMCID: PMC11914509 DOI: 10.1038/s41467-025-58011-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/07/2025] [Indexed: 03/19/2025] Open
Abstract
The mediodorsal (MD) thalamus is a critical partner for the prefrontal cortex (PFC) in cognitive control. Accumulating evidence has shown that the MD regulates task uncertainty in decision making and enhance cognitive flexibility. However, the computational mechanism of this cognitive process remains unclear. Here we trained biologically-constrained computational models to delineate the mechanistic role of MD in context-dependent decision making. We show that the addition of a feedforward MD structure to the recurrent PFC increases robustness to low cueing signal-to-noise ratio, enhances working memory, and enables rapid context switching. Incorporating genetically identified thalamocortical connectivity and interneuron cell types into the model replicates key neurophysiological findings in task-performing animals. Our model reveals computational mechanisms and geometric interpretations of MD in regulating cue uncertainty and context switching to enable cognitive flexibility. Our model makes experimentally testable predictions linking cognitive deficits with disrupted thalamocortical connectivity, prefrontal excitation-inhibition imbalance and dysfunctional inhibitory cell types.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Arghya Mukherjee
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Michael M Halassa
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Zhe Sage Chen
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY, USA.
| |
Collapse
|
4
|
Wang X, Chen M, Mei D, Shi S, Guo J, Gao C, Wang Q, Zhao S, Yan X, Zhang H, Wang Y, Guo B, Zhang Y. Somatostatin-expressing interneurons of prefrontal cortex modulate social deficits in the Magel2 mouse model of autism. Mol Autism 2025; 16:18. [PMID: 40069835 PMCID: PMC11895276 DOI: 10.1186/s13229-025-00653-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/19/2025] [Indexed: 03/15/2025] Open
Abstract
Dysfunction in social interactions is a core symptom of autism spectrum disorder (ASD). Nevertheless, the neural mechanisms underlying social deficits in ASD are poorly understood. By integrating electrophysiological, in vivo fiber photometry, viral-mediated tracing, optogenetic and pharmacological stimulation, we show reduced intrinsic excitability and hypoactivity of SOM interneurons in medial prefrontal cortex (mPFC) in Magel2-deficient mice, an established ASD model, were required to social defects. Chemogenetic inhibition of mPFC SOM-containing interneurons resulted in reduced social interaction in wild-type Magel2 mice. These sociability deficits can be rescued by optogenetic activation by excitability of SOM in the mPFC and mPFCSOM-LS inhibitory pathway in Magel 2 knockout mice. These results demonstrate the hypoactivity for SOM action in the mPFC in social impairments, and suggest targeting this mechanism that may prove therapeutically beneficial for mitigating social behavioral disturbances observed in ASD.
Collapse
Affiliation(s)
- Xiaona Wang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China.
| | - Mengyuan Chen
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Daoqi Mei
- Department of Neurology, Children's Hospital of Suzhou University, Suzhou, China
| | - Shengli Shi
- Department of Image, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jisheng Guo
- Department of Pathology, School of Basic Medical Sciences, Yantai Campus of Binzhou Medical University, Yantai City, Shandong, China
| | - Chao Gao
- Department of Rehabilitation, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Qi Wang
- Department of Histology and Embryology, Guizhou Medical University, Guizhou, China
| | - Shuai Zhao
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Xingxue Yan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China
| | - Huichun Zhang
- Department of Rehabilitation, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yanli Wang
- Department of Rehabilitation, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Bin Guo
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Yaodong Zhang
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Henan Children's Neurodevelopment Engineering Research Center, Zhengzhou, China.
| |
Collapse
|
5
|
Kourdougli N, Nomura T, Wu MW, Heuvelmans A, Dobler Z, Contractor A, Portera-Cailliau C. The NKCC1 Inhibitor Bumetanide Restores Cortical Feedforward Inhibition and Lessens Sensory Hypersensitivity in Early Postnatal Fragile X Mice. Biol Psychiatry 2025; 97:507-516. [PMID: 38950809 PMCID: PMC11825064 DOI: 10.1016/j.biopsych.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Exaggerated responses to sensory stimuli, a hallmark of fragile X syndrome, contribute to anxiety and learning challenges. Sensory hypersensitivity is recapitulated in the Fmr1 knockout (KO) mouse model of fragile X syndrome. Recent studies in Fmr1 KO mice have demonstrated differences in the activity of cortical interneurons and a delayed switch in the polarity of GABA (gamma-aminobutyric acid) signaling during development. Previously, we reported that blocking the chloride transporter NKCC1 with the diuretic bumetanide could rescue synaptic circuit phenotypes in the primary somatosensory cortex (S1) of Fmr1 KO mice. However, it remains unknown whether bumetanide can rescue earlier circuit phenotypes or sensory hypersensitivity in Fmr1 KO mice. METHODS We used acute and chronic systemic administration of bumetanide in Fmr1 KO mice and performed in vivo 2-photon calcium imaging to record neuronal activity, while tracking mouse behavior with high-resolution videos. RESULTS We demonstrated that layer 2/3 pyramidal neurons in the S1 of Fmr1 KO mice showed a higher frequency of synchronous events on postnatal day 6 than wild-type controls. This was reversed by acute administration of bumetanide. Furthermore, chronic bumetanide treatment (postnatal days 5-14) restored S1 circuit differences in Fmr1 KO mice, including reduced neuronal adaptation to repetitive whisker stimulation, and ameliorated tactile defensiveness. Bumetanide treatment also rectified the reduced feedforward inhibition of layer 2/3 neurons in the S1 and boosted the circuit participation of parvalbumin interneurons. CONCLUSIONS This further supports the notion that synaptic, circuit, and sensory behavioral phenotypes in Fmr1 KO can be mitigated by inhibitors of NKCC1, such as the Food and Drug Administration-approved diuretic bumetanide.
Collapse
Affiliation(s)
- Nazim Kourdougli
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | - Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Michelle W Wu
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, California; UCLA-Caltech Medical Scientist Training Program, University of California, Los Angeles, Los Angeles, California
| | - Anouk Heuvelmans
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | - Zoë Dobler
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, California
| | - Anis Contractor
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Carlos Portera-Cailliau
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Department of Neurobiology, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
6
|
Padmashri R, Dunaevsky A. Targeting GABA Polarity During Cortical Development Improves Circuit and Sensory Deficits in Fragile X Mice. Biol Psychiatry 2025; 97:420-421. [PMID: 39919885 PMCID: PMC12004231 DOI: 10.1016/j.biopsych.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025]
Affiliation(s)
- Ragunathan Padmashri
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Anna Dunaevsky
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
7
|
DeSpenza T, Kiziltug E, Allington G, Barson DG, McGee S, O'Connor D, Robert SM, Mekbib KY, Nanda P, Greenberg ABW, Singh A, Duy PQ, Mandino F, Zhao S, Lynn A, Reeves BC, Marlier A, Getz SA, Nelson-Williams C, Shimelis H, Walsh LK, Zhang J, Wang W, Prina ML, OuYang A, Abdulkareem AF, Smith H, Shohfi J, Mehta NH, Dennis E, Reduron LR, Hong J, Butler W, Carter BS, Deniz E, Lake EMR, Constable RT, Sahin M, Srivastava S, Winden K, Hoffman EJ, Carlson M, Gunel M, Lifton RP, Alper SL, Jin SC, Crair MC, Moreno-De-Luca A, Luikart BW, Kahle KT. PTEN mutations impair CSF dynamics and cortical networks by dysregulating periventricular neural progenitors. Nat Neurosci 2025; 28:536-557. [PMID: 39994410 PMCID: PMC12038823 DOI: 10.1038/s41593-024-01865-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/05/2024] [Indexed: 02/26/2025]
Abstract
Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (ventriculomegaly) is a defining feature of congenital hydrocephalus (CH) and an under-recognized concomitant of autism. Here, we show that de novo mutations in the autism risk gene PTEN are among the most frequent monogenic causes of CH and primary ventriculomegaly. Mouse Pten-mutant ventriculomegaly results from aqueductal stenosis due to hyperproliferation of periventricular Nkx2.1+ neural progenitor cells (NPCs) and increased CSF production from hyperplastic choroid plexus. Pten-mutant ventriculomegalic cortices exhibit network dysfunction from increased activity of Nkx2.1+ NPC-derived inhibitory interneurons. Raptor deletion or postnatal everolimus treatment corrects ventriculomegaly, rescues cortical deficits and increases survival by antagonizing mTORC1-dependent Nkx2.1+ NPC pathology. Thus, PTEN mutations concurrently alter CSF dynamics and cortical networks by dysregulating Nkx2.1+ NPCs. These results implicate a nonsurgical treatment for CH, demonstrate a genetic association of ventriculomegaly and ASD, and help explain neurodevelopmental phenotypes refractory to CSF shunting in select individuals with CH.
Collapse
Affiliation(s)
- Tyrone DeSpenza
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Emre Kiziltug
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Garrett Allington
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons and New York Presbyterian Hospital, New York, NY, USA
| | - Daniel G Barson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | | | - David O'Connor
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie M Robert
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Kedous Y Mekbib
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pranav Nanda
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana B W Greenberg
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Amrita Singh
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Phan Q Duy
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Francesca Mandino
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Shujuan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Anna Lynn
- Medical Scientist Training Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Stephanie A Getz
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Carol Nelson-Williams
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Hermela Shimelis
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Lauren K Walsh
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
| | - Junhui Zhang
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Wei Wang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Mackenzi L Prina
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Annaliese OuYang
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Asan F Abdulkareem
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA
| | - Hannah Smith
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - John Shohfi
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Neel H Mehta
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Evan Dennis
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laetitia R Reduron
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Jennifer Hong
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - William Butler
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Engin Deniz
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Evelyn M R Lake
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - R Todd Constable
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kellen Winden
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ellen J Hoffman
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Marina Carlson
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Seth L Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael C Crair
- Interdepartmental Neuroscience Program, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Andres Moreno-De-Luca
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA, USA
- Department of Radiology, Diagnostic Medicine Institute, Geisinger, Danville, PA, USA
| | - Bryan W Luikart
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Neurobiology, UAB Heersink School of Medicine, Birmingham, AL, USA.
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
8
|
Rosés-Novella C, Bernard C. Dynamic regulation of cortical interneuron wiring. Curr Opin Neurobiol 2025; 92:102980. [PMID: 40015134 DOI: 10.1016/j.conb.2025.102980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 03/01/2025]
Abstract
Inhibitory interneurons play crucial roles in modulating the circuits and activity patterns of the cerebral cortex. In particular, interneurons must adapt to changes in cortical activity and environmental information to drive appropriate responses. In this review, we focus on the latest progress in our understanding of the processes that regulate interneuron wiring adaptability. We discuss newly identified types of regulatory processes, from structural synaptic changes to long-range neuromodulation, and provide an update on the activity-dependent molecular underpinnings at the transcriptional and post-transcriptional levels.
Collapse
Affiliation(s)
- Claudia Rosés-Novella
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom. https://twitter.com/@claudiarsnv
| | - Clémence Bernard
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 4PS, United Kingdom.
| |
Collapse
|
9
|
Kumaraguru S, Morgan J, Wong FK. Activity-dependent regulation of microglia numbers by pyramidal cells during development shape cortical functions. SCIENCE ADVANCES 2025; 11:eadq5842. [PMID: 39970202 PMCID: PMC11838000 DOI: 10.1126/sciadv.adq5842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025]
Abstract
Beyond their role as immune sentinels, microglia are actively involved in establishing and maintaining cortical circuits. Alteration in microglial numbers has been associated with abnormal behaviors akin to those observed in neurodevelopmental disorders. Consequently, establishing the appropriate microglial numbers during development is crucial for ensuring normal cortical function. Here, we uncovered a dynamic relationship between pyramidal cells and microglia that tunes microglial numbers and development through distinct phases of mouse postnatal development. Changes in pyramidal cell activity during development induce differential release of activity-dependent proteins such as Activin A, which, in turn, adjusts microglial numbers accordingly. Decoupling of this relationship not only changes microglial numbers but has a long-term consequence on their role as synaptic organizers, which ultimately affects cortical function. Our findings reveal that microglia adapt their numbers to changes in pyramidal cell activity during a critical time window in development, consequently adjusting their numbers and function to the demands of the developing local circuits.
Collapse
Affiliation(s)
- Sanjana Kumaraguru
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - James Morgan
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Fong Kuan Wong
- Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, UK
| |
Collapse
|
10
|
Zhang X, Wu M, Cheng L, Cao W, Liu Z, Yang SB, Kim MS. Fast-spiking parvalbumin-positive interneurons: new perspectives of treatment and future challenges in dementia. Mol Psychiatry 2025; 30:693-704. [PMID: 39695324 DOI: 10.1038/s41380-024-02756-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/26/2024] [Accepted: 09/12/2024] [Indexed: 12/20/2024]
Abstract
Central nervous system parvalbumin-positive interneurons (PV-INs) are crucial and highly vulnerable to various stressors. They also play a significant role in the pathological processes of many neuropsychiatric diseases, especially those associated with cognitive impairment, such as Alzheimer's disease (AD), vascular dementia (VD), Lewy body dementia, and schizophrenia. Although accumulating evidence suggests that the loss of PV-INs is associated with memory impairment in dementia, the precise molecular mechanisms remain elusive. In this review, we delve into the current evidence regarding the physiological properties of PV-INs and summarize the latest insights into how their loss contributes to cognitive decline in dementia, particularly focusing on AD and VD. Additionally, we discuss the influence of PV-INs on brain development, the variations in their characteristics across different types of dementia, and how their loss affects the etiology and progression of cognitive impairments. Ultimately, our goal is to provide a comprehensive overview of PV-INs and to consider their potential as novel therapeutic targets in dementia treatment.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Department of Pathology, Affiliated Hospital of Jiujiang University, Jiujiang, China
- Jiujiang Clinical Precision Clinical Medicine Research Center, Jiujiang, Jiangxi, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Moxin Wu
- Jiujiang Clinical Precision Clinical Medicine Research Center, Jiujiang, Jiangxi, China
| | - Lin Cheng
- Jiujiang Clinical Precision Clinical Medicine Research Center, Jiujiang, Jiangxi, China
| | - Wa Cao
- Jiujiang Clinical Precision Clinical Medicine Research Center, Jiujiang, Jiangxi, China
| | - Ziying Liu
- Jiujiang Clinical Precision Clinical Medicine Research Center, Jiujiang, Jiangxi, China
| | - Seung-Bum Yang
- Department of Paramedicine, Wonkwang Health Science University, Iksan, Republic of Korea
| | - Min-Sun Kim
- Center for Nitric Oxide Metabolite, Wonkwang University, Iksan, Republic of Korea.
| |
Collapse
|
11
|
Hu Y, Feng Y, Luo H, Zhu XN, Chen S, Yang K, Deng Z, Luo M, Du W, Wang Q, Wang S, Wei K, Hu J, Wang Y. Dissociation-related behaviors in mice emerge from the inhibition of retrosplenial cortex parvalbumin interneurons. Cell Rep 2025; 44:115086. [PMID: 39708317 DOI: 10.1016/j.celrep.2024.115086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/11/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
Dissociation, characterized by altered consciousness and perception, underlies multiple mental disorders, but the specific neuronal subtypes involved remain elusive. In mice, we find that dissociation-inducing doses of ketamine significantly inhibit retrosplenial cortex (RSC) parvalbumin interneurons (PV-INs), enhancing delta oscillations (1-3 Hz) and delta-gamma phase-amplitude coupling (δ-γ PAC) and inducing dissociation-like behaviors. Optogenetic inhibition of RSC PV-INs triggers delta oscillations, δ-γ PAC, and some dissociation-like behaviors without ketamine. Furthermore, activation of RSC PV-INs or knockdown of the N-methyl-D-aspartate receptor subunit NR1 and the hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1) in RSC PV-INs attenuates ketamine-induced delta oscillations, δ-γ PAC, and certain dissociation-like behaviors. These findings reveal that PV-INs regulate delta oscillations and δ-γ PAC and identify NR1 and HCN1 as ketamine targets in PV-INs that may cooperatively affect dissociation, possibly providing potential therapeutic targets for dissociative symptoms.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yifan Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Siyu Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziqing Deng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qi Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shubai Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kai Wei
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
12
|
Shih YT, Alipio JB, Klaft ZJ, Green N, Wong LP, Sadreyev R, Hyun JH, Dulla C, Sahay A. Pro-cognitive restoration of experience-dependent parvalbumin inhibitory neuron plasticity in neurodevelopmental disorders. RESEARCH SQUARE 2025:rs.3.rs-5624085. [PMID: 39877097 PMCID: PMC11774442 DOI: 10.21203/rs.3.rs-5624085/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
The hippocampus forms memories of our experiences by registering processed sensory information in coactive populations of excitatory principal cells or ensembles1-3. Fast-spiking parvalbumin-expressing inhibitory neurons (PV INs) in the dentate gyrus (DG)-CA3/CA2 circuit contribute to memory encoding by exerting precise temporal control of excitatory principal cell activity through mossy fiber-dependent feed-forward inhibition4-13. PV INs respond to input-specific information by coordinating changes in their intrinsic excitability, input-output synaptic-connectivity, synaptic-physiology and synaptic-plasticity9,13-17, referred to here as experience-dependent PV IN plasticity, to influence hippocampal functions. PV IN impairments in early life, when neural circuitry is highly sensitive to experience-dependent refinement, are thought to result in imbalanced excitation and inhibition, impaired cognition, network hyperexcitability and seizures: hallmarks of neurodevelopmental disorders (NDDs) such as Autism Spectrum Disorder and epilepsy18-20. Discovery of transcriptional regulators of experience-dependent PV IN plasticity in the adult hippocampus may permit reversal of these developmental impairments. Here, in a screen designed to capture the PV IN intrinsic program induced by increased mossy fiber inputs, a trigger for experience-dependent PV IN plasticity, we identify the homeobox gene Meis2 21 as a regulator of experience-dependent PV IN plasticity gene (XPG) in the adult DG-CA3/CA2 circuit. We found that a significant number of upregulated XPGs also exhibit haploinsufficiency in ASDs, epilepsies, and schizophrenia. We demonstrate that virally-mediated rescue of experience-dependent Meis2 upregulation in CA3/CA2 PV INs in a NDD risk mouse model in adulthood is sufficient to restore experience-dependent PV IN plasticity, spatial and social memory, ensemble specificity, suppression of network hyperexcitability and seizures. Together, these findings suggest that experience-dependent PV IN plasticity is a convergent mechanism for NDD risk genes that can be re-instated in adulthood to reverse developmental deficits in circuitry, network excitability and cognition.
Collapse
Affiliation(s)
- Yu-Tzu Shih
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jason Bondoc Alipio
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Zin-Juan Klaft
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Nathaniel Green
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Lai Ping Wong
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology. Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Chris Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- BROAD Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
Monday HR, Nieto AM, Yohannes SA, Luxu S, Wong KW, Bolio FE, Feldman DE. Physiological and molecular impairment of PV circuit homeostasis in mouse models of autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.632056. [PMID: 39829892 PMCID: PMC11741295 DOI: 10.1101/2025.01.08.632056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Circuit dysfunction in autism may involve a failure of homeostatic plasticity. To test this, we studied parvalbumin (PV) interneurons which exhibit rapid homeostatic plasticity of intrinsic excitability following whisker deprivation in mouse somatosensory cortex. Brief deprivation reduces PV excitability by increasing Kv1 current to increase PV spike threshold. We found that PV homeostatic plasticity is disrupted in Tsc2 +/- and Fmr1 -/- models of autism. In wildtype mice, deprivation elevates the transcription factor ER81 which drives Kcna1 transcription, increasing Kv1.1 protein in the axon initial segment and soma. These molecular signatures of homeostasis were absent in Tsc2 +/- and Fmr1 -/- . Whisker enrichment increased PV excitability, but not in Tsc2 +/- , indicating that homeostasis is lost bidirectionally. Deprivation reduced feedforward L4-L2/3 inhibition in wildtype but not Tsc2 +/- mice. Thus, two autism models show a convergent loss of PV circuit homeostasis at physiological and molecular levels, potentially contributing to sensory processing impairments.
Collapse
|
14
|
Chalkiadaki K, Statoulla E, Zafeiri M, Voudouri G, Amvrosiadis T, Typou A, Theodoridou N, Moschovas D, Avgeropoulos A, Samiotaki M, Mason JO, Gkogkas CG. GABA/Glutamate Neuron Differentiation Imbalance and Increased AKT/mTOR Signaling in CNTNAP2 -/- Cerebral Organoids. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100413. [PMID: 39758604 PMCID: PMC11699409 DOI: 10.1016/j.bpsgos.2024.100413] [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: 07/30/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 01/07/2025] Open
Abstract
Background The polygenic nature of autism spectrum disorder (ASD) requires the identification of converging genetic pathways during early development to elucidate its complexity and varied manifestations. Methods We developed a human cerebral organoid model from induced pluripotent stem cells with targeted genome editing to abolish protein expression of the CNTNAP2 ASD risk gene. Results CNTNAP2-/- cerebral organoids displayed accelerated cell cycle, ventricular zone disorganization, and increased cortical folding. Proteomic analysis revealed disruptions in glutamatergic/GABAergic (gamma-aminobutyric acidergic) synaptic pathways and neurodevelopment, and transcriptomic analysis revealed differentially expressed genes belonging to inhibitory neuron-related gene networks. Interestingly, there was a weak correlation between the 2 datasets, suggesting nuanced translational control mechanisms. Along these lines, we found upregulated AKT/mTOR (mechanistic target of rapamycin) signaling in CNTNAP2-/- organoids. Spatial transcriptomic analysis of CNTNAP2-/- ventricular-like zones demonstrated pervasive changes in gene expression, implicating upregulation of cell cycle regulation, synaptic, and glutamatergic/GABAergic pathways. We noted significant overlap of all day-30 organoid omics datasets differentially expressed genes from idiopathic ASD (macrocephaly) induced pluripotent stem cell-derived telencephalic organoids, where FOXG1 was upregulated. Moreover, we detected increased GAD1-expressing and decreased TBR1-expressing cells, suggesting altered GABAergic/glutamatergic neuron development. Conclusions These findings potentially highlight a shared mechanism in the early cortical development of various forms of ASD, further elucidate the role of CNTNAP2 in ASD pathophysiology and cortical development, and pave the way for targeted therapies that use cerebral organoids as preclinical models.
Collapse
Affiliation(s)
- Kleanthi Chalkiadaki
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Elpida Statoulla
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Maria Zafeiri
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Georgia Voudouri
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | | | - Alexandra Typou
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Niki Theodoridou
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| | - Dimitrios Moschovas
- Department of Materials Science Engineering, University of Ioannina, Ioannina, Greece
| | | | - Martina Samiotaki
- Biomedical Sciences Research Center “Alexander Fleming”, Vari, Greece
| | - John O. Mason
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
| | - Christos G. Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, Ioannina, Greece
| |
Collapse
|
15
|
Ibáñez-Sandoval DN, Hidalgo-Balbuena AE, Velázquez Contreras R, Saderi N, Flores G, Rueda-Orozco PE, Ibáñez-Sandoval O. Striatal Interneuron Imbalance in a Valproic Acid-Induced Model of Autism in Rodents Is Accompanied by Atypical Somatosensory Processing. eNeuro 2024; 11:ENEURO.0326-24.2024. [PMID: 39572246 PMCID: PMC11653103 DOI: 10.1523/eneuro.0326-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 12/16/2024] Open
Abstract
Autism spectrum disorder (ASD) is characterized by deficits in social interaction and communication, cognitive rigidity, and atypical sensory processing. Recent studies suggest that the basal ganglia, specifically the striatum (NSt), plays an important role in ASD. While striatal interneurons, including cholinergic (ChAT+) and parvalbumin-positive (PV+) GABAergic neurons, have been described to be altered in animal models of ASD, their specific contribution remains elusive. Here, we combined behavioral, anatomical, and electrophysiological quantifications to explore if interneuron balance could be implicated in atypical sensory processing in cortical and striatal somatosensory regions of rats subjected to a valproic acid (VPA) model of ASD. We found that VPA animals showed a significant decrease in the number of ChAT+ and PV+ cells in multiple regions (including the sensorimotor region) of the NSt. We also observed significantly different sensory-evoked responses at the single-neuron and population levels in both striatal and cortical regions, as well as corticostriatal interactions. Therefore, selective elimination of striatal PV+ neurons only partially recapitulated the effects of VPA, indicating that the mechanisms behind the VPA phenotype are much more complex than the elimination of a particular neural subpopulation. Our results indicate that VPA exposure induced significant histological changes in ChAT+ and PV+ cells accompanied by atypical sensory-evoked corticostriatal population dynamics that could partially explain the sensory processing differences associated with ASD.
Collapse
Affiliation(s)
- Dayna N Ibáñez-Sandoval
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Ana E Hidalgo-Balbuena
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM, Querétaro 76230, México
| | | | - Nadia Saderi
- Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78295, México
| | - Gonzalo Flores
- Instituto de Fisiología, Universidad Autónoma de Puebla, Puebla CP 72570, México
| | - Pavel E Rueda-Orozco
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM, Querétaro 76230, México
| | - Osvaldo Ibáñez-Sandoval
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| |
Collapse
|
16
|
Subramanian D, Eisenberg C, Huang A, Baek J, Naveed H, Komatireddy S, Shiflett MW, Tran TS, Santhakumar V. Dysregulation of neuropilin-2 expression in inhibitory neurons impairs hippocampal circuit development and enhances risk for autism-related behaviors and seizures. Mol Psychiatry 2024:10.1038/s41380-024-02839-4. [PMID: 39578518 DOI: 10.1038/s41380-024-02839-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
Dysregulation of development, migration, and function of interneurons, collectively termed interneuronopathies, have been proposed as a shared mechanism for autism spectrum disorders (ASDs) and childhood epilepsy. Neuropilin-2 (Nrp2), a candidate ASD gene, is a critical regulator of interneuron migration from the median ganglionic eminence (MGE) to the pallium, including the hippocampus. While clinical studies have identified Nrp2 polymorphisms in patients with ASD, whether selective dysregulation of Nrp2-dependent interneuron migration contributes to pathogenesis of ASD and enhances the risk for seizures has not been evaluated. We tested the hypothesis that the lack of Nrp2 in MGE-derived interneuron precursors disrupts the excitation/inhibition balance in hippocampal circuits, thus predisposing the network to seizures and behavioral patterns associated with ASD. Embryonic deletion of Nrp2 during the developmental period for migration of MGE derived interneuron precursors (iCKO) significantly reduced parvalbumin, neuropeptide Y, and somatostatin positive neurons in the hippocampal CA1. Consequently, when compared to controls, the frequency of inhibitory synaptic currents in CA1 pyramidal cells was reduced while frequency of excitatory synaptic currents was increased in iCKO mice. Although passive and active membrane properties of CA1 pyramidal cells were unchanged, iCKO mice showed enhanced susceptibility to chemically evoked seizures. Moreover, iCKO mice exhibited selective behavioral deficits in both preference for social novelty and goal-directed learning, which are consistent with ASD-like phenotype. Together, our findings show that disruption of developmental Nrp2 regulation of interneuron circuit establishment, produces ASD-like behaviors and enhanced risk for epilepsy. These results support the developmental interneuronopathy hypothesis of ASD epilepsy comorbidity.
Collapse
Affiliation(s)
- Deepak Subramanian
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
| | - Carol Eisenberg
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Andrew Huang
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Jiyeon Baek
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Haniya Naveed
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Samiksha Komatireddy
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
| | | | - Tracy S Tran
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA.
| | - Vijayalakshmi Santhakumar
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA.
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA.
| |
Collapse
|
17
|
Sutley-Koury SN, Taitano-Johnson C, Kulinich AO, Farooq N, Wagner VA, Robles M, Hickmott PW, Santhakumar V, Mimche PN, Ethell IM. EphB2 Signaling Is Implicated in Astrocyte-Mediated Parvalbumin Inhibitory Synapse Development. J Neurosci 2024; 44:e0154242024. [PMID: 39327008 PMCID: PMC11551896 DOI: 10.1523/jneurosci.0154-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Impaired inhibitory synapse development is suggested to drive neuronal hyperactivity in autism spectrum disorders (ASD) and epilepsy. We propose a novel mechanism by which astrocytes control the development of parvalbumin (PV)-specific inhibitory synapses in the hippocampus, implicating ephrin-B/EphB signaling. Here, we utilize genetic approaches to assess functional and structural connectivity between PV and pyramidal cells (PCs) through whole-cell patch-clamp electrophysiology, optogenetics, immunohistochemical analysis, and behaviors in male and female mice. While inhibitory synapse development is adversely affected by PV-specific expression of EphB2, a strong candidate ASD risk gene, astrocytic ephrin-B1 facilitates PV→PC connectivity through a mechanism involving EphB signaling in PV boutons. In contrast, the loss of astrocytic ephrin-B1 reduces PV→PC connectivity and inhibition, resulting in increased seizure susceptibility and an ASD-like phenotype. Our findings underscore the crucial role of astrocytes in regulating inhibitory circuit development and discover a new role of EphB2 receptors in PV-specific inhibitory synapse development.
Collapse
Affiliation(s)
- Samantha N Sutley-Koury
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Christopher Taitano-Johnson
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| | - Anna O Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Nadia Farooq
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Victoria A Wagner
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| | - Marissa Robles
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
| | - Peter W Hickmott
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| | | | - Patrice N Mimche
- Department of Dermatology, and Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis Indiana 46202
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California 92521
- Neuroscience Graduate Program, University of California Riverside, Riverside, California 92521
| |
Collapse
|
18
|
Barron JJ, Mroz NM, Taloma SE, Dahlgren MW, Ortiz-Carpena JF, Keefe MG, Escoubas CC, Dorman LC, Vainchtein ID, Chiaranunt P, Kotas ME, Nowakowski TJ, Bender KJ, Molofsky AB, Molofsky AV. Group 2 innate lymphoid cells promote inhibitory synapse development and social behavior. Science 2024; 386:eadi1025. [PMID: 39480923 PMCID: PMC11995778 DOI: 10.1126/science.adi1025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 02/22/2024] [Accepted: 09/02/2024] [Indexed: 11/02/2024]
Abstract
The innate immune system shapes brain development and is implicated in neurodevelopmental diseases. It is critical to define the relevant immune cells and signals and their impact on brain circuits. In this work, we found that group 2 innate lymphoid cells (ILC2s) and their cytokine interleukin-13 (IL-13) signaled directly to inhibitory interneurons to increase inhibitory synapse density in the developing mouse brain. ILC2s expanded and produced IL-13 in the developing brain meninges. Loss of ILC2s or IL-13 signaling to interneurons decreased inhibitory, but not excitatory, cortical synapses. Conversely, ILC2s and IL-13 were sufficient to increase inhibitory synapses. Loss of this signaling pathway led to selective impairments in social interaction. These data define a type 2 neuroimmune circuit in early life that shapes inhibitory synapse development and behavior.
Collapse
Affiliation(s)
- Jerika J. Barron
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nicholas M. Mroz
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sunrae E. Taloma
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Madelene W. Dahlgren
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
- Lung biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jorge F. Ortiz-Carpena
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Matthew G. Keefe
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Caroline C. Escoubas
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Leah C. Dorman
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Ilia D. Vainchtein
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Pailin Chiaranunt
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Maya E. Kotas
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tomasz J. Nowakowski
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kevin J. Bender
- Kavli Institute for Fundamental Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ari B. Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Anna V. Molofsky
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Kavli Institute for Fundamental Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
19
|
Rocha Neto HG, Lessa JLM, Koiller LM, Pereira AM, de Souza Gomes BM, Veloso Filho CL, Telleria CHC, Cavalcanti MT, Telles-Correia D. Non-standard diagnostic assessment reliability in psychiatry: a study in a Brazilian outpatient setting using Kappa. Eur Arch Psychiatry Clin Neurosci 2024; 274:1759-1770. [PMID: 38085328 DOI: 10.1007/s00406-023-01730-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/21/2023] [Indexed: 09/25/2024]
Abstract
The use of Structured Diagnostic Assessments (SDAs) is a solution for unreliability in psychiatry and the gold standard for diagnosis. However, except for studies between the 50 s and 70 s, reliability without the use of Non-SDAs (NSDA) is seldom tested, especially in non-Western, Educated, Industrialized, Rich, and Democratic (WEIRD) countries. We aim to measure reliability between examiners with NSDAs for psychiatric disorders. We compared diagnostic agreement after clinician change, in an outpatient academic setting. We used inter-rater Kappa measuring 8 diagnostic groups: Depression (DD: F32, F33), Anxiety Related Disorders (ARD: F40-F49, F50-F59), Personality Disorders (PD: F60-F69), Bipolar Disorder (BD: F30, F31, F34.0, F38.1), Organic Mental Disorders (Org: F00-F09), Neurodevelopment Disorders (ND: F70-F99) and Schizophrenia Spectrum Disorders (SSD: F20-F29). Cohen's Kappa measured agreement between groups, and Baphkar's test assessed if any diagnostic group have a higher tendency to change after a new diagnostic assessment. We analyzed 739 reevaluation pairs, from 99 subjects who attended IPUB's outpatient clinic. Overall inter-rater Kappa was moderate, and none of the groups had a different tendency to change. NSDA evaluation was moderately reliable, but the lack of some prevalent hypothesis inside the pairs raised concerns about NSDA sensitivity to some diagnoses. Diagnostic momentum bias (that is, a tendency to keep the last diagnosis observed) may have inflated the observed agreement. This research was approved by IPUB's ethical committee, registered under the CAAE33603220.1.0000.5263, and the UTN-U1111-1260-1212.
Collapse
Affiliation(s)
- Helio G Rocha Neto
- Programa de Pós-Graduação em Psiquiatria e Saúde Mental - PROPSAM, Instituto de Psiquiatria da UFRJ, Av.Venceslau Brás, nº71 Fundos, Gabinete da Direção, Botafogo, Rio de Janeiro, RJ, Brazil.
- Programa de Doutoramento do Centro Acadêmico de Medicina da Universidade de Lisboa - PhD CAML, Lisbon, Portugal.
| | - José Luiz Martins Lessa
- Instituto de Psiquiatria, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Luisa Mendez Koiller
- Instituto de Psiquiatria, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Amanda Machado Pereira
- Instituto de Psiquiatria, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | | | - Carlos Linhares Veloso Filho
- Programa de Pós-Graduação em Psiquiatria e Saúde Mental - PROPSAM, Instituto de Psiquiatria da UFRJ, Av.Venceslau Brás, nº71 Fundos, Gabinete da Direção, Botafogo, Rio de Janeiro, RJ, Brazil
| | - Carlos Henrique Casado Telleria
- Medicine Faculty, Centro de Ciências da Saúde - CCS, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Maria T Cavalcanti
- Programa de Pós-Graduação em Psiquiatria e Saúde Mental - PROPSAM, Instituto de Psiquiatria da UFRJ, Av.Venceslau Brás, nº71 Fundos, Gabinete da Direção, Botafogo, Rio de Janeiro, RJ, Brazil
- Medicine Faculty, Centro de Ciências da Saúde - CCS, Universidade Federal do Rio de Janeiro - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Diogo Telles-Correia
- Clinica Universitária de Psiquiatria e Psicologia Médica, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Programa de Doutoramento do Centro Acadêmico de Medicina da Universidade de Lisboa - PhD CAML, Lisbon, Portugal
| |
Collapse
|
20
|
Zhang B, Chen K, Dai Y, Luo X, Xiong Z, Zhang W, Huang X, So KF, Zhang L. Human α-synuclein aggregation activates ferroptosis leading to parvalbumin interneuron degeneration and motor learning impairment. Commun Biol 2024; 7:1227. [PMID: 39349708 PMCID: PMC11443099 DOI: 10.1038/s42003-024-06896-x] [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: 07/05/2023] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
The accumulation of α-synuclein induces neuronal loss in midbrain nuclei and leads to the disruption of motor circuits, while the pathology of α-synuclein in cortical regions remains elusive. To better characterize cortical synucleinopathy, here we generate a mouse model with the overexpression of human α-synuclein in the primary motor cortex (M1) of mice. A combination of molecular, in vivo recording, and behavioral approaches reveal that cortical expression of human α-synuclein results in the overexcitation of cortical pyramidal neurons (PNs), which are regulated by the decreased inhibitory inputs from parvalbumin-interneurons (PV-INs) to impair complex motor skill learning. Further mechanistic dissections reveal that human α-synuclein aggregation activates ferroptosis, contributing to PV-IN degeneration and motor circuit dysfunction. Taken together, the current study adds more knowledge to the emerging role and pathogenic mechanism of ferroptosis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Borui Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
| | - Kai Chen
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
| | - Yelin Dai
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
| | - Xi Luo
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
| | - Ziwei Xiong
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
| | - Weijia Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
| | - Xiaodan Huang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
| | - Kwok-Fai So
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China
- State Key Laboratory of Brain and Cognitive Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P. R. China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, P. R. China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, China
| | - Li Zhang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, P. R. China.
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, P. R. China.
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China.
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
21
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. iScience 2024; 27:110800. [PMID: 39310747 PMCID: PMC11416532 DOI: 10.1016/j.isci.2024.110800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement pathway dysregulation in PV cells drives disease pathogenesis, we have developed a transgenic line that permits cell-type specific overexpression of the schizophrenia-associated C4 gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific alterations in anxiety-like behavior and deficits in synaptic connectivity and excitability of PFC PV cells. Using a computational model, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that perturbations of this neuroimmune gene in fast-spiking neurons are especially detrimental to circuits associated with anxiety-like behavior. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics MS Program, Boston University, Boston, MA, USA
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, USA
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- NeuroTechnology Center (NTC), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
22
|
Dumontier D, Liebman SA, Le VH, George S, Valdemar D, Van Aelst L, Pouchelon G. Restoring transient connectivity during development improves dysfunctions in fragile X mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.08.611918. [PMID: 39314327 PMCID: PMC11419037 DOI: 10.1101/2024.09.08.611918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Early-generated circuits are critical for the maturation of cortical network activity and the formation of excitation/inhibition (E/I) balance. This process involves the maturation of specific populations of inhibitory neurons. While parvalbumin (PV)-expressing neurons have been associated with E/I impairments observed in neurodevelopmental disorders, somatostatin-expressing (SST) neurons have recently been shown to regulate PV neuron maturation by controlling neural dynamics in the developing cortex. SST neurons receive transient connections from the sensory thalamus, yet the implications of transient connectivity in neurodevelopmental disorders remain unknown. Here, we show that thalamocortical connectivity to SST neurons is persistent rather than transient in a mouse model of Fragile X syndrome. We were able to restore the transient dynamics using chemogenetics, which led to the recovery of fragile X-associated dysfunctions in circuit maturation and sensory-dependent behavior. Overall, our findings unveil the role of early transient dynamics in controlling downstream maturation of sensory functions.
Collapse
Affiliation(s)
| | | | - Viet-Hang Le
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Shanu George
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | | | | |
Collapse
|
23
|
Subramanian D, Eisenberg C, Huang A, Baek J, Naveed H, Komatireddy S, Shiflett MW, Tran TS, Santhakumar V. Dysregulation of Neuropilin-2 Expression in Inhibitory Neurons Impairs Hippocampal Circuit Development and Enhances Risk for Autism-Related Behaviors and Seizures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578976. [PMID: 38370800 PMCID: PMC10871171 DOI: 10.1101/2024.02.05.578976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Dysregulation of development, migration, and function of interneurons, collectively termed interneuronopathies, have been proposed as a shared mechanism for autism spectrum disorders (ASDs) and childhood epilepsy. Neuropilin-2 (Nrp2), a candidate ASD gene, is a critical regulator of interneuron migration from the median ganglionic eminence (MGE) to the pallium, including the hippocampus. While clinical studies have identified Nrp2 polymorphisms in patients with ASD, whether selective dysregulation of Nrp2-dependent interneuron migration contributes to pathogenesis of ASD and enhances the risk for seizures has not been evaluated. We tested the hypothesis that the lack of Nrp2 in MGE-derived interneuron precursors disrupts the excitation/inhibition balance in hippocampal circuits, thus predisposing the network to seizures and behavioral patterns associated with ASD. Embryonic deletion of Nrp2 during the developmental period for migration of MGE derived interneuron precursors (iCKO) significantly reduced parvalbumin, neuropeptide Y, and somatostatin positive neurons in the hippocampal CA1. Consequently, when compared to controls, the frequency of inhibitory synaptic currents in CA1 pyramidal cells was reduced while frequency of excitatory synaptic currents was increased in iCKO mice. Although passive and active membrane properties of CA1 pyramidal cells were unchanged, iCKO mice showed enhanced susceptibility to chemically evoked seizures. Moreover, iCKO mice exhibited selective behavioral deficits in both preference for social novelty and goal-directed learning, which are consistent with ASD-like phenotype. Together, our findings show that disruption of developmental Nrp2 regulation of interneuron circuit establishment, produces ASD-like behaviors and enhanced risk for epilepsy. These results support the developmental interneuronopathy hypothesis of ASD epilepsy comorbidity.
Collapse
Affiliation(s)
- Deepak Subramanian
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA
| | - Carol Eisenberg
- Department of Biological Sciences, Rutgers University, Newark, NJ
| | - Andrew Huang
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA
| | - Jiyeon Baek
- Department of Biological Sciences, Rutgers University, Newark, NJ
| | - Haniya Naveed
- Department of Biological Sciences, Rutgers University, Newark, NJ
| | - Samiksha Komatireddy
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA
| | | | - Tracy S. Tran
- Department of Biological Sciences, Rutgers University, Newark, NJ
| | - Vijayalakshmi Santhakumar
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA
| |
Collapse
|
24
|
Nussinov R, Yavuz BR, Jang H. Single cell spatial biology over developmental time can decipher pediatric brain pathologies. Neurobiol Dis 2024; 199:106597. [PMID: 38992777 DOI: 10.1016/j.nbd.2024.106597] [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: 03/27/2024] [Revised: 06/18/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024] Open
Abstract
Pediatric low grade brain tumors and neurodevelopmental disorders share proteins, signaling pathways, and networks. They also share germline mutations and an impaired prenatal differentiation origin. They may differ in the timing of the events and proliferation. We suggest that their pivotal distinct, albeit partially overlapping, outcomes relate to the cell states, which depend on their spatial location, and timing of gene expression during brain development. These attributes are crucial as the brain develops sequentially, and single-cell spatial organization influences cell state, thus function. Our underlying premise is that the root cause in neurodevelopmental disorders and pediatric tumors is impaired prenatal differentiation. Data related to pediatric brain tumors, neurodevelopmental disorders, brain cell (sub)types, locations, and timing of expression in the developing brain are scant. However, emerging single cell technologies, including transcriptomic, spatial biology, spatial high-resolution imaging performed over the brain developmental time, could be transformational in deciphering brain pathologies thereby pharmacology.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
25
|
Hanson KL, Greiner DM, Schumann CM, Semendeferi K. Inhibitory Systems in Brain Evolution: Pathways of Vulnerability in Neurodevelopmental Disorders. BRAIN, BEHAVIOR AND EVOLUTION 2024; 100:29-48. [PMID: 39137740 PMCID: PMC11822052 DOI: 10.1159/000540865] [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: 10/31/2023] [Accepted: 08/08/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND The evolution of the primate brain has been characterized by the reorganization of key structures and circuits underlying derived specializations in sensory systems, as well as social behavior and cognition. Among these, expansion and elaboration of the prefrontal cortex has been accompanied by alterations to the connectivity and organization of subcortical structures, including the striatum and amygdala, underlying advanced aspects of executive function, inhibitory behavioral control, and socioemotional cognition seen in our lineages. At the cellular level, the primate brain has further seen an increase in the diversity and number of inhibitory GABAergic interneurons. A prevailing hypothesis holds that disruptions in the balance of excitatory to inhibitory activity in the brain underlies the pathophysiology of many neurodevelopmental and psychiatric disorders. SUMMARY This review highlights the evolution of inhibitory brain systems and circuits and suggests that recent evolutionary modifications to GABAergic circuitry may provide the substrate for vulnerability to aberrant neurodevelopment. We further discuss how modifications to primate and human social organization and life history may shape brain development in ways that contribute to neurodivergence and the origins of neurodevelopmental disorders. KEY MESSAGES Many brain systems have seen functional reorganization in the mammalian, primate, and human brain. Alterations to inhibitory circuitry in frontostriatal and frontoamygdalar systems support changes in social behavior and cognition. Increased complexity of inhibitory systems may underlie vulnerabilities to neurodevelopmental and psychiatric disorders, including autism and schizophrenia. Changes observed in Williams syndrome may further elucidate the mechanisms by which alterations in inhibitory systems lead to changes in behavior and cognition. Developmental processes, including altered neuroimmune function and age-related vulnerability of inhibitory cells and synapses, may lead to worsening symptomatology in neurodevelopmental and psychiatric disorders. BACKGROUND The evolution of the primate brain has been characterized by the reorganization of key structures and circuits underlying derived specializations in sensory systems, as well as social behavior and cognition. Among these, expansion and elaboration of the prefrontal cortex has been accompanied by alterations to the connectivity and organization of subcortical structures, including the striatum and amygdala, underlying advanced aspects of executive function, inhibitory behavioral control, and socioemotional cognition seen in our lineages. At the cellular level, the primate brain has further seen an increase in the diversity and number of inhibitory GABAergic interneurons. A prevailing hypothesis holds that disruptions in the balance of excitatory to inhibitory activity in the brain underlies the pathophysiology of many neurodevelopmental and psychiatric disorders. SUMMARY This review highlights the evolution of inhibitory brain systems and circuits and suggests that recent evolutionary modifications to GABAergic circuitry may provide the substrate for vulnerability to aberrant neurodevelopment. We further discuss how modifications to primate and human social organization and life history may shape brain development in ways that contribute to neurodivergence and the origins of neurodevelopmental disorders. KEY MESSAGES Many brain systems have seen functional reorganization in the mammalian, primate, and human brain. Alterations to inhibitory circuitry in frontostriatal and frontoamygdalar systems support changes in social behavior and cognition. Increased complexity of inhibitory systems may underlie vulnerabilities to neurodevelopmental and psychiatric disorders, including autism and schizophrenia. Changes observed in Williams syndrome may further elucidate the mechanisms by which alterations in inhibitory systems lead to changes in behavior and cognition. Developmental processes, including altered neuroimmune function and age-related vulnerability of inhibitory cells and synapses, may lead to worsening symptomatology in neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Kari L. Hanson
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
- MIND Institute, UC Davis School of Medicine, Sacramento, CA, USA
| | - Demi M.Z. Greiner
- Department of Anthropology, University of California San Diego, La Jolla, CA, USA
| | - Cynthia M. Schumann
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, Sacramento, CA, USA
- MIND Institute, UC Davis School of Medicine, Sacramento, CA, USA
| | - Katerina Semendeferi
- Department of Anthropology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
26
|
Wu MW, Kourdougli N, Portera-Cailliau C. Network state transitions during cortical development. Nat Rev Neurosci 2024; 25:535-552. [PMID: 38783147 PMCID: PMC11825063 DOI: 10.1038/s41583-024-00824-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Mammalian cortical networks are active before synaptogenesis begins in earnest, before neuronal migration is complete, and well before an animal opens its eyes and begins to actively explore its surroundings. This early activity undergoes several transformations during development. The most important of these is a transition from episodic synchronous network events, which are necessary for patterning the neocortex into functionally related modules, to desynchronized activity that is computationally more powerful and efficient. Network desynchronization is perhaps the most dramatic and abrupt developmental event in an otherwise slow and gradual process of brain maturation. In this Review, we summarize what is known about the phenomenology of developmental synchronous activity in the rodent neocortex and speculate on the mechanisms that drive its eventual desynchronization. We argue that desynchronization of network activity is a fundamental step through which the cortex transitions from passive, bottom-up detection of sensory stimuli to active sensory processing with top-down modulation.
Collapse
Affiliation(s)
- Michelle W Wu
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Neuroscience Interdepartmental Graduate Program, University of California Los Angeles, Los Angeles, CA, USA
- UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nazim Kourdougli
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
27
|
Machold R, Rudy B. Genetic approaches to elucidating cortical and hippocampal GABAergic interneuron diversity. Front Cell Neurosci 2024; 18:1414955. [PMID: 39113758 PMCID: PMC11303334 DOI: 10.3389/fncel.2024.1414955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
GABAergic interneurons (INs) in the mammalian forebrain represent a diverse population of cells that provide specialized forms of local inhibition to regulate neural circuit activity. Over the last few decades, the development of a palette of genetic tools along with the generation of single-cell transcriptomic data has begun to reveal the molecular basis of IN diversity, thereby providing deep insights into how different IN subtypes function in the forebrain. In this review, we outline the emerging picture of cortical and hippocampal IN speciation as defined by transcriptomics and developmental origin and summarize the genetic strategies that have been utilized to target specific IN subtypes, along with the technical considerations inherent to each approach. Collectively, these methods have greatly facilitated our understanding of how IN subtypes regulate forebrain circuitry via cell type and compartment-specific inhibition and thus have illuminated a path toward potential therapeutic interventions for a variety of neurocognitive disorders.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
28
|
Kon K, Ode KL, Mano T, Fujishima H, Takahashi RR, Tone D, Shimizu C, Shiono S, Yada S, Matsuzawa K, Yoshida SY, Yoshida Garçon J, Kaneko M, Shinohara Y, Yamada RG, Shi S, Miyamichi K, Sumiyama K, Kiyonari H, Susaki EA, Ueda HR. Cortical parvalbumin neurons are responsible for homeostatic sleep rebound through CaMKII activation. Nat Commun 2024; 15:6054. [PMID: 39025867 PMCID: PMC11258272 DOI: 10.1038/s41467-024-50168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/01/2024] [Indexed: 07/20/2024] Open
Abstract
The homeostatic regulation of sleep is characterized by rebound sleep after prolonged wakefulness, but the molecular and cellular mechanisms underlying this regulation are still unknown. In this study, we show that Ca2+/calmodulin-dependent protein kinase II (CaMKII)-dependent activity control of parvalbumin (PV)-expressing cortical neurons is involved in homeostatic regulation of sleep in male mice. Prolonged wakefulness enhances cortical PV-neuron activity. Chemogenetic suppression or activation of cortical PV neurons inhibits or induces rebound sleep, implying that rebound sleep is dependent on increased activity of cortical PV neurons. Furthermore, we discovered that CaMKII kinase activity boosts the activity of cortical PV neurons, and that kinase activity is important for homeostatic sleep rebound. Here, we propose that CaMKII-dependent PV-neuron activity represents negative feedback inhibition of cortical neural excitability, which serves as the distributive cortical circuits for sleep homeostatic regulation.
Collapse
Affiliation(s)
- Kazuhiro Kon
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Kennedy Krieger Institute, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koji L Ode
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Tomoyuki Mano
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Information Physics and Computing, Graduate School of Information Science and Technology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Computational Neuroethology Unit, Okinawa Institute of Science and Technology, Onna, Okinawa, Japan
| | - Hiroshi Fujishima
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan
| | - Riina R Takahashi
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Daisuke Tone
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Chika Shimizu
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Shinnosuke Shiono
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Saori Yada
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kyoko Matsuzawa
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Shota Y Yoshida
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
| | - Junko Yoshida Garçon
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research (BDR), Chuou-ku, Kobe, Hyogo, Japan
| | - Yuta Shinohara
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Rikuhiro G Yamada
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan
| | - Shoi Shi
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- International Institute for Integrative Sleep Medicine (IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connectomics, RIKEN Center for Biosystems Dynamics Research (BDR), Chuou-ku, Kobe, Hyogo, Japan
| | - Kenta Sumiyama
- Laboratory for Mouse Genetic Engineering, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Laboratory of Animal Genetics and Breeding, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa-ku, Nagoya, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research (BDR), Chuou-ku, Kobe, Hyogo, Japan
| | - Etsuo A Susaki
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Hiroki R Ueda
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, Japan.
- Department of Information Physics and Computing, Graduate School of Information Science and Technology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
- Department of Systems Biology, Institute of Life Science, Kurume University, Kurume, Fukuoka, Japan.
| |
Collapse
|
29
|
Xu QW, Larosa A, Wong TP. Roles of AMPA receptors in social behaviors. Front Synaptic Neurosci 2024; 16:1405510. [PMID: 39056071 PMCID: PMC11269240 DOI: 10.3389/fnsyn.2024.1405510] [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: 03/23/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
As a crucial player in excitatory synaptic transmission, AMPA receptors (AMPARs) contribute to the formation, regulation, and expression of social behaviors. AMPAR modifications have been associated with naturalistic social behaviors, such as aggression, sociability, and social memory, but are also noted in brain diseases featuring impaired social behavior. Understanding the role of AMPARs in social behaviors is timely to reveal therapeutic targets for treating social impairment in disorders, such as autism spectrum disorder and schizophrenia. In this review, we will discuss the contribution of the molecular composition, function, and plasticity of AMPARs to social behaviors. The impact of targeting AMPARs in treating brain disorders will also be discussed.
Collapse
Affiliation(s)
- Qi Wei Xu
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Amanda Larosa
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Tak Pan Wong
- Douglas Hospital Research Centre, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
30
|
Qiu H, Miraucourt LS, Petitjean H, Xu M, Theriault C, Davidova A, Soubeyre V, Poulen G, Lonjon N, Vachiery-Lahaye F, Bauchet L, Levesque-Damphousse P, Estall JL, Bourinet E, Sharif-Naeini R. Parvalbumin gates chronic pain through the modulation of firing patterns in inhibitory neurons. Proc Natl Acad Sci U S A 2024; 121:e2403777121. [PMID: 38916998 PMCID: PMC11228497 DOI: 10.1073/pnas.2403777121] [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: 02/22/2024] [Accepted: 05/14/2024] [Indexed: 06/27/2024] Open
Abstract
Spinal cord dorsal horn inhibition is critical to the processing of sensory inputs, and its impairment leads to mechanical allodynia. How this decreased inhibition occurs and whether its restoration alleviates allodynic pain are poorly understood. Here, we show that a critical step in the loss of inhibitory tone is the change in the firing pattern of inhibitory parvalbumin (PV)-expressing neurons (PVNs). Our results show that PV, a calcium-binding protein, controls the firing activity of PVNs by enabling them to sustain high-frequency tonic firing patterns. Upon nerve injury, PVNs transition to adaptive firing and decrease their PV expression. Interestingly, decreased PV is necessary and sufficient for the development of mechanical allodynia and the transition of PVNs to adaptive firing. This transition of the firing pattern is due to the recruitment of calcium-activated potassium (SK) channels, and blocking them during chronic pain restores normal tonic firing and alleviates chronic pain. Our findings indicate that PV is essential for controlling the firing pattern of PVNs and for preventing allodynia. Developing approaches to manipulate these mechanisms may lead to different strategies for chronic pain relief.
Collapse
Affiliation(s)
- Haoyi Qiu
- Department of Physiology, McGill University, Montreal, QCH3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QCH3A 2B4, Canada
| | - Loïs S. Miraucourt
- Department of Physiology, McGill University, Montreal, QCH3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QCH3A 2B4, Canada
| | - Hugues Petitjean
- Department of Physiology, McGill University, Montreal, QCH3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QCH3A 2B4, Canada
| | - Mengyi Xu
- Department of Physiology, McGill University, Montreal, QCH3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QCH3A 2B4, Canada
| | - Catherine Theriault
- Department of Physiology, McGill University, Montreal, QCH3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QCH3A 2B4, Canada
| | - Albena Davidova
- Department of Physiology, McGill University, Montreal, QCH3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QCH3A 2B4, Canada
| | - Vanessa Soubeyre
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier34000, France
| | - Gaetan Poulen
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier34295, France
| | - Nicolas Lonjon
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier34295, France
| | - Florence Vachiery-Lahaye
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier34295, France
| | - Luc Bauchet
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier34000, France
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier34295, France
| | | | - Jennifer L. Estall
- Institut de Recherches Cliniques de Montréal, Montreal, QCH2W 1R7, Canada
| | - Emmanuel Bourinet
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier34000, France
| | - Reza Sharif-Naeini
- Department of Physiology, McGill University, Montreal, QCH3G 1Y6, Canada
- Alan Edwards Center for Research on Pain, McGill University, Montreal, QCH3A 2B4, Canada
| |
Collapse
|
31
|
Martinez JD, Wilson LG, Brancaleone WP, Peterson KG, Popke DS, Garzon VC, Perez Tremble RE, Donnelly MJ, Mendez Ortega SL, Torres D, Shaver JJ, Jiang S, Yang Z, Aton SJ. Hypnotic treatment improves sleep architecture and EEG disruptions and rescues memory deficits in a mouse model of fragile X syndrome. Cell Rep 2024; 43:114266. [PMID: 38787724 PMCID: PMC11910971 DOI: 10.1016/j.celrep.2024.114266] [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: 07/07/2023] [Revised: 12/20/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Fragile X syndrome (FXS) is associated with disrupted cognition and sleep abnormalities. Sleep loss negatively impacts cognitive function, and one untested possibility is that disrupted cognition in FXS is exacerbated by abnormal sleep. We tested whether ML297, a hypnotic acting on G-protein-activated inward-rectifying potassium (GIRK) channels, could reverse sleep phenotypes and disrupted memory in Fmr1-/y mice. Fmr1-/y mice exhibit reduced non-rapid eye movement (NREM) sleep and fragmented NREM architecture, altered sleep electroencephalogram (EEG) oscillations, and reduced EEG coherence between cortical areas; these are partially reversed following ML297 administration. Treatment following contextual fear or spatial learning restores disrupted memory consolidation in Fmr1-/y mice. During memory recall, Fmr1-/y mice show an altered balance of activity among hippocampal principal neurons vs. parvalbumin-expressing interneurons; this is partially reversed by ML297. Because sleep disruption could impact neurophysiological phenotypes in FXS, augmenting sleep may improve disrupted cognition in this disorder.
Collapse
Affiliation(s)
- Jessy D Martinez
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lydia G Wilson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William P Brancaleone
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn G Peterson
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Donald S Popke
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | - Valentina Caicedo Garzon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Roxanne E Perez Tremble
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcus J Donnelly
- Undergraduate Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Daniel Torres
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Shaver
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sha Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhongying Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara J Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
32
|
Deng J, Labarta-Bajo L, Brandebura AN, Kahn SB, Pinto AFM, Diedrich JK, Allen NJ. Suppression of astrocyte BMP signaling improves fragile X syndrome molecular signatures and functional deficits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599752. [PMID: 38979341 PMCID: PMC11230279 DOI: 10.1101/2024.06.19.599752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Fragile X syndrome (FXS) is a monogenic neurodevelopmental disorder with manifestations spanning molecular, neuroanatomical, and behavioral changes. Astrocytes contribute to FXS pathogenesis and show hundreds of dysregulated genes and proteins; targeting upstream pathways mediating astrocyte changes in FXS could therefore be a point of intervention. To address this, we focused on the bone morphogenetic protein (BMP) pathway, which is upregulated in FXS astrocytes. We generated a conditional KO (cKO) of Smad4 in astrocytes to suppress BMP signaling, and found this lessens audiogenic seizure severity in FXS mice. To ask how this occurs on a molecular level, we performed in vivo transcriptomic and proteomic profiling of cortical astrocytes, finding upregulation of metabolic pathways, and downregulation of secretory machinery and secreted proteins in FXS astrocytes, with these alterations no longer present when BMP signaling is suppressed. Functionally, astrocyte Smad4 cKO restores deficits in inhibitory synapses present in FXS auditory cortex. Thus, astrocytes contribute to FXS molecular and functional phenotypes, and targeting astrocytes can mitigate FXS symptoms.
Collapse
Affiliation(s)
- James Deng
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Lara Labarta-Bajo
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ashley N Brandebura
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Samuel B Kahn
- Department of Biology, University of California, San Diego, La Jolla, CA, USA
| | - Antonio F M Pinto
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene K Diedrich
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
33
|
Mòdol L, Moissidis M, Selten M, Oozeer F, Marín O. Somatostatin interneurons control the timing of developmental desynchronization in cortical networks. Neuron 2024; 112:2015-2030.e5. [PMID: 38599213 DOI: 10.1016/j.neuron.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/21/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Synchronous neuronal activity is a hallmark of the developing brain. In the mouse cerebral cortex, activity decorrelates during the second week of postnatal development, progressively acquiring the characteristic sparse pattern underlying the integration of sensory information. The maturation of inhibition seems critical for this process, but the interneurons involved in this crucial transition of network activity in the developing cortex remain unknown. Using in vivo longitudinal two-photon calcium imaging during the period that precedes the change from highly synchronous to decorrelated activity, we identify somatostatin-expressing (SST+) interneurons as critical modulators of this switch in mice. Modulation of the activity of SST+ cells accelerates or delays the decorrelation of cortical network activity, a process that involves regulating the maturation of parvalbumin-expressing (PV+) interneurons. SST+ cells critically link sensory inputs with local circuits, controlling the neural dynamics in the developing cortex while modulating the integration of other interneurons into nascent cortical circuits.
Collapse
Affiliation(s)
- Laura Mòdol
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Monika Moissidis
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
34
|
Marshall AH, Boyle DJ, Hanson MA, Nagarajan D, Bibi N, Safa A, Johantges AC, Wester JC. Arid1b haploinsufficiency in cortical inhibitory interneurons causes cell-type-dependent changes in cellular and synaptic development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597984. [PMID: 38895260 PMCID: PMC11185764 DOI: 10.1101/2024.06.07.597984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Autism spectrum disorder (ASD) presents with diverse cognitive and behavioral abnormalities beginning during early development. Although the neural circuit mechanisms remain unclear, recent work suggests pathology in cortical inhibitory interneurons (INs) plays a crucial role. However, we lack fundamental information regarding changes in the physiology of synapses to and from INs in ASD. Here, we used transgenic mice to conditionally knockout one copy of the high confidence ASD risk gene Arid1b from the progenitors of parvalbumin-expressing fast-spiking (PV-FS) INs and somatostatin-expressing non-fast-spiking (SST-NFS) INs. In brain slices, we performed paired whole-cell recordings between INs and excitatory projection neurons (PNs) to investigate changes in synaptic physiology. In neonates, we found reduced synaptic input to INs but not PNs, with a concomitant reduction in the frequency of spontaneous network events, which are driven by INs in immature circuits. In mature mice, we found a reduction in the number of PV-FS INs in cortical layers 2/3 and 5. However, changes in PV-FS IN synaptic physiology were cortical layer and PN cell-type dependent. In layer 5, synapses from PV-FS INs to subcortical-projecting PNs were weakened. In contrast, in layer 2/3, synapses to and from PV-FS INs and corticocortical-projecting PNs were strengthened, leading to enhanced feedforward inhibition of input from layer 4. Finally, we found a novel synaptic deficit among SST-NFS INs, in which excitatory synapses from layer 2/3 PNs failed to facilitate. Our data highlight that changes in unitary synaptic dynamics among INs in ASD depend on neuronal cell-type.
Collapse
|
35
|
Marshall AH, Hanson MA, Boyle DJ, Nagarajan D, Bibi N, Fitzgerald J, Gaitten E, Kokiko-Cochran ON, Gu B, Wester JC. Arid1b haploinsufficiency in pyramidal neurons causes cellular and circuit changes in neocortex but is not sufficient to produce behavioral or seizure phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597344. [PMID: 38895205 PMCID: PMC11185765 DOI: 10.1101/2024.06.04.597344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Arid1b is a high confidence risk gene for autism spectrum disorder that encodes a subunit of a chromatin remodeling complex expressed in neuronal progenitors. Haploinsufficiency causes a broad range of social, behavioral, and intellectual disability phenotypes, including Coffin-Siris syndrome. Recent work using transgenic mouse models suggests pathology is due to deficits in proliferation, survival, and synaptic development of cortical neurons. However, there is conflicting evidence regarding the relative roles of excitatory projection neurons and inhibitory interneurons in generating abnormal cognitive and behavioral phenotypes. Here, we conditionally knocked out either one or both copies of Arid1b from excitatory projection neuron progenitors and systematically investigated the effects on intrinsic membrane properties, synaptic physiology, social behavior, and seizure susceptibility. We found that disrupting Arid1b expression in excitatory neurons alters their membrane properties, including hyperpolarizing action potential threshold; however, these changes depend on neuronal subtype. Using paired whole-cell recordings, we found increased synaptic connectivity rate between projection neurons. Furthermore, we found reduced strength of excitatory synapses to parvalbumin (PV)-expression inhibitory interneurons. These data suggest an increase in the ratio of excitation to inhibition. However, the strength of inhibitory synapses from PV interneurons to excitatory neurons was enhanced, which may rebalance this ratio. Indeed, Arid1b haploinsufficiency in projection neurons was insufficient to cause social deficits and seizure phenotypes observed in a preclinical germline haploinsufficient mouse model. Our data suggest that while excitatory projection neurons likely contribute to autistic phenotypes, pathology in these cells is not the primary cause.
Collapse
|
36
|
Espinoza F, Carrazana R, Retamal-Fredes E, Ávila D, Papes F, Muotri AR, Ávila A. Tcf4 dysfunction alters dorsal and ventral cortical neurogenesis in Pitt-Hopkins syndrome mouse model showing sexual dimorphism. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167178. [PMID: 38636614 DOI: 10.1016/j.bbadis.2024.167178] [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: 11/15/2023] [Revised: 03/28/2024] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by haploinsufficiency of transcription factor 4 (TCF4). In this work, we focused on the cerebral cortex and investigated in detail the progenitor cell dynamics and the outcome of neurogenesis in a PTHS mouse model. Labeling and quantification of progenitors and newly generated neurons at various time points during embryonic development revealed alterations affecting the dynamic of cortical progenitors since the earliest stages of cortex formation in PTHS mice. Consequently, establishment of neuronal populations and layering of the cortex were found to be altered in heterozygotes subjects at birth. Interestingly, defective layering process of pyramidal neurons was partially rescued by reintroducing TCF4 expression using focal in utero electroporation in the cerebral cortex. Coincidentally with a defective dorsal neurogenesis, we found that ventral generation of interneurons was also defective in this model, which may lead to an excitation/inhibition imbalance in PTHS. Overall, sex-dependent differences were detected with more marked effects evidenced in males compared with females. All of this contributes to expand our understanding of PTHS, paralleling the advances of research in autism spectrum disorder and further validating the PTHS mouse model as an important tool to advance preclinical studies.
Collapse
Affiliation(s)
- Francisca Espinoza
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción (UCSC), Concepción, Chile
| | - Ramón Carrazana
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción (UCSC), Concepción, Chile
| | - Eduardo Retamal-Fredes
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción (UCSC), Concepción, Chile
| | - Denisse Ávila
- Department of Biochemical Engineering, University College of London (UCL), London, UK
| | - Fabio Papes
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ariel Ávila
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción (UCSC), Concepción, Chile.
| |
Collapse
|
37
|
Bhandari K, Kanodia H, Donato F, Caroni P. Selective vulnerability of the ventral hippocampus-prelimbic cortex axis parvalbumin interneuron network underlies learning deficits of fragile X mice. Cell Rep 2024; 43:114124. [PMID: 38630591 DOI: 10.1016/j.celrep.2024.114124] [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: 10/06/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
High-penetrance mutations affecting mental health can involve genes ubiquitously expressed in the brain. Whether the specific patterns of dysfunctions result from ubiquitous circuit deficits or might reflect selective vulnerabilities of targetable subnetworks has remained unclear. Here, we determine how loss of ubiquitously expressed fragile X mental retardation protein (FMRP), the cause of fragile X syndrome, affects brain networks in Fmr1y/- mice. We find that in wild-type mice, area-specific knockout of FMRP in the adult mimics behavioral consequences of area-specific silencing. By contrast, the functional axis linking the ventral hippocampus (vH) to the prelimbic cortex (PreL) is selectively affected in constitutive Fmr1y/- mice. A chronic alteration in late-born parvalbumin interneuron networks across the vH-PreL axis rescued by VIP signaling specifically accounts for deficits in vH-PreL theta-band network coherence, ensemble assembly, and learning functions of Fmr1y/- mice. Therefore, vH-PreL axis function exhibits a selective vulnerability to loss of FMRP in the vH or PreL, leading to learning and memory dysfunctions in fragile X mice.
Collapse
Affiliation(s)
- Komal Bhandari
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Harsh Kanodia
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Flavio Donato
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Pico Caroni
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.
| |
Collapse
|
38
|
Morimoto K, Takahashi R, Takahashi G, Miyajima M, Nakajima K. Maternal immunoglobulins are distributed in the offspring's brain to support the maintenance of cortical interneurons in the postnatal period. Inflamm Regen 2024; 44:24. [PMID: 38750507 PMCID: PMC11094934 DOI: 10.1186/s41232-024-00336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
It is known that maternal immunoglobulins (Igs) are transferred to the offspring across the placenta. However, receiving maternal Igs, especially before the blood-brain barrier (BBB) is formed in the offspring's brain, carries the risk of transferring some brain-reactive Igs. It is thus hypothesized that there may be some unknown benefit to the offspring's brain that overweighs this risk. In this study, we show that the Ig detected in the embryonic/perinatal mouse brain is IgG not produced by the pups themselves, but is basically transferred from the mother across the placenta using the neonatal Fc receptor (FcRn) during embryonic stages. The amount of IgG in the brain gradually decreases after birth, and almost disappears within 3 weeks postnatally. IgG is detected on axon bundles, microglia, and some meningeal cells, including border-associated macrophages (BAMs), endothelial cells, and fibroblasts. Using Fcer1g knock-out (KO) mice, we show that BAMs and microglia receive maternal IgG in an Fc receptor γ chain (FcRγ)-dependent manner, but IgG on other meningeal cells and axon bundles is received independently of the FcRγ. These results suggest that maternal IgG may be used in multiple ways by different mechanisms. In maternal IgG-deficient mice, the number of interneurons in the cerebral cortex is not altered around birth but is reduced postnatally, suggesting that receipt of maternal IgG is necessary for the maintenance of cortical interneurons in the postnatal period. These data suggest that maternal IgG has an important function in the developing brain, where neither obvious inflammation nor infection is observed.
Collapse
Affiliation(s)
- Keiko Morimoto
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Rikuo Takahashi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Goro Takahashi
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Michio Miyajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
39
|
Arutiunian V, Santhosh M, Neuhaus E, Borland H, Tompkins C, Bernier RA, Bookheimer SY, Dapretto M, Gupta AR, Jack A, Jeste S, McPartland JC, Naples A, Van Horn JD, Pelphrey KA, Webb SJ. The relationship between gamma-band neural oscillations and language skills in youth with Autism Spectrum Disorder and their first-degree relatives. Mol Autism 2024; 15:19. [PMID: 38711098 PMCID: PMC11075235 DOI: 10.1186/s13229-024-00598-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Most children with Autism Spectrum Disorder (ASD) have co-occurring language impairments and some of these autism-specific language difficulties are also present in their non-autistic first-degree relatives. One of the possible neural mechanisms associated with variability in language functioning is alterations in cortical gamma-band oscillations, hypothesized to be related to neural excitation and inhibition balance. METHODS We used a high-density 128-channel electroencephalography (EEG) to register brain response to speech stimuli in a large sex-balanced sample of participants: 125 youth with ASD, 121 typically developing (TD) youth, and 40 unaffected siblings (US) of youth with ASD. Language skills were assessed with Clinical Evaluation of Language Fundamentals. RESULTS First, during speech processing, we identified significantly elevated gamma power in ASD participants compared to TD controls. Second, across all youth, higher gamma power was associated with lower language skills. Finally, the US group demonstrated an intermediate profile in both language and gamma power, with nonverbal IQ mediating the relationship between gamma power and language skills. LIMITATIONS We only focused on one of the possible neural contributors to variability in language functioning. Also, the US group consisted of a smaller number of participants in comparison to the ASD or TD groups. Finally, due to the timing issue in EEG system we have provided only non-phase-locked analysis. CONCLUSIONS Autistic youth showed elevated gamma power, suggesting higher excitation in the brain in response to speech stimuli and elevated gamma power was related to lower language skills. The US group showed an intermediate pattern of gamma activity, suggesting that the broader autism phenotype extends to neural profiles.
Collapse
Affiliation(s)
- Vardan Arutiunian
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Megha Santhosh
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Emily Neuhaus
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Heather Borland
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA
| | - Chris Tompkins
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Raphael A Bernier
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA
| | - Susan Y Bookheimer
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Mirella Dapretto
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Abha R Gupta
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Allison Jack
- Department of Psychology, George Mason University, Fairfax, VA, USA
| | - Shafali Jeste
- Department of Neurology, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | | | - Adam Naples
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - John D Van Horn
- School of Data Science, University of Virginia, Charlottesville, VA, USA
| | - Kevin A Pelphrey
- Department of Neurology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Sara Jane Webb
- Center for Child Health, Behavior and Development, Seattle Children's Research Institute, 1920 Terry Ave., Seattle, WA, 98101, USA.
- Department of Psychiatry and Behavioral Science, University of Washington, Seattle, WA, USA.
- Institute of Human Development and Disability, University of Washington, Seattle, WA, USA.
| |
Collapse
|
40
|
Dummer PD, Lee DI, Hossain S, Wang R, Evard A, Newman G, Ho C, Schneider-Mizell CM, Menon V, Au E. Multidimensional analysis of cortical interneuron synaptic features reveals underlying synaptic heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586340. [PMID: 38659827 PMCID: PMC11042224 DOI: 10.1101/2024.03.22.586340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cortical interneurons represent a diverse set of neuronal subtypes characterized in part by their striking degree of synaptic specificity. However, little is known about the extent of synaptic diversity because of the lack of unbiased methods to extract synaptic features among interneuron subtypes. Here, we develop an approach to aggregate image features from fluorescent confocal images of interneuron synapses and their post-synaptic targets, in order to characterize the heterogeneity of synapses at fine scale. We started by training a model that recognizes pre- and post-synaptic compartments and then determines the target of each genetically-identified interneuron synapse in vitro and in vivo. Our model extracts hundreds of spatial and intensity features from each analyzed synapse, constructing a multidimensional data set, consisting of millions of synapses, which allowed us to perform an unsupervised analysis on this dataset, uncovering novel synaptic subgroups. The subgroups were spatially distributed in a highly structured manner that revealed the local underlying topology of the postsynaptic environment. Dendrite-targeting subgroups were clustered onto subdomains of the dendrite along the proximal to distal axis. Soma-targeting subgroups were enriched onto different postsynaptic cell types. We also find that the two main subclasses of interneurons, basket cells and somatostatin interneurons, utilize distinct strategies to enact inhibitory coverage. Thus, our analysis of multidimensional synaptic features establishes a conceptual framework for studying interneuron synaptic diversity.
Collapse
Affiliation(s)
- Patrick D. Dummer
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Dylan I. Lee
- Department of Neurology, Columbia Irving Medical Center, New York NY, 10032
| | - Sakib Hossain
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Runsheng Wang
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Andre Evard
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Gabriel Newman
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | - Claire Ho
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
| | | | - Vilas Menon
- Department of Neurology, Columbia Irving Medical Center, New York NY, 10032
| | - Edmund Au
- Department of Pathology and Cell Biology, Columbia Irving Medical Center, New York NY, 10032
- Columbia Translation Neuroscience Initiative Fellow, Columbia Irving Medical Center, New York NY, 10032
| |
Collapse
|
41
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.575409. [PMID: 38328248 PMCID: PMC10849664 DOI: 10.1101/2024.01.27.575409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement dysregulation - a prevalent locus of brain disease etiology - in PV cells may drive disease pathogenesis, we have developed a transgenic mouse line that permits cell-type specific overexpression of the schizophrenia-associated complement component 4 (C4) gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific behavioral alterations and concomitant deficits in synaptic connectivity and excitability of PV cells of the prefrontal cortex. Using a computational network, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that C4 perturbations in fast-spiking neurons are more harmful to brain function than pan-neuronal alterations. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, Massachusetts, United States
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts, United States
- Hearing Research Center, Boston University, Boston, Massachusetts, United States
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
- Bioinformatics MS Program, Boston University, Boston, MA, United States
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, United States
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, United States
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, United States
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, Massachusetts, United States
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts, United States
- Hearing Research Center, Boston University, Boston, Massachusetts, United States
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, United States
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, United States
| |
Collapse
|
42
|
Vlasits AL, Syeda M, Wickman A, Guzman P, Schmidt TM. Atypical retinal function in a mouse model of Fragile X syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585283. [PMID: 38559003 PMCID: PMC10980068 DOI: 10.1101/2024.03.15.585283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Altered function of peripheral sensory neurons is an emerging mechanism for symptoms of autism spectrum disorders. Visual sensitivities are common in autism, but whether differences in the retina might underlie these sensitivities is not well-understood. We explored retinal function in the Fmr1 knockout model of Fragile X syndrome, focusing on a specific type of retinal neuron, the "sustained On alpha" retinal ganglion cell. We found that these cells exhibit changes in dendritic structure and dampened responses to light in the Fmr1 knockout. We show that decreased light sensitivity is due to increased inhibitory input and reduced E-I balance. The change in E-I balance supports maintenance of circuit excitability similar to what has been observed in cortex. These results show that loss of Fmr1 in the mouse retina affects sensory function of one retinal neuron type. Our findings suggest that the retina may be relevant for understanding visual function in Fragile X syndrome.
Collapse
Affiliation(s)
- Anna L Vlasits
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Department of Ophthalmology, University of Illinois, Chicago, IL, USA
- Lead contact
| | - Maria Syeda
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Annelise Wickman
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Pedro Guzman
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Department of Ophthalmology, Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
43
|
Juarez P, Salcedo-Arellano MJ, Dufour B, Martinez-Cerdeño V. Fragile X cortex is characterized by decreased parvalbumin-expressing interneurons. Cereb Cortex 2024; 34:bhae103. [PMID: 38521994 PMCID: PMC10960956 DOI: 10.1093/cercor/bhae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/25/2024] Open
Abstract
Fragile X syndrome is a genetic neurodevelopmental disorder caused by a mutation of the fragile X messenger ribonucleoprotein 1 (FMR1) gene in the X chromosome. Many fragile X syndrome cases present with autism spectrum disorder and fragile X syndrome cases account for up to 5% of all autism spectrum disorder cases. The cellular composition of the fragile X syndrome cortex is not well known. We evaluated alterations in the number of Calbindin, Calretinin, and Parvalbumin expressing interneurons across 5 different cortical areas, medial prefrontal cortex (BA46), primary somatosensory cortex (BA3), primary motor cortex (BA4), superior temporal cortex (BA22), and anterior cingulate cortex (BA24) of fragile X syndrome and neurotypical brains. Compared with neurotypical cases, fragile X syndrome brains displayed a significant reduction in the number of PV+ interneurons in all areas and of CR+ interneurons in BA22 and BA3. The number of CB+ interneurons did not differ. These findings are the first to demonstrate that fragile X syndrome brains are characterized by cortical wide PV+ interneuron deficits across multiple cortical areas. These add to the idea that deficits in PV+ interneurons could disrupt the cortical balance and promote clinical deficits in fragile X syndrome patients and help to develop novel therapies for neurodevelopmental disorders like fragile X syndrome and autism spectrum disorder.
Collapse
Affiliation(s)
- Pablo Juarez
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
| | - Maria Jimena Salcedo-Arellano
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
| | - Brett Dufour
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
| | - Veronica Martinez-Cerdeño
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine; Institute for Pediatric Regenerative Medicine and Shriners Hospitals for Children of Northern CaliforniaSacramento, CA 95817, United States
- MIND Institute, University of California, Davis, Sacramento, CA 95817, United States
| |
Collapse
|
44
|
García-Cerro S, Gómez-Garrido A, Garcia G, Crespo-Facorro B, Brites D. Exploratory Analysis of MicroRNA Alterations in a Neurodevelopmental Mouse Model for Autism Spectrum Disorder and Schizophrenia. Int J Mol Sci 2024; 25:2786. [PMID: 38474035 DOI: 10.3390/ijms25052786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
MicroRNAs (miRNAs) play a crucial role in the regulation of gene expression levels and have been implicated in the pathogenesis of autism spectrum disorder (ASD) and schizophrenia (SCZ). In this study, we examined the adult expression profiles of specific miRNAs in the prefrontal cortex (PFC) of a neurodevelopmental mouse model for ASD and SCZ that mimics perinatal pathology, such as NMDA receptor hypofunction, and exhibits behavioral and neurophysiological phenotypes related to these disorders during adulthood. To model the early neuropathogenesis of the disorders, mouse pups were administered subcutaneously with ketamine (30 mg/Kg) at postnatal days 7, 9, and 11. We focused on a set of miRNAs most frequently altered in ASD (miR-451a and miR-486-3p) and in SCZ (miR-132-3p and miR-137-3p) according to human studies. Additionally, we explored miRNAs whose alterations have been identified in both disorders (miR-21-5p, miR-92a-2-5p, miR-144-3p, and miR-146a-5p). We placed particular emphasis on studying the sexual dimorphism in the dynamics of these miRNAs. Our findings revealed significant alterations in the PFC of this ASD- and SCZ-like mouse model. Specifically, we observed upregulated miR-451a and downregulated miR-137-3p. Furthermore, we identified sexual dimorphism in the expression of miR-132-3p, miR-137-3p, and miR-92a-2-5p. From a translational perspective, our results emphasize the potential involvement of miR-92a-2-5p, miR-132-3p, miR-137-3p, and miR-451a in the pathophysiology of ASD and SCZ and strengthen their potential as biomarkers and therapeutic targets of such disorders.
Collapse
Affiliation(s)
- Susana García-Cerro
- Translational Psychiatry Group, Ibis-Biomedicine Institute of Sevilla-CSIC, Manuel Siurot AV, 41013 Seville, Spain
- Spanish Network for Research in Mental Health (CIBERSAM), Monforte de Lemos AV, 3-5, 28029 Madrid, Spain
| | - Ana Gómez-Garrido
- Translational Psychiatry Group, Ibis-Biomedicine Institute of Sevilla-CSIC, Manuel Siurot AV, 41013 Seville, Spain
- Spanish Network for Research in Mental Health (CIBERSAM), Monforte de Lemos AV, 3-5, 28029 Madrid, Spain
| | - Gonçalo Garcia
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Benedicto Crespo-Facorro
- Translational Psychiatry Group, Ibis-Biomedicine Institute of Sevilla-CSIC, Manuel Siurot AV, 41013 Seville, Spain
- Spanish Network for Research in Mental Health (CIBERSAM), Monforte de Lemos AV, 3-5, 28029 Madrid, Spain
- Mental Health Unit, Virgen del Rocio University Hospital, Manuel Siurot AV, 41013 Seville, Spain
- Department of Psychiatry, Faculty of Medicine, University of Seville, Sánchez Pizjuán AV, 41013 Seville, Spain
| | - Dora Brites
- Neuroinflammation, Signaling and Neuroregeneration Lab, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| |
Collapse
|
45
|
Rhodes CT, Asokumar D, Sohn M, Naskar S, Elisha L, Stevenson P, Lee DR, Zhang Y, Rocha PP, Dale RK, Lee S, Petros TJ. Loss of Ezh2 in the medial ganglionic eminence alters interneuron fate, cell morphology and gene expression profiles. Front Cell Neurosci 2024; 18:1334244. [PMID: 38419656 PMCID: PMC10899338 DOI: 10.3389/fncel.2024.1334244] [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: 11/06/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Enhancer of zeste homolog 2 (Ezh2) is responsible for trimethylation of histone 3 at lysine 27 (H3K27me3), resulting in repression of gene expression. Here, we explore the role of Ezh2 in forebrain GABAergic interneuron development. Methods We removed Ezh2 in the MGE by generating Nkx2-1Cre;Ezh2 conditional knockout mice. We then characterized changes in MGE-derived interneuron fate and electrophysiological properties in juvenile mice, as well as alterations in gene expression, chromatin accessibility and histone modifications in the MGE. Results Loss of Ezh2 increases somatostatin-expressing (SST+) and decreases parvalbumin-expressing (PV+) interneurons in the forebrain. We observe fewer MGE-derived interneurons in the first postnatal week, indicating reduced interneuron production. Intrinsic electrophysiological properties in SST+ and PV+ interneurons are normal, but PV+ interneurons display increased axonal complexity in Ezh2 mutant mice. Single nuclei multiome analysis revealed differential gene expression patterns in the embryonic MGE that are predictive of these cell fate changes. Lastly, CUT&Tag analysis revealed that some genomic loci are particularly resistant or susceptible to shifts in H3K27me3 levels in the absence of Ezh2, indicating differential selectivity to epigenetic perturbation. Discussion Thus, loss of Ezh2 in the MGE alters interneuron fate, morphology, and gene expression and regulation. These findings have important implications for both normal development and potentially in disease etiologies.
Collapse
Affiliation(s)
- Christopher T Rhodes
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Dhanya Asokumar
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
- Unit on Genome Structure and Regulation, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Mira Sohn
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Shovan Naskar
- Unit on Functional Neural Circuits, National Institute of Mental Health (NIMH), NIH, Bethesda, MD, United States
| | - Lielle Elisha
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Parker Stevenson
- Unit on Functional Neural Circuits, National Institute of Mental Health (NIMH), NIH, Bethesda, MD, United States
| | - Dongjin R Lee
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Yajun Zhang
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Pedro P Rocha
- Unit on Genome Structure and Regulation, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
- National Cancer Institute (NCI), NIH, Bethesda, MD, United States
| | - Ryan K Dale
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Soohyun Lee
- Unit on Functional Neural Circuits, National Institute of Mental Health (NIMH), NIH, Bethesda, MD, United States
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| |
Collapse
|
46
|
Palicz R, Pater B, Truschow P, Witte M, Staiger JF. Intersectional strategy to study cortical inhibitory parvalbumin-expressing interneurons. Sci Rep 2024; 14:2829. [PMID: 38310185 PMCID: PMC10838283 DOI: 10.1038/s41598-024-52901-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/24/2024] [Indexed: 02/05/2024] Open
Abstract
Parvalbumin-expressing (PV) interneurons are key neuronal elements to a global excitatory-inhibitory balance in normal cortical functioning. To better understand the circuit functions of PV interneurons, reliable animal models are needed. This study investigated the sensitivity and specificity of the most frequently used PV-Cre/tdTomato mouse line in this regard. The colocalization of the transgene (tdTomato) with the parvalbumin protein, with GAD1 (a conclusive inhibitory cell marker) and Vglut1 (a conclusive excitatory cell marker) as well as with a marker for perineuronal nets (WFA) was assessed and a substantial proportion of layer 5 PV neurons was found to be excitatory and not inhibitory in the PV-Cre/tdTomato mouse. The intersectional transgenic mouse line Vgat-Cre/PV-Flp/tdTomato provided a solution, since no colocalization of tdTomato with the Vglut1 probe was found there. In conclusion, the Vgat-Cre/PV-Flp/tdTomato mouse line seems to be a more reliable animal model for functional studies of GABAergic PV interneurons.
Collapse
Affiliation(s)
- Rebeka Palicz
- Center Anatomy, Institute for Neuroanatomy, University of Göttingen, Göttingen, Germany.
| | - Bettina Pater
- Center Anatomy, Institute for Neuroanatomy, University of Göttingen, Göttingen, Germany
| | - Pavel Truschow
- Center Anatomy, Institute for Neuroanatomy, University of Göttingen, Göttingen, Germany
| | - Mirko Witte
- Center Anatomy, Institute for Neuroanatomy, University of Göttingen, Göttingen, Germany
| | - Jochen F Staiger
- Center Anatomy, Institute for Neuroanatomy, University of Göttingen, Göttingen, Germany
| |
Collapse
|
47
|
Rylaarsdam L, Rakotomamonjy J, Pope E, Guemez-Gamboa A. iPSC-derived models of PACS1 syndrome reveal transcriptional and functional deficits in neuron activity. Nat Commun 2024; 15:827. [PMID: 38280846 PMCID: PMC10821916 DOI: 10.1038/s41467-024-44989-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 01/11/2024] [Indexed: 01/29/2024] Open
Abstract
PACS1 syndrome is a neurodevelopmental disorder characterized by intellectual disability and distinct craniofacial abnormalities resulting from a de novo p.R203W variant in phosphofurin acidic cluster sorting protein 1 (PACS1). PACS1 is known to have functions in the endosomal pathway and nucleus, but how the p.R203W variant affects developing neurons is not fully understood. Here we differentiated stem cells towards neuronal models including cortical organoids to investigate the impact of the PACS1 syndrome-causing variant on neurodevelopment. While few deleterious effects were detected in PACS1(+/R203W) neural precursors, mature PACS1(+/R203W) glutamatergic neurons exhibited impaired expression of genes involved in synaptic signaling processes. Subsequent characterization of neural activity using calcium imaging and multielectrode arrays revealed the p.R203W PACS1 variant leads to a prolonged neuronal network burst duration mediated by an increased interspike interval. These findings demonstrate the impact of the PACS1 p.R203W variant on developing human neural tissue and uncover putative electrophysiological underpinnings of disease.
Collapse
Affiliation(s)
- Lauren Rylaarsdam
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jennifer Rakotomamonjy
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eleanor Pope
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alicia Guemez-Gamboa
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
48
|
Chen H, Wang YD, Blan AW, Almanza-Fuerte EP, Bonkowski ES, Bajpai R, Pruett-Miller SM, Mefford HC. Patient derived model of UBA5-associated encephalopathy identifies defects in neurodevelopment and highlights potential therapies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577254. [PMID: 38328212 PMCID: PMC10849720 DOI: 10.1101/2024.01.25.577254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
UBA5 encodes for the E1 enzyme of the UFMylation cascade, which plays an essential role in ER homeostasis. The clinical phenotypes of UBA5-associated encephalopathy include developmental delays, epilepsy and intellectual disability. To date, there is no humanized neuronal model to study the cellular and molecular consequences of UBA5 pathogenic variants. We developed and characterized patient-derived cortical organoid cultures and identified defects in GABAergic interneuron development. We demonstrated aberrant neuronal firing and microcephaly phenotypes in patient-derived organoids. Mechanistically, we show that ER homeostasis is perturbed along with exacerbated unfolded protein response pathway in cells and organoids expressing UBA5 pathogenic variants. We also assessed two gene expression modalities that augmented UBA5 expression to rescue aberrant molecular and cellular phenotypes. Our study provides a novel humanized model that allows further investigations of UBA5 variants in the brain and highlights novel systemic approaches to alleviate cellular aberrations for this rare, developmental disorder.
Collapse
Affiliation(s)
- Helen Chen
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis TN, USA
| | - Aidan W. Blan
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Edith P. Almanza-Fuerte
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Emily S. Bonkowski
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Richa Bajpai
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis TN, USA
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis TN, USA
| | - Shondra M. Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis TN, USA
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis TN, USA
| | - Heather C. Mefford
- Center for Pediatric Neurological Disease Research, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
49
|
Janz P, Knoflach F, Bleicher K, Belli S, Biemans B, Schnider P, Ebeling M, Grundschober C, Benekareddy M. Selective oxytocin receptor activation prevents prefrontal circuit dysfunction and social behavioral alterations in response to chronic prefrontal cortex activation in male rats. Front Cell Neurosci 2023; 17:1286552. [PMID: 38145283 PMCID: PMC10745491 DOI: 10.3389/fncel.2023.1286552] [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: 08/31/2023] [Accepted: 11/08/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Social behavioral changes are a hallmark of several neurodevelopmental and neuropsychiatric conditions, nevertheless the underlying neural substrates of such dysfunction remain poorly understood. Building evidence points to the prefrontal cortex (PFC) as one of the key brain regions that orchestrates social behavior. We used this concept with the aim to develop a translational rat model of social-circuit dysfunction, the chronic PFC activation model (CPA). Methods Chemogenetic designer receptor hM3Dq was used to induce chronic activation of the PFC over 10 days, and the behavioral and electrophysiological signatures of prolonged PFC hyperactivity were evaluated. To test the sensitivity of this model to pharmacological interventions on longer timescales, and validate its translational potential, the rats were treated with our novel highly selective oxytocin receptor (OXTR) agonist RO6958375, which is not activating the related vasopressin V1a receptor. Results CPA rats showed reduced sociability in the three-chamber sociability test, and a concomitant decrease in neuronal excitability and synaptic transmission within the PFC as measured by electrophysiological recordings in acute slice preparation. Sub-chronic treatment with a low dose of the novel OXTR agonist following CPA interferes with the emergence of PFC circuit dysfunction, abnormal social behavior and specific transcriptomic changes. Discussion These results demonstrate that sustained PFC hyperactivity modifies circuit characteristics and social behaviors in ways that can be modulated by selective OXTR activation and that this model may be used to understand the circuit recruitment of prosocial therapies in drug discovery.
Collapse
Affiliation(s)
- Philipp Janz
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Frederic Knoflach
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Konrad Bleicher
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Sara Belli
- Roche Pharma Research and Early Development, Pharmaceutical Science, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Barbara Biemans
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Patrick Schnider
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Martin Ebeling
- Roche Pharma Research and Early Development, Pharmaceutical Science, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Christophe Grundschober
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Madhurima Benekareddy
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
- Calico Life Sciences, South San Francisco, CA, United States
| |
Collapse
|
50
|
Li E, Kampmann M. Toward a CRISPR understanding of gene function in human brain development. Cell Stem Cell 2023; 30:1561-1562. [PMID: 38065064 DOI: 10.1016/j.stem.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023]
Abstract
Genome-wide association studies pinpoint genetic risk factors for neurodevelopmental disorders (NDDs), but the next challenge is to understand the mechanisms through which these genes affect brain development. Two recent CRISPR screens in human brain organoids1,2 interrogate the function of risk genes for autism spectrum disorder and other NDDs.
Collapse
Affiliation(s)
- Emmy Li
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|