1
|
Darwish M, Passarell J, Maxwell K, Bradley H, Bishop KM, Youakim JM. Population Pharmacokinetics of Trofinetide in a Pediatric Population Aged 2-4 Years with Rett Syndrome. Adv Ther 2025; 42:1009-1025. [PMID: 39692837 PMCID: PMC11787236 DOI: 10.1007/s12325-024-03058-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/25/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Weight-banded trofinetide dosing improved physician- and caregiver-rated efficacy measures and had acceptable tolerability in patients aged 2‒4 years (DAFFODIL study) and 5‒20 years (LAVENDER study) with Rett syndrome (RTT). Selection of weight-banded dosing regimens for these studies was based on population pharmacokinetic (popPK) modeling and exposure simulations. This study applied an updated popPK model to confirm steady-state trofinetide exposures achieved in DAFFODIL patients were within target range. METHODS A popPK model was developed using data from 14 clinical studies of trofinetide in healthy volunteers and pediatric and adult patients, including the LAVENDER and DAFFODIL studies. Individual exposure measures (area under concentration-time curve over 0-12 h [AUC0-12] were generated via integration of the predicted concentration-time profile for each DAFFODIL study participant based on the popPK model and individual empiric Bayesian pharmacokinetic parameter estimates. Distributions of steady-state AUC0-12 values for each body-weight group were compared against target exposure (AUC0-12 = 800‒1200 µg·h/mL). RESULTS Distribution and box plots of simulated steady-state AUC0-12 values achieved with the weight-banded DAFFODIL dosing regimen used in younger individuals aged 2-4 years with RTT (twice daily trofinetide 5 g [≥ 9 to < 12 kg] or 6 g [≥ 12 to < 20 kg]) indicated good overlap with the target exposure range. Median steady-state AUC0-12 values for both body-weight bands fell within the target exposure range. CONCLUSIONS PopPK model-based simulations confirm that the weight-banded dosing regimen used in DAFFODIL is adequate to achieve target trofinetide exposure in 2- to 4-year-olds with RTT.
Collapse
Affiliation(s)
- Mona Darwish
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA.
| | - Julie Passarell
- Clinical Pharmacology and Pharmacometrics Division, Simulations Plus Inc., Buffalo, NY, USA
| | - Kelly Maxwell
- Clinical Pharmacology and Pharmacometrics Division, Simulations Plus Inc., Buffalo, NY, USA
| | - Heather Bradley
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| | - Kathie M Bishop
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| | - James M Youakim
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| |
Collapse
|
2
|
Dominguez G, Wu Y, Zhou J. Epigenetic Regulation and Neurodevelopmental Disorders: From MeCP2 to the TCF20/PHF14 Complex. Genes (Basel) 2024; 15:1653. [PMID: 39766920 PMCID: PMC11728296 DOI: 10.3390/genes15121653] [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: 12/01/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) affect approximately 15% of children and adolescents worldwide. This group of disorders is often polygenic with varying risk factors, with many associated genes converging on shared molecular pathways, including chromatin regulation and transcriptional control. Understanding how NDD-associated chromatin regulators and protein complexes orchestrate these regulatory pathways is crucial for elucidating NDD pathogenesis and developing targeted therapeutic strategies. Recently, the TCF20/PHF14 chromatin complex was identified in the mammalian brain, expanding the list of chromatin regulatory remodelers implicated in NDDs. This complex-which includes MeCP2, RAI1, TCF20, PHF14, and HMG20A-plays a vital role in epigenetic and transcriptional regulation. METHODS We review and summarize current research and clinical reports pertaining to the different components of the MeCP2-interacting TCF20/PHF14 complex. We examine the NDDs associated with the TCF20/PHF14 complex, explore the molecular and neuronal functions of its components, and discuss emerging therapeutic strategies targeting this complex to mitigate symptoms, with broader applicability to other NDDs. RESULTS Mutations in the genes encoding the components of the MeCP2-interacting TCF20/PHF14 complex have been linked to various NDDs, underscoring its critical contribution to brain development and NDD pathogenesis. CONCLUSIONS The MeCP2-interacting TCF20/PHF14 complex and its associated NDDs could serve as a model system to provide insight into the interplay between epigenetic regulation and NDD pathogenesis.
Collapse
Affiliation(s)
- Gaea Dominguez
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
| | - Yongji Wu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
| | - Jian Zhou
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (G.D.); (Y.W.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
3
|
Esposito A, Seri T, Breccia M, Indrigo M, De Rocco G, Nuzzolillo F, Denti V, Pappacena F, Tartaglione G, Serrao S, Paglia G, Murru L, de Pretis S, Cioni JM, Landsberger N, Guarnieri FC, Palmieri M. Unraveling autophagic imbalances and therapeutic insights in Mecp2-deficient models. EMBO Mol Med 2024; 16:2795-2826. [PMID: 39402139 PMCID: PMC11555085 DOI: 10.1038/s44321-024-00151-w] [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: 12/22/2023] [Revised: 09/16/2024] [Accepted: 09/27/2024] [Indexed: 11/13/2024] Open
Abstract
Loss-of-function mutations in MECP2 are associated to Rett syndrome (RTT), a severe neurodevelopmental disease. Mainly working as a transcriptional regulator, MeCP2 absence leads to gene expression perturbations resulting in deficits of synaptic function and neuronal activity. In addition, RTT patients and mouse models suffer from a complex metabolic syndrome, suggesting that related cellular pathways might contribute to neuropathogenesis. Along this line, autophagy is critical in sustaining developing neuron homeostasis by breaking down dysfunctional proteins, lipids, and organelles.Here, we investigated the autophagic pathway in RTT and found reduced content of autophagic vacuoles in Mecp2 knock-out neurons. This correlates with defective lipidation of LC3B, probably caused by a deficiency of the autophagic membrane lipid phosphatidylethanolamine. The administration of the autophagy inducer trehalose recovers LC3B lipidation, autophagosomes content in knock-out neurons, and ameliorates their morphology, neuronal activity and synaptic ultrastructure. Moreover, we provide evidence for attenuation of motor and exploratory impairment in Mecp2 knock-out mice upon trehalose administration. Overall, our findings open new perspectives for neurodevelopmental disorders therapies based on the concept of autophagy modulation.
Collapse
Affiliation(s)
- Alessandro Esposito
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Tommaso Seri
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Breccia
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy
| | - Marzia Indrigo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppina De Rocco
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy
| | | | - Vanna Denti
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Francesca Pappacena
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gaia Tartaglione
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simone Serrao
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giuseppe Paglia
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Luca Murru
- CNR Institute of Neuroscience, Vedano al Lambro, Italy
| | - Stefano de Pretis
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jean-Michel Cioni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicoletta Landsberger
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, Italy
| | - Fabrizia Claudia Guarnieri
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- CNR Institute of Neuroscience, Vedano al Lambro, Italy.
| | - Michela Palmieri
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
4
|
Percy AK, Neul JL, Benke TA, Berry-Kravis EM, Glaze DG, Marsh ED, Barrett AM, An D, Bishop KM, Youakim JM. Trofinetide for the treatment of Rett syndrome: Long-term safety and efficacy results of the 32-month, open-label LILAC-2 study. MED 2024; 5:1275-1281.e2. [PMID: 39025065 DOI: 10.1016/j.medj.2024.06.007] [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: 03/15/2024] [Revised: 05/23/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Trofinetide was approved for the treatment of Rett syndrome (RTT) in patients aged ≥2 years based on the results of the 12-week, randomized, phase 3 LAVENDER study. In LILAC, a 40-week, open-label extension study of LAVENDER, trofinetide continued to improve the symptoms of RTT, with a similar safety profile as LAVENDER. Here, we report long-term safety and efficacy results of LILAC-2, a 32-month, open-label extension study. METHODS Females aged 5-22 years who completed LILAC were eligible to enter LILAC-2. Safety and tolerability were assessed with the incidence of adverse events (AEs). Efficacy was assessed with Rett Syndrome Behaviour Questionnaire (RSBQ) and Clinical Global Impression-Improvement (CGI-I) scores. Caregiver interviews explored the patient's experience with RTT and the efficacy of trofinetide during study participation. FINDINGS In total, 77 participants were enrolled in LILAC-2. The most common AEs were diarrhea (53.2%), COVID-19 (27.3%), and vomiting (19.5%). The mean (standard error [SE]) change in RSBQ score from LAVENDER baseline to week 104 of LILAC-2 was -11.8 (2.45). The mean (SE) CGI-I score from LILAC baseline to week 12 of LILAC-2 was 3.1 (0.10). Most caregivers (96%; n = 24/25) were satisfied or very satisfied with the benefits of trofinetide. CONCLUSIONS Long-term treatment with trofinetide continued to improve RTT symptoms, without new safety concerns. Caregivers reported satisfaction with trofinetide related to improvements that were meaningful for their child and themselves. FUNDING The study was supported by Acadia Pharmaceuticals (San Diego, CA, USA). This study was registered at ClinicalTrials.gov: NCT04776746.
Collapse
Affiliation(s)
- Alan K Percy
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeffrey L Neul
- Department of Pediatrics, Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy A Benke
- Department of Pediatrics, Children's Hospital of Colorado/University of Colorado School of Medicine, Aurora, CO, USA
| | - Elizabeth M Berry-Kravis
- Departments of Pediatrics, Neurological Sciences and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Daniel G Glaze
- Department of Pediatrics and Neurology, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, USA
| | - Eric D Marsh
- Division of Child Neurology, Children's Hospital of Philadelphia, Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy M Barrett
- Patient-Centered Outcomes Assessment, RTI Health Solutions, Research Triangle Park, NC, USA
| | - Di An
- Acadia Pharmaceuticals, Inc., San Diego, CA, USA
| | | | | |
Collapse
|
5
|
Bahram Sangani N, Koetsier J, Gomes AR, Diogo MM, Fernandes TG, Bouwman FG, Mariman ECM, Ghazvini M, Gribnau J, Curfs LMG, Reutelingsperger CP, Eijssen LMT. Involvement of extracellular vesicle microRNA clusters in developing healthy and Rett syndrome brain organoids. Cell Mol Life Sci 2024; 81:410. [PMID: 39305343 PMCID: PMC11416455 DOI: 10.1007/s00018-024-05409-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/19/2024] [Accepted: 08/10/2024] [Indexed: 09/25/2024]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by de novo mutations in the MECP2 gene. Although miRNAs in extracellular vesicles (EVs) have been suggested to play an essential role in several neurological conditions, no prior study has utilized brain organoids to profile EV-derived miRNAs during normal and RTT-affected neuronal development. Here we report the spatiotemporal expression pattern of EV-derived miRNAs in region-specific forebrain organoids generated from female hiPSCs with a MeCP2:R255X mutation and the corresponding isogenic control. EV miRNA and protein expression profiles were characterized at day 0, day 13, day 40, and day 75. Several members of the hsa-miR-302/367 cluster were identified as having a time-dependent expression profile with RTT-specific alterations at the latest developmental stage. Moreover, the miRNA species of the chromosome 14 miRNA cluster (C14MC) exhibited strong upregulation in RTT forebrain organoids irrespective of their spatiotemporal location. Together, our results suggest essential roles of the C14MC and hsa-miR-302/367 clusters in EVs during normal and RTT-associated neurodevelopment, displaying promising prospects as biomarkers for monitoring RTT progression.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Jarno Koetsier
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Ana Rita Gomes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Margarida Diogo
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Freek G Bouwman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Mehrnaz Ghazvini
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Joost Gribnau
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Developmental Biology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Leopold M G Curfs
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands.
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands.
| | - Lars M T Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- Department of Bioinformatics-BiGCaT, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
6
|
Percy AK, Neul JL, Benke TA, Berry-Kravis EM, Glaze DG, Marsh ED, An D, Bishop KM, Youakim JM. Trofinetide for the treatment of Rett syndrome: Results from the open-label extension LILAC study. MED 2024; 5:1178-1189.e3. [PMID: 38917793 DOI: 10.1016/j.medj.2024.05.018] [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: 10/05/2023] [Revised: 04/04/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Trofinetide was approved for the treatment of Rett syndrome based on the results of the phase 3, randomized, placebo-controlled, 12-week LAVENDER study. Rett syndrome is a chronic disorder requiring long-term treatment. We report the efficacy and safety results of LILAC, a 40-week, open-label extension study of LAVENDER. METHODS Females with Rett syndrome aged 5-21 years received open-label treatment with trofinetide for 40 weeks. The primary endpoint was long-term safety of trofinetide; secondary endpoints included the change from baseline at week 40 in the Rett Syndrome Behaviour Questionnaire score and the Clinical Global Impression-Improvement score at week 40. FINDINGS Overall, 154 participants were enrolled and treated with trofinetide in LILAC. The most common adverse events in LILAC were diarrhea (74.7%), vomiting (28.6%), and COVID-19 (11.0%). Diarrhea was the most common adverse event leading to treatment withdrawal (21.4%). The Rett Syndrome Behaviour Questionnaire mean score (standard error) improvement from the LAVENDER baseline to week 40 in LILAC was -7.3 (1.62) and -7.0 (1.61) for participants treated with trofinetide and placebo in LAVENDER, respectively. Mean Clinical Global Impression-Improvement scores (standard error) at week 40 rated from the LILAC baseline were 3.1 (0.11) and 3.2 (0.14) for participants treated with trofinetide and placebo in LAVENDER, respectively. CONCLUSIONS Treatment with trofinetide for ≤40 weeks continued to improve symptoms of Rett syndrome. Trofinetide had a similar safety profile in LILAC as in LAVENDER. FUNDING The study was supported by Acadia Pharmaceuticals Inc. (San Diego, CA, USA). This trial was registered at ClinicalTrials.gov (NCT04279314).
Collapse
Affiliation(s)
- Alan K Percy
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jeffrey L Neul
- Department of Pediatrics, Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Timothy A Benke
- Department of Pediatrics, Children's Hospital of Colorado/University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Elizabeth M Berry-Kravis
- Departments of Pediatrics, Neurological Sciences and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Daniel G Glaze
- Department of Pediatrics and Neurology, Texas Children's Hospital/Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric D Marsh
- Division of Child Neurology, Children's Hospital of Philadelphia, Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Di An
- Acadia Pharmaceuticals Inc., San Diego, CA 92130, USA
| | | | | |
Collapse
|
7
|
Dennys CN, Vermudez SAD, Deacon RJM, Sierra-Delgado JA, Rich K, Zhang X, Buch A, Weiss K, Moxley Y, Rajpal H, Espinoza FD, Powers S, Ávila AS, Gogliotti RG, Cogram P, Niswender CM, Meyer KC. MeCP2 gene therapy ameliorates disease phenotype in mouse model for Pitt Hopkins syndrome. Neurotherapeutics 2024; 21:e00376. [PMID: 38876822 PMCID: PMC11579869 DOI: 10.1016/j.neurot.2024.e00376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 06/16/2024] Open
Abstract
The neurodevelopmental disorder Pitt Hopkins syndrome (PTHS) causes clinical symptoms similar to Rett syndrome (RTT) patients. However, RTT is caused by MECP2 mutations whereas mutations in the TCF4 gene lead to PTHS. The mechanistic commonalities underling these two disorders are unknown, but their shared symptomology suggest that convergent pathway-level disruption likely exists. We reprogrammed patient skin derived fibroblasts into induced neuronal progenitor cells. Interestingly, we discovered that MeCP2 levels were decreased in PTHS patient iNPCs relative to healthy controls and that both iNPCs and iAstrocytes displayed defects in function and differentiation in a mutation-specific manner. When Tcf4+/- mice were genetically crossed with mice overexpressing MeCP2, molecular and phenotypic defects were significantly ameliorated, underlining and important role of MeCP2 in PTHS pathology. Importantly, post-natal intracerebroventricular gene replacement therapy with adeno-associated viral vector serotype 9 (AAV9)-expressing MeCP2 (AAV9.P546.MeCP2) significantly improved iNPC and iAstrocyte function and effectively ameliorated histological and behavioral defects in Tcf4+/- mice. Combined, our data suggest a previously unknown role of MeCP2 in PTHS pathology and common pathways that might be affected in multiple neurodevelopmental disorders. Our work highlights potential novel therapeutic targets for PTHS, including upregulation of MeCP2 expression or its downstream targets or, potentially, MeCP2-based gene therapy.
Collapse
Affiliation(s)
- Cassandra N Dennys
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Sheryl Anne D Vermudez
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Robert J M Deacon
- Department of Genetics, Institute of Ecology and Biodiversity, Faculty of Science, University of Chile, Chile
| | - J Andrea Sierra-Delgado
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kelly Rich
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Xiaojin Zhang
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Aditi Buch
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Kelly Weiss
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Yuta Moxley
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Hemangi Rajpal
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Francisca D Espinoza
- Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, 4090541, Chile
| | - Samantha Powers
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ariel S Ávila
- Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, 4090541, Chile
| | - Rocco G Gogliotti
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago, Maywood, IL 60153, USA
| | - Patricia Cogram
- Department of Genetics, Institute of Ecology and Biodiversity, Faculty of Science, University of Chile, Chile
| | - Colleen M Niswender
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kathrin C Meyer
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University Medical Center, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
8
|
Darwish M, Obianom ON, Youakim JM, Darling I, Lukacova V, Bradley H. Effect of Hepatic Impairment on Trofinetide Exposures Using an In Silico Physiologically Based Pharmacokinetic Model. Adv Ther 2024; 41:3328-3341. [PMID: 38963587 PMCID: PMC11263405 DOI: 10.1007/s12325-024-02926-6] [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: 04/12/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Trofinetide is the first drug to be approved for the treatment of Rett syndrome. Hepatic impairment is not expected to affect the pharmacokinetic (PK) profile of trofinetide because of predominant renal excretion. This study was conducted to help understand the potential impact of any hepatic impairment on trofinetide PK. METHODS This study used physiologically based PK modeling to estimate trofinetide exposure (maximum drug concentration and area under the concentration-time curve from time zero to infinity) in virtual patients with mild, moderate, and severe hepatic impairment (per Child-Pugh classification) compared with virtual healthy subjects following a 12 g oral trofinetide dose. RESULTS In individual deterministic simulations for matched individuals and stochastic simulations at the population level (100 virtual individuals simulated per population), as anticipated, predicted plasma exposures were similar for healthy subjects and for patients with mild, moderate, and severe hepatic impairment. However, predicted blood concentration exposures slightly increased with increasing severity of hepatic impairment because of change in hematocrit levels. CONCLUSION This study indicates that hepatic impairment is not expected to have a clinically relevant effect on exposure to trofinetide.
Collapse
Affiliation(s)
- Mona Darwish
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA.
| | | | - James M Youakim
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| | | | | | - Heather Bradley
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| |
Collapse
|
9
|
Parent HH, Niswender CM. Therapeutic Potential for Metabotropic Glutamate Receptor 7 Modulators in Cognitive Disorders. Mol Pharmacol 2024; 105:348-358. [PMID: 38423750 PMCID: PMC11026152 DOI: 10.1124/molpharm.124.000874] [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: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is the most highly conserved and abundantly expressed mGlu receptor in the human brain. The presynaptic localization of mGlu7, coupled with its low affinity for its endogenous agonist, glutamate, are features that contribute to the receptor's role in modulating neuronal excitation and inhibition patterns, including long-term potentiation, in various brain regions. These characteristics suggest that mGlu7 modulation may serve as a novel therapeutic strategy in disorders of cognitive dysfunction, including neurodevelopmental disorders that cause impairments in learning, memory, and attention. Primary mutations in the GRM7 gene have recently been identified as novel causes of neurodevelopmental disorders, and these patients exhibit profound intellectual and cognitive disability. Pharmacological tools, such as agonists, antagonists, and allosteric modulators, have been the mainstay for targeting mGlu7 in its endogenous homodimeric form to probe effects of its function and modulation in disease models. However, recent research has identified diversity in dimerization, as well as trans-synaptic interacting proteins, that also play a role in mGlu7 signaling and pharmacological properties. These novel findings represent exciting opportunities in the field of mGlu receptor drug discovery and highlight the importance of further understanding the functions of mGlu7 in complex neurologic conditions at both the molecular and physiologic levels. SIGNIFICANCE STATEMENT: Proper expression and function of mGlu7 is essential for learning, attention, and memory formation at the molecular level within neural circuits. The pharmacological targeting of mGlu7 is undergoing a paradigm shift by incorporating an understanding of receptor interaction with other cis- and trans- acting synaptic proteins, as well as various intracellular signaling pathways. Based upon these new findings, mGlu7's potential as a drug target in the treatment of cognitive disorders and learning impairments is primed for exploration.
Collapse
Affiliation(s)
- Harrison H Parent
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Colleen M Niswender
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| |
Collapse
|
10
|
Darwish M, Passarell J, Youakim JM, Bradley H, Bishop KM. Exposure-Response Efficacy Modeling to Support Trofinetide Dosing in Individuals with Rett Syndrome. Adv Ther 2024; 41:1462-1480. [PMID: 38363467 PMCID: PMC10960884 DOI: 10.1007/s12325-024-02796-y] [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: 09/26/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024]
Abstract
INTRODUCTION Trofinetide was recently approved for the treatment of Rett syndrome (RTT) on the basis of the efficacy and safety findings of the phase 3 LAVENDER study, which used a body weight-based dosing regimen. Exposure-response (E-R) efficacy modeling was used to characterize relationships between trofinetide exposure measures (maximum drug concentration and area under the concentration-time curve for the dosing interval of 0-12 h [AUC0-12]) and efficacy endpoints in RTT clinical studies to support the trofinetide dosing regimen. METHODS Efficacy endpoints were modeled using trofinetide exposure measures predicted from the population pharmacokinetic model and Bayesian estimates. The analysis population for each E-R model comprised individuals receiving placebo or trofinetide who had available trofinetide exposure measures. Efficacy endpoints were scores from the Rett Syndrome Behaviour Questionnaire (RSBQ), the Clinical Global Impression-Improvement, the Communication and Symbolic Behavior Scales Developmental Profile™ Infant-Toddler Checklist (CSBS-DP-IT) Social Composite, and the Rett Syndrome Clinician Rating of Ability to Communicate Choices (RTT-COMC). RESULTS Higher trofinetide exposure was associated with improvements in RSBQ, CSBS-DP-IT Social Composite, and RTT-COMC scores. Assuming target trofinetide AUC0-12 values of 800-1200 μg·h/mL, the reductions in RSBQ total scores at week 12 were approximately five- to seven-fold greater with trofinetide (range 3.55-4.94) versus placebo (0.76). Significant E-R relationships were also found for the CSBS-DP-IT Social Composite and RTT-COMC scores. CONCLUSION E-R efficacy modeling demonstrated significant relationships between trofinetide exposure and RSBQ, CSBS-DP-IT Social Composite, and RTT-COMC scores. Trofinetide is efficacious within the target exposure range, supporting the approved dosing regimen for trofinetide. TRIAL REGISTRATION NCT01703533, NCT02715115, NCT04181723.
Collapse
Affiliation(s)
- Mona Darwish
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA.
| | - Julie Passarell
- Cognigen Corporation (a Simulations Plus Company), Buffalo, NY, USA
| | - James M Youakim
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| | - Heather Bradley
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| | - Kathie M Bishop
- Acadia Pharmaceuticals Inc., 12830 El Camino Real, Suite 400, San Diego, CA, 92130, USA
| |
Collapse
|
11
|
Kennedy M, Glass L, Glaze DG, Kaminsky S, Percy AK, Neul JL, Jones NE, Tropea D, Horrigan JP, Nues P, Bishop KM, Youakim JM. Development of trofinetide for the treatment of Rett syndrome: from bench to bedside. Front Pharmacol 2024; 14:1341746. [PMID: 38318312 PMCID: PMC10839050 DOI: 10.3389/fphar.2023.1341746] [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: 11/20/2023] [Accepted: 12/18/2023] [Indexed: 02/07/2024] Open
Abstract
Rett syndrome (RTT) is rare neurodevelopmental disorder caused by mutations in the MECP2 gene that encodes methyl-CpG-binding protein 2 (MeCP2), a DNA-binding protein with roles in epigenetic regulation of gene expression. Functional loss of MeCP2 results in abnormal neuronal maturation and plasticity, characterized by loss of verbal communication and loss of fine and gross motor function, among others. Trofinetide, a synthetic analog of glycine-proline-glutamate, was approved by the US Food and Drug Administration for the treatment of RTT in adult and pediatric patients aged 2 years and older. Here, we present the development of trofinetide from bench research to clinical studies and emphasize how the collaboration between academia, the pharmaceutical industry, and patient advocacy led to the recent approval. The bench-to-bedside development of trofinetide underscores the value of collaboration between these groups in the development and approval of treatments for rare diseases.
Collapse
Affiliation(s)
- Melissa Kennedy
- International Rett Syndrome Foundation, Cincinnati, OH, United States
| | - Larry Glass
- Neuren Pharmaceuticals Ltd., Melbourne, VIC, Australia
| | - Daniel G. Glaze
- Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, United States
| | - Steve Kaminsky
- International Rett Syndrome Foundation, Cincinnati, OH, United States
| | - Alan K. Percy
- Division of Pediatric Neurology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jeffrey L. Neul
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | - Daniela Tropea
- Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Joseph P. Horrigan
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, United States
| | - Paige Nues
- International Rett Syndrome Foundation, Cincinnati, OH, United States
| | | | | |
Collapse
|
12
|
Oluigbo DC. Rett Syndrome: A Tale of Altered Genetics, Synaptic Plasticity, and Neurodevelopmental Dynamics. Cureus 2023; 15:e41555. [PMID: 37554594 PMCID: PMC10405636 DOI: 10.7759/cureus.41555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is a leading cause of severe cognitive and physical impairment. RTT typically occurs in females, although rare cases of males with the disease exist. Its genetic cause, symptoms, and clinical progression timeline have also become well-documented since its initial discovery. However, a relatively late diagnosis and lack of an available cure signify that our understanding of the disease is incomplete. Innovative research methods and tools are thereby helping to fill gaps in our knowledge of RTT. Specifically, mouse models of RTT, video analysis, and retrospective parental analysis are well-established tools that provide valuable insights into RTT. Moreover, current and anticipated treatment options are improving the quality of life of the RTT patient population. Collectively, these developments are creating optimistic future perspectives for RTT.
Collapse
Affiliation(s)
- David C Oluigbo
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, USA
| |
Collapse
|
13
|
Neul JL, Percy AK, Benke TA, Berry-Kravis EM, Glaze DG, Marsh ED, Lin T, Stankovic S, Bishop KM, Youakim JM. Trofinetide for the treatment of Rett syndrome: a randomized phase 3 study. Nat Med 2023; 29:1468-1475. [PMID: 37291210 PMCID: PMC10287558 DOI: 10.1038/s41591-023-02398-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/12/2023] [Indexed: 06/10/2023]
Abstract
Rett syndrome is a rare, genetic neurodevelopmental disorder. Trofinetide is a synthetic analog of glycine-proline-glutamate, the N-terminal tripeptide of the insulin-like growth factor 1 protein, and has demonstrated clinical benefit in phase 2 studies in Rett syndrome. In this phase 3 study ( https://clinicaltrials.gov identifier NCT04181723 ), females with Rett syndrome received twice-daily oral trofinetide (n = 93) or placebo (n = 94) for 12 weeks. For the coprimary efficacy endpoints, least squares mean (LSM) change from baseline to week 12 in the Rett Syndrome Behaviour Questionnaire for trofinetide versus placebo was -4.9 versus -1.7 (P = 0.0175; Cohen's d effect size, 0.37), and LSM Clinical Global Impression-Improvement at week 12 was 3.5 versus 3.8 (P = 0.0030; effect size, 0.47). For the key secondary efficacy endpoint, LSM change from baseline to week 12 in the Communication and Symbolic Behavior Scales Developmental Profile Infant-Toddler Checklist Social Composite score was -0.1 versus -1.1 (P = 0.0064; effect size, 0.43). Common treatment-emergent adverse events included diarrhea (80.6% for trofinetide versus 19.1% for placebo), which was mostly mild to moderate in severity. Significant improvement for trofinetide compared with placebo was observed for the coprimary efficacy endpoints, suggesting that trofinetide provides benefit in treating the core symptoms of Rett syndrome.
Collapse
Affiliation(s)
- Jeffrey L Neul
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alan K Percy
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Timothy A Benke
- Children's Hospital of Colorado and University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Daniel G Glaze
- Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA
| | - Eric D Marsh
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tim Lin
- Acadia Pharmaceuticals Inc., San Diego, CA, USA
| | | | | | | |
Collapse
|
14
|
Palmieri M, Pozzer D, Landsberger N. Advanced genetic therapies for the treatment of Rett syndrome: state of the art and future perspectives. Front Neurosci 2023; 17:1172805. [PMID: 37304036 PMCID: PMC10248472 DOI: 10.3389/fnins.2023.1172805] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/02/2023] [Indexed: 06/13/2023] Open
Abstract
Loss and gain of functions mutations in the X-linked MECP2 (methyl-CpG-binding protein 2) gene are responsible for a set of generally severe neurological disorders that can affect both genders. In particular, Mecp2 deficiency is mainly associated with Rett syndrome (RTT) in girls, while duplication of the MECP2 gene leads, mainly in boys, to the MECP2 duplication syndrome (MDS). No cure is currently available for MECP2 related disorders. However, several studies have reported that by re-expressing the wild-type gene is possible to restore defective phenotypes of Mecp2 null animals. This proof of principle endorsed many laboratories to search for novel therapeutic strategies to cure RTT. Besides pharmacological approaches aimed at modulating MeCP2-downstream pathways, genetic targeting of MECP2 or its transcript have been largely proposed. Remarkably, two studies focused on augmentative gene therapy were recently approved for clinical trials. Both use molecular strategies to well-control gene dosage. Notably, the recent development of genome editing technologies has opened an alternative way to specifically target MECP2 without altering its physiological levels. Other attractive approaches exclusively applicable for nonsense mutations are the translational read-through (TR) and t-RNA suppressor therapy. Reactivation of the MECP2 locus on the silent X chromosome represents another valid choice for the disease. In this article, we intend to review the most recent genetic interventions for the treatment of RTT, describing the current state of the art, and the related advantages and concerns. We will also discuss the possible application of other advanced therapies, based on molecular delivery through nanoparticles, already proposed for other neurological disorders but still not tested in RTT.
Collapse
Affiliation(s)
- Michela Palmieri
- Rett Research Unit, Division of Neuroscience, San Raffaele Hospital (IRCCS), Milan, Italy
| | - Diego Pozzer
- Rett Research Unit, Division of Neuroscience, San Raffaele Hospital (IRCCS), Milan, Italy
| | - Nicoletta Landsberger
- Rett Research Unit, Division of Neuroscience, San Raffaele Hospital (IRCCS), Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| |
Collapse
|
15
|
Freitas GA, Niswender CM. GRM7 gene mutations and consequences for neurodevelopment. Pharmacol Biochem Behav 2023; 225:173546. [PMID: 37003303 PMCID: PMC10192299 DOI: 10.1016/j.pbb.2023.173546] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
The metabotropic glutamate receptor 7 (mGlu7), encoded by the GRM7 gene in humans, is a presynaptic, G protein-coupled glutamate receptor that is essential for modulating neurotransmission. Mutations in or reduced expression of GRM7 have been identified in different genetic neurodevelopmental disorders (NDDs), and rare biallelic missense variants have been proposed to underlie a subset of NDDs. Clinical GRM7 variants have been associated with a range of symptoms consistent with neurodevelopmental molecular features, including hypomyelination, brain atrophy and defects in axon outgrowth. Here, we review the newest findings regarding the cellular and molecular defects caused by GRM7 variants in NDD patients.
Collapse
Affiliation(s)
- Geanne A Freitas
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37212, United States of America
| | - Colleen M Niswender
- Department of Pharmacology and Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37212, United States of America; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America.
| |
Collapse
|
16
|
Carli S, Chaabane L, De Rocco G, Albizzati E, Sormonta I, Calligaro S, Bonizzi P, Frasca A, Landsberger N. A comprehensive longitudinal study of magnetic resonance imaging identifies novel features of the Mecp2 deficient mouse brain. Neurobiol Dis 2023; 180:106083. [PMID: 36931532 DOI: 10.1016/j.nbd.2023.106083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/17/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Rett syndrome (RTT) is a X-linked neurodevelopmental disorder which represents the leading cause of severe incurable intellectual disability in females worldwide. The vast majority of RTT cases are caused by mutations in the X-linked MECP2 gene, and preclinical studies on RTT largely benefit from the use of mouse models of Mecp2, which present a broad spectrum of symptoms phenocopying those manifested by RTT patients. Neurons represent the core targets of the pathology; however, neuroanatomical abnormalities that regionally characterize the Mecp2 deficient mammalian brain remain ill-defined. Neuroimaging techniques, such as MRI and MRS, represent a key approach for assessing in vivo anatomic and metabolic changes in brain. Being non-invasive, these analyses also permit to investigate how the disease progresses over time through longitudinal studies. To foster the biological comprehension of RTT and identify useful biomarkers, we have performed a thorough in vivo longitudinal study of MRI and MRS in Mecp2 deficient mouse brains. Analyses were performed on both genders of two different mouse models of RTT, using an automatic atlas-based segmentation tool that permitted to obtain a detailed and unbiased description of the whole RTT mouse brain. We found that the most robust alteration of the RTT brain consists in an overall reduction of the brain volume. Accordingly, Mecp2 deficiency generally delays brain growth, eventually leading, in heterozygous older animals, to stagnation and/or contraction. Most but not all brain regions participate in the observed deficiency in brain size; similarly, the volumetric defect progresses diversely in different brain areas also depending on the specific Mecp2 genetic lesion and gender. Interestingly, in some regions volumetric defects anticipate overt symptoms, possibly revealing where the pathology originates and providing a useful biomarker for assessing drug efficacy in pre-clinical studies.
Collapse
Affiliation(s)
- Sara Carli
- Neuroscience Division, IRCCS San Raffaele Scientific Institute, Milan I-20132, Italy.
| | - Linda Chaabane
- Institute of Experimental Neurology (INSPE) and Experimental Imaging Center (CIS), IRCCS San Raffaele Scientific Institute, Milan I-20132, Italy.
| | - Giuseppina De Rocco
- Neuroscience Division, IRCCS San Raffaele Scientific Institute, Milan I-20132, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan) I-20090, Italy.
| | - Elena Albizzati
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan) I-20090, Italy.
| | - Irene Sormonta
- Neuroscience Division, IRCCS San Raffaele Scientific Institute, Milan I-20132, Italy.
| | - Stefano Calligaro
- Neuroscience Division, IRCCS San Raffaele Scientific Institute, Milan I-20132, Italy.
| | - Pietro Bonizzi
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, the Netherlands.
| | - Angelisa Frasca
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan) I-20090, Italy.
| | - Nicoletta Landsberger
- Neuroscience Division, IRCCS San Raffaele Scientific Institute, Milan I-20132, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan) I-20090, Italy.
| |
Collapse
|
17
|
Panayotis N, Ehinger Y, Felix MS, Roux JC. State-of-the-art therapies for Rett syndrome. Dev Med Child Neurol 2023; 65:162-170. [PMID: 36056801 PMCID: PMC10087176 DOI: 10.1111/dmcn.15383] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 01/04/2023]
Abstract
Rett syndrome (RTT) is an X-linked neurogenetic disorder caused by mutations of the MECP2 (methyl-CpG-binding protein 2) gene. Over two decades of work established MeCP2 as a protein with pivotal roles in the regulation of the epigenome, neuronal physiology, synaptic maintenance, and behaviour. Given the genetic aetiology of RTT and the proof of concept of its reversal in a mouse model, considerable efforts have been made to design therapeutic approaches to re-express MeCP2. By being at the forefront of the development of innovative gene therapies, research on RTT is of paramount importance for the treatment of monogenic neurological diseases. Here we discuss the recent advances and challenges of promising genetic strategies for the treatment of RTT including gene replacement therapies, gene/RNA editing strategies, and reactivation of the silenced X chromosome. WHAT THIS PAPER ADDS: Recent advances shed light on the promises of gene replacement therapy with new vectors designed to control the levels of MeCP2 expression. New developments in DNA/RNA editing approaches or reactivation of the silenced X chromosome open the possibility to re-express the native MeCP2 locus at endogenous levels. Current strategies still face limitations in transduction efficiency and future work is needed to improve brain delivery.
Collapse
Affiliation(s)
- Nicolas Panayotis
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.,Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.,Université Paris Cité, CNRS, Saints-Pères Paris Institute for the Neurosciences, Paris, France
| | - Yann Ehinger
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
18
|
Haase F, Singh R, Gloss B, Tam P, Gold W. Meta-Analysis Identifies BDNF and Novel Common Genes Differently Altered in Cross-Species Models of Rett Syndrome. Int J Mol Sci 2022; 23:11125. [PMID: 36232428 PMCID: PMC9570315 DOI: 10.3390/ijms231911125] [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: 06/28/2022] [Revised: 09/06/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Rett syndrome (RTT) is a rare disorder and one of the most abundant causes of intellectual disabilities in females. Single mutations in the gene coding for methyl-CpG-binding protein 2 (MeCP2) are responsible for the disorder. MeCP2 regulates gene expression as a transcriptional regulator as well as through epigenetic imprinting and chromatin condensation. Consequently, numerous biological pathways on multiple levels are influenced. However, the exact molecular pathways from genotype to phenotype are currently not fully elucidated. Treatment of RTT is purely symptomatic as no curative options for RTT have yet to reach the clinic. The paucity of this is mainly due to an incomplete understanding of the underlying pathophysiology of the disorder with no clinically useful common disease drivers, biomarkers, or therapeutic targets being identified. With the premise of identifying universal and robust disease drivers and therapeutic targets, here, we interrogated a range of RTT transcriptomic studies spanning different species, models, and MECP2 mutations. A meta-analysis using RNA sequencing data from brains of RTT mouse models, human post-mortem brain tissue, and patient-derived induced pluripotent stem cell (iPSC) neurons was performed using weighted gene correlation network analysis (WGCNA). This study identified a module of genes common to all datasets with the following ten hub genes driving the expression: ATRX, ADCY7, ADCY9, SOD1, CACNA1A, PLCG1, CCT5, RPS9, BDNF, and MECP2. Here, we discuss the potential benefits of these genes as therapeutic targets.
Collapse
Affiliation(s)
- Florencia Haase
- School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| | - Rachna Singh
- School of Medicine Sydney, The University of Notre Dame, Chippendale, NSW 2007, Australia
| | - Brian Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
| | - Patrick Tam
- School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Embryology Research Unit, Children’s Medical Research Institute, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Wendy Gold
- School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Kids Neuroscience Centre, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
- Molecular Neurobiology Research Laboratory, Kids Research, Children’s Hospital at Westmead, Westmead, NSW 2145, Australia
| |
Collapse
|
19
|
Yildirim M, Delepine C, Feldman D, Pham VA, Chou S, Ip J, Nott A, Tsai LH, Ming GL, So PTC, Sur M. Label-free three-photon imaging of intact human cerebral organoids for tracking early events in brain development and deficits in Rett syndrome. eLife 2022; 11:78079. [PMID: 35904330 PMCID: PMC9337854 DOI: 10.7554/elife.78079] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/08/2022] [Indexed: 12/20/2022] Open
Abstract
Human cerebral organoids are unique in their development of progenitor-rich zones akin to ventricular zones from which neuronal progenitors differentiate and migrate radially. Analyses of cerebral organoids thus far have been performed in sectioned tissue or in superficial layers due to their high scattering properties. Here, we demonstrate label-free three-photon imaging of whole, uncleared intact organoids (~2 mm depth) to assess early events of early human brain development. Optimizing a custom-made three-photon microscope to image intact cerebral organoids generated from Rett Syndrome patients, we show defects in the ventricular zone volumetric structure of mutant organoids compared to isogenic control organoids. Long-term imaging live organoids reveals that shorter migration distances and slower migration speeds of mutant radially migrating neurons are associated with more tortuous trajectories. Our label-free imaging system constitutes a particularly useful platform for tracking normal and abnormal development in individual organoids, as well as for screening therapeutic molecules via intact organoid imaging.
Collapse
Affiliation(s)
- Murat Yildirim
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Neuroscience, Cleveland Clinic Lerner Research Institute, Cleveland, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Chloe Delepine
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Danielle Feldman
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Vincent A Pham
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Stephanie Chou
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Jacque Ip
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alexi Nott
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Li-Huei Tsai
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Peter T C So
- Deparment of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Mriganka Sur
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
20
|
Zhang WJ, Shi LL, Zhang L. Dysregulated cortical synaptic plasticity under methyl-CpG binding protein 2 deficiency and its implication in motor impairments. World J Psychiatry 2022; 12:673-682. [PMID: 35663301 PMCID: PMC9150038 DOI: 10.5498/wjp.v12.i5.673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/16/2021] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
Caused by the mutation of methyl-CpG binding protein 2 (MeCP2), Rett syndrome leads to a battery of severe neural dysfunctions including the regression of motor coordination and motor learning. Current understanding has revealed the motor cortex as the critical region mediating voluntary movement. In this review article, we will summarize major findings from human patients and animal models regarding the cortical synaptic plasticity under the regulation of MeCP2. We will also discuss how mutation of MeCP2 leads to the disruption of cortical circuitry homeostasis to cause motor deficits. Lastly, potential values of physical exercise and neuromodulation approaches to recover neural plasticity and motor function will be evaluated. All of this evidence may help to accelerate timely diagnosis and effective interventions for Rett syndrome patients.
Collapse
Affiliation(s)
- Wei-Jia Zhang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Ling-Ling Shi
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Li Zhang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
21
|
Exploration of group II metabotropic glutamate receptor modulation in mouse models of Rett syndrome and MECP2 Duplication syndrome. Neuropharmacology 2022; 209:109022. [PMID: 35248529 PMCID: PMC8973998 DOI: 10.1016/j.neuropharm.2022.109022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/11/2022] [Accepted: 02/27/2022] [Indexed: 01/01/2023]
Abstract
Rett syndrome (RTT) and MECP2 Duplication syndrome (MDS) have opposing molecular origins in relation to expression and function of the transcriptional regulator Methyl-CpG-binding protein 2 (MeCP2). Several clinical and preclinical phenotypes, however, are shared between these disorders. Modulation of MeCP2 levels has recently emerged as a potential treatment option for both of these diseases. However, toxicity concerns remain with these approaches. Here, we focus on pharmacologically modulating the group II metabotropic glutamate receptors (mGlu), mGlu2 and mGlu3, which are two downstream targets of MeCP2 that are bidirectionally affected in expression in RTT patients and mice (Mecp2Null/+) versus an MDS mouse model (MECP2Tg1/o). Mecp2Null/+ and MECP2Tg1/o animals also exhibit contrasting phenotypes in trace fear acquisition, a form of temporal associative learning and memory, with trace fear deficiency observed in Mecp2Null/+ mice and abnormally enhanced trace fear acquisition in MECP2Tg1/o animals. In Mecp2Null/+ mice, treatment with the mGlu2/3 agonist LY379268 reverses the deficit in trace fear acquisition, and mGlu2/3 antagonism with LY341495 normalizes the abnormal trace fear learning and memory phenotype in MECP2Tg1/o mice. Altogether, these data highlight the role of group II mGlu receptors in RTT and MDS and demonstrate that both mGlu2 and mGlu3 may be potential therapeutic targets for these disorders.
Collapse
|
22
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
23
|
Dogra S, Stansley BJ, Xiang Z, Qian W, Gogliotti RG, Nicoletti F, Lindsley CW, Niswender CM, Joffe ME, Conn PJ. Activating mGlu 3 Metabotropic Glutamate Receptors Rescues Schizophrenia-like Cognitive Deficits Through Metaplastic Adaptations Within the Hippocampus. Biol Psychiatry 2021; 90:385-398. [PMID: 33965197 PMCID: PMC8403106 DOI: 10.1016/j.biopsych.2021.02.970] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Polymorphisms in GRM3, the gene encoding the mGlu3 metabotropic glutamate receptor, are associated with impaired cognition and neuropsychiatric disorders such as schizophrenia. Limited availability of selective genetic and molecular tools has hindered progress in developing a clear understanding of the mechanisms through which mGlu3 receptors regulate synaptic plasticity and cognition. METHODS We examined associative learning in mice with trace fear conditioning, a hippocampal-dependent learning task disrupted in patients with schizophrenia. Underlying cellular mechanisms were assessed using ex vivo hippocampal slice preparations with selective pharmacological tools and selective genetic deletion of mGlu3 receptor expression in specific neuronal subpopulations. RESULTS mGlu3 receptor activation enhanced trace fear conditioning and reversed deficits induced by subchronic phencyclidine. Mechanistic studies revealed that mGlu3 receptor activation induced metaplastic changes, biasing afferent stimulation to induce long-term potentiation through an mGlu5 receptor-dependent, endocannabinoid-mediated, disinhibitory mechanism. Selective genetic deletion of either mGlu3 or mGlu5 from hippocampal pyramidal cells eliminated effects of mGlu3 activation, revealing a novel mechanism by which mGlu3 and mGlu5 interact to enhance cognitive function. CONCLUSIONS These data demonstrate that activation of mGlu3 receptors in hippocampal pyramidal cells enhances hippocampal-dependent cognition in control and impaired mice by inducing a novel form of metaplasticity to regulate circuit function, providing a clear mechanism through which genetic variation in GRM3 can contribute to cognitive deficits. Developing approaches to positively modulate mGlu3 receptor function represents an encouraging new avenue for treating cognitive disruption in schizophrenia and other psychiatric diseases.
Collapse
Affiliation(s)
- Shalini Dogra
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Branden J. Stansley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Weilun Qian
- Vanderbilt University, Nashville, TN 37232, USA
| | - Rocco G. Gogliotti
- Molecular Pharmacology and Neuroscience Department, Loyola University Chicago, Maywood, IL, 60153, USA
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy,Department of Physiology and Pharmacology, University Sapienza of Roma, Roma, Italy
| | - Craig W. Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Colleen M. Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA,Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Max E. Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA,Correspondence to: Max E. Joffe, Ph.D., Research Instructor, Department of Pharmacology, Vanderbilt University, 12475E MRB4, Nashville, TN 37232-0697, Tel. (615) 322-6730, Fax. (615) 343-3088, , Twitter: @mejoffe; P. Jeffrey Conn, Ph.D., Lee E. Limbird Professor of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, 1205 Light Hall, Nashville, TN 37232-0697, Tel. (615) 936-2478, Fax. (615) 343-3088,
| | - P. Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA,Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA,Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, 37232, USA,Vanderbilt Kennedy Center, Vanderbilt University, Nashville, TN 37232, USA,Correspondence to: Max E. Joffe, Ph.D., Research Instructor, Department of Pharmacology, Vanderbilt University, 12475E MRB4, Nashville, TN 37232-0697, Tel. (615) 322-6730, Fax. (615) 343-3088, , Twitter: @mejoffe; P. Jeffrey Conn, Ph.D., Lee E. Limbird Professor of Pharmacology, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, 1205 Light Hall, Nashville, TN 37232-0697, Tel. (615) 936-2478, Fax. (615) 343-3088,
| |
Collapse
|
24
|
Marballi K, MacDonald JL. Proteomic and transcriptional changes associated with MeCP2 dysfunction reveal nodes for therapeutic intervention in Rett syndrome. Neurochem Int 2021; 148:105076. [PMID: 34048843 PMCID: PMC8286335 DOI: 10.1016/j.neuint.2021.105076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 04/13/2021] [Accepted: 05/17/2021] [Indexed: 12/28/2022]
Abstract
Mutations in the methyl-CpG binding protein 2 (MECP2) gene cause Rett syndrome (RTT), an X-linked neurodevelopmental disorder predominantly impacting females. MECP2 is an epigenetic transcriptional regulator acting mainly to repress gene expression, though it plays multiple gene regulatory roles and has distinct molecular targets across different cell types and specific developmental stages. In this review, we summarize MECP2 loss-of-function associated transcriptome and proteome disruptions, delving deeper into the latter which have been comparatively severely understudied. These disruptions converge on multiple biochemical and cellular pathways, including those involved in synaptic function and neurodevelopment, NF-κB signaling and inflammation, and the vitamin D pathway. RTT is a complex neurological disorder characterized by myriad physiological disruptions, in both the central nervous system and peripheral systems. Thus, treating RTT will likely require a combinatorial approach, targeting multiple nodes within the interactomes of these cellular pathways. To this end, we discuss the use of dietary supplements and factors, namely, vitamin D and polyunsaturated fatty acids (PUFAs), as possible partial therapeutic agents given their demonstrated benefit in RTT and their ability to restore homeostasis to multiple disrupted cellular pathways simultaneously. Further unravelling the complex molecular alterations induced by MECP2 loss-of-function, and contextualizing them at the level of proteome homeostasis, will identify new therapeutic avenues for this complex disorder.
Collapse
Affiliation(s)
- Ketan Marballi
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, USA
| | - Jessica L MacDonald
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, USA.
| |
Collapse
|
25
|
Sharifi O, Yasui DH. The Molecular Functions of MeCP2 in Rett Syndrome Pathology. Front Genet 2021; 12:624290. [PMID: 33968128 PMCID: PMC8102816 DOI: 10.3389/fgene.2021.624290] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
MeCP2 protein, encoded by the MECP2 gene, binds to DNA and affects transcription. Outside of this activity the true range of MeCP2 function is still not entirely clear. As MECP2 gene mutations cause the neurodevelopmental disorder Rett syndrome in 1 in 10,000 female births, much of what is known about the biologic function of MeCP2 comes from studying human cell culture models and rodent models with Mecp2 gene mutations. In this review, the full scope of MeCP2 research available in the NIH Pubmed (https://pubmed.ncbi.nlm.nih.gov/) data base to date is considered. While not all original research can be mentioned due to space limitations, the main aspects of MeCP2 and Rett syndrome research are discussed while highlighting the work of individual researchers and research groups. First, the primary functions of MeCP2 relevant to Rett syndrome are summarized and explored. Second, the conflicting evidence and controversies surrounding emerging aspects of MeCP2 biology are examined. Next, the most obvious gaps in MeCP2 research studies are noted. Finally, the most recent discoveries in MeCP2 and Rett syndrome research are explored with a focus on the potential and pitfalls of novel treatments and therapies.
Collapse
Affiliation(s)
- Osman Sharifi
- LaSalle Laboratory, Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, CA, United States
| | - Dag H Yasui
- LaSalle Laboratory, Department of Medical Microbiology and Immunology, UC Davis School of Medicine, Davis, CA, United States
| |
Collapse
|
26
|
Scaramuzza L, De Rocco G, Desiato G, Cobolli Gigli C, Chiacchiaretta M, Mirabella F, Pozzi D, De Simone M, Conforti P, Pagani M, Benfenati F, Cesca F, Bedogni F, Landsberger N. The enhancement of activity rescues the establishment of Mecp2 null neuronal phenotypes. EMBO Mol Med 2021; 13:e12433. [PMID: 33665914 PMCID: PMC8033520 DOI: 10.15252/emmm.202012433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 01/24/2021] [Accepted: 01/27/2021] [Indexed: 01/29/2023] Open
Abstract
MECP2 mutations cause Rett syndrome (RTT), a severe and progressive neurodevelopmental disorder mainly affecting females. Although RTT patients exhibit delayed onset of symptoms, several evidences demonstrate that MeCP2 deficiency alters early development of the brain. Indeed, during early maturation, Mecp2 null cortical neurons display widespread transcriptional changes, reduced activity, and defective morphology. It has been proposed that during brain development these elements are linked in a feed-forward cycle where neuronal activity drives transcriptional and morphological changes that further increase network maturity. We hypothesized that the enhancement of neuronal activity during early maturation might prevent the onset of RTT-typical molecular and cellular phenotypes. Accordingly, we show that the enhancement of excitability, obtained by adding to neuronal cultures Ampakine CX546, rescues transcription of several genes, neuronal morphology, and responsiveness to stimuli. Greater effects are achieved in response to earlier treatments. In vivo, short and early administration of CX546 to Mecp2 null mice prolongs lifespan, delays the disease progression, and rescues motor abilities and spatial memory, thus confirming the value for RTT of an early restoration of neuronal activity.
Collapse
Affiliation(s)
- Linda Scaramuzza
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Present address:
Department of Bioscience, University of Milan, Milan, Italy; Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Giuseppina De Rocco
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Genni Desiato
- IRCCS Humanitas Research HospitalMilanItaly
- CNR Institute of NeuroscienceMilanItaly
| | - Clementina Cobolli Gigli
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Present address:
Francis Crick InstituteLondonUK
| | - Martina Chiacchiaretta
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- Present address:
Department of NeuroscienceTufts University School of MedicineBostonMAUSA
| | - Filippo Mirabella
- IRCCS Humanitas Research HospitalMilanItaly
- Department of Biomedical Sciences, Humanitas UniversityMilanItaly
| | - Davide Pozzi
- IRCCS Humanitas Research HospitalMilanItaly
- Department of Biomedical Sciences, Humanitas UniversityMilanItaly
| | - Marco De Simone
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
- Present address:
Department of Radiation Oncology, Cedars-Sinai Medical CenterLos Angeles, CAUSA
| | - Paola Conforti
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
- Department of BiosciencesUniversity of MilanMilanItaly
| | - Massimiliano Pagani
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
- Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi”MilanItaly
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
- Present address:
Francis Crick InstituteLondonUK
| | - Fabrizia Cesca
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- Department of Life SciencesUniversity of TriesteTriesteItaly
- Present address:
Department of Radiation Oncology, Cedars-Sinai Medical CenterLos Angeles, CAUSA
| | - Francesco Bedogni
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Present address:
Neuroscience and Mental Health Research Institute (NMHRI)Division of NeuroscienceSchool of BiosciencesCardiffUK
| | - Nicoletta Landsberger
- Division of NeuroscienceIRCCS San Raffaele Scientific InstituteMilanItaly
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| |
Collapse
|
27
|
D'Mello SR. MECP2 and the Biology of MECP2 Duplication Syndrome. J Neurochem 2021; 159:29-60. [PMID: 33638179 DOI: 10.1111/jnc.15331] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/21/2021] [Accepted: 02/18/2021] [Indexed: 11/27/2022]
Abstract
MECP2 duplication syndrome (MDS), a rare X-linked genomic disorder affecting predominantly males, is caused by duplication of the chromosomal region containing the methyl CpG binding protein-2 (MECP2) gene, which encodes methyl-CpG-binding protein 2 (MECP2), a multi-functional protein required for proper brain development and maintenance of brain function during adulthood. Disease symptoms include severe motor and cognitive impairment, delayed or absent speech development, autistic features, seizures, ataxia, recurrent respiratory infections and shortened lifespan. The cellular and molecular mechanisms by which a relatively modest increase in MECP2 protein causes such severe disease symptoms are poorly understood and consequently there are no treatments available for this fatal disorder. This review summarizes what is known to date about the structure and complex regulation of MECP2 and its many functions in the developing and adult brain. Additionally, recent experimental findings on the cellular and molecular underpinnings of MDS based on cell culture and mouse models of the disorder are reviewed. The emerging picture from these studies is that MDS is a neurodegenerative disorder in which neurons die in specific parts of the central nervous system, including the cortex, hippocampus, cerebellum and spinal cord. Neuronal death likely results from astrocytic dysfunction, including a breakdown of glutamate homeostatic mechanisms. The role of elevations in the expression of glial acidic fibrillary protein (GFAP) in astrocytes and the microtubule-associated protein, Tau, in neurons to the pathogenesis of MDS is discussed. Lastly, potential therapeutic strategies to potentially treat MDS are discussed.
Collapse
|
28
|
Pizzamiglio L, Focchi E, Cambria C, Ponzoni L, Ferrara S, Bifari F, Desiato G, Landsberger N, Murru L, Passafaro M, Sala M, Matteoli M, Menna E, Antonucci F. The DNA repair protein ATM as a target in autism spectrum disorder. JCI Insight 2021; 6:133654. [PMID: 33373327 PMCID: PMC7934840 DOI: 10.1172/jci.insight.133654] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/16/2020] [Indexed: 12/20/2022] Open
Abstract
Impairment of the GABAergic system has been reported in epilepsy, autism, attention deficit hyperactivity disorder, and schizophrenia. We recently demonstrated that ataxia telangiectasia mutated (ATM) directly shapes the development of the GABAergic system. Here, we show for the first time to our knowledge how the abnormal expression of ATM affects the pathological condition of autism. We exploited 2 different animal models of autism, the methyl CpG binding protein 2-null (Mecp2y/-) mouse model of Rett syndrome and mice prenatally exposed to valproic acid, and found increased ATM levels. Accordingly, treatment with the specific ATM kinase inhibitor KU55933 (KU) normalized molecular, functional, and behavioral defects in these mouse models, such as (a) delayed GABAergic development, (b) hippocampal hyperexcitability, (c) low cognitive performances, and (d) social impairments. Mechanistically, we demonstrate that KU administration to WT hippocampal neurons leads to (a) higher early growth response 4 activity on Kcc2b promoter, (b) increased expression of Mecp2, and (c) potentiated GABA transmission. These results provide evidence and molecular substrates for the pharmacological development of ATM inhibition in autism spectrum disorders.
Collapse
Affiliation(s)
- Lara Pizzamiglio
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| | - Elisa Focchi
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| | - Clara Cambria
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| | | | - Silvia Ferrara
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| | - Francesco Bifari
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| | - Genni Desiato
- Humanitas Clinical and Research Center – IRCCS, Rozzano, Milan, Italy
| | - Nicoletta Landsberger
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| | - Luca Murru
- Institute of Neuroscience, IN-CNR, Milan, Italy
| | | | | | - Michela Matteoli
- Institute of Neuroscience, IN-CNR, Milan, Italy
- Humanitas Clinical and Research Center – IRCCS, Rozzano, Milan, Italy
| | - Elisabetta Menna
- Institute of Neuroscience, IN-CNR, Milan, Italy
- Humanitas Clinical and Research Center – IRCCS, Rozzano, Milan, Italy
| | - Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Milan, Italy
| |
Collapse
|
29
|
Pathogenic GRM7 Mutations Associated with Neurodevelopmental Disorders Impair Axon Outgrowth and Presynaptic Terminal Development. J Neurosci 2021; 41:2344-2359. [PMID: 33500274 DOI: 10.1523/jneurosci.2108-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/11/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is an inhibitory heterotrimeric G-protein-coupled receptor that modulates neurotransmitter release and synaptic plasticity at presynaptic terminals in the mammalian central nervous system. Recent studies have shown that rare mutations in glutamate receptors and synaptic scaffold proteins are associated with neurodevelopmental disorders (NDDs). However, the role of presynaptic mGlu7 in the pathogenesis of NDDs remains largely unknown. Recent whole-exome sequencing (WES) studies in families with NDDs have revealed that several missense mutations (c.1865G>A:p.R622Q; c.461T>C:p.I154T; c.1972C>T:p.R658W and c.2024C>A:p.T675K) or a nonsense mutation (c.1757G>A:p.W586X) in the GRM7 gene may be linked to NDDs. In the present study, we investigated the mechanistic links between GRM7 point mutations and NDD pathology. We find that the pathogenic GRM7 I154T and R658W/T675K mutations lead to the degradation of the mGlu7 protein. In particular, the GRM7 R658W/T675K mutation results in a lack of surface mGlu7 expression in heterologous cells and cultured neurons isolated from male and female rat embryos. We demonstrate that the expression of mGlu7 variants or exposure to mGlu7 antagonists impairs axon outgrowth through the mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling pathway during early neuronal development, which subsequently leads to a decrease in the number of presynaptic terminals in mature neurons. Treatment with an mGlu7 agonist restores the pathologic phenotypes caused by mGlu7 I154T but not by mGlu7 R658W/T675K because of its lack of neuronal surface expression. These findings provide evidence that stable neuronal surface expression of mGlu7 is essential for neural development and that mGlu7 is a promising therapeutic target for NDDs.SIGNIFICANCE STATEMENT Neurodevelopmental disorders (NDDs) affect brain development and function by multiple etiologies. Metabotropic glutamate receptor 7 (mGlu7) is a receptor that controls excitatory neurotransmission and synaptic plasticity. Since accumulating evidence indicates that the GRM7 gene locus is associated with NDD risk, we analyzed the functional effects of human GRM7 variants identified in patients with NDDs. We demonstrate that stable neuronal surface expression of mGlu7 is essential for axon outgrowth and presynaptic terminal development in neurons. We found that mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling and subsequent cytoskeletal dynamics are defective because of the degradation of mGlu7 variants. Finally, we show that the defects caused by mGlu7 I154T can be reversed by agonists, providing the rationale for proposing mGlu7 as a potential therapeutic target for NDDs.
Collapse
|
30
|
Basilico B, Morandell J, Novarino G. Molecular mechanisms for targeted ASD treatments. Curr Opin Genet Dev 2020; 65:126-137. [PMID: 32659636 DOI: 10.1016/j.gde.2020.06.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/30/2022]
Abstract
The possibility to generate construct valid animal models enabled the development and testing of therapeutic strategies targeting the core features of autism spectrum disorders (ASDs). At the same time, these studies highlighted the necessity of identifying sensitive developmental time windows for successful therapeutic interventions. Animal and human studies also uncovered the possibility to stratify the variety of ASDs in molecularly distinct subgroups, potentially facilitating effective treatment design. Here, we focus on the molecular pathways emerging as commonly affected by mutations in diverse ASD-risk genes, on their role during critical windows of brain development and the potential treatments targeting these biological processes.
Collapse
Affiliation(s)
| | - Jasmin Morandell
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
31
|
Frasca A, Spiombi E, Palmieri M, Albizzati E, Valente MM, Bergo A, Leva B, Kilstrup‐Nielsen C, Bianchi F, Di Carlo V, Di Cunto F, Landsberger N. MECP2 mutations affect ciliogenesis: a novel perspective for Rett syndrome and related disorders. EMBO Mol Med 2020; 12:e10270. [PMID: 32383329 PMCID: PMC7278541 DOI: 10.15252/emmm.201910270] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/19/2020] [Accepted: 03/26/2020] [Indexed: 12/20/2022] Open
Abstract
Mutations in MECP2 cause several neurological disorders of which Rett syndrome (RTT) represents the best-defined condition. Although mainly working as a transcriptional repressor, MeCP2 is a multifunctional protein revealing several activities, the involvement of which in RTT remains obscure. Besides being mainly localized in the nucleus, MeCP2 associates with the centrosome, an organelle from which primary cilia originate. Primary cilia function as "sensory antennae" protruding from most cells, and a link between primary cilia and mental illness has recently been reported. We herein demonstrate that MeCP2 deficiency affects ciliogenesis in cultured cells, including neurons and RTT fibroblasts, and in the mouse brain. Consequently, the cilium-related Sonic Hedgehog pathway, which is essential for brain development and functioning, is impaired. Microtubule instability participates in these phenotypes that can be rescued by HDAC6 inhibition together with the recovery of RTT-related neuronal defects. Our data indicate defects of primary cilium as a novel pathogenic mechanism that by contributing to the clinical features of RTT might impact on proper cerebellum/brain development and functioning, thus providing a novel therapeutic target.
Collapse
Affiliation(s)
- Angelisa Frasca
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Eleonora Spiombi
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Michela Palmieri
- Neuroscience DivisionIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Elena Albizzati
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Maria Maddalena Valente
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | - Anna Bergo
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | - Barbara Leva
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | - Charlotte Kilstrup‐Nielsen
- Department of Biotechnology and Life SciencesCentre of NeuroscienceUniversity of InsubriaBusto ArsizioItaly
| | | | - Valerio Di Carlo
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri OttolenghiOrbassanoItaly
- Department of NeuroscienceUniversity of TorinoTorinoItaly
| | - Nicoletta Landsberger
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanMilanItaly
- Neuroscience DivisionIRCCS San Raffaele Scientific InstituteMilanItaly
| |
Collapse
|
32
|
Fagiolini M, Patrizi A, LeBlanc J, Jin LW, Maezawa I, Sinnett S, Gray SJ, Molholm S, Foxe JJ, Johnston MV, Naidu S, Blue M, Hossain A, Kadam S, Zhao X, Chang Q, Zhou Z, Zoghbi H. Intellectual and Developmental Disabilities Research Centers: A Multidisciplinary Approach to Understand the Pathogenesis of Methyl-CpG Binding Protein 2-related Disorders. Neuroscience 2020; 445:190-206. [PMID: 32360592 DOI: 10.1016/j.neuroscience.2020.04.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/20/2022]
Abstract
Disruptions in the gene encoding methyl-CpG binding protein 2 (MECP2) underlie complex neurodevelopmental disorders including Rett Syndrome (RTT), MECP2 duplication disorder, intellectual disabilities, and autism. Significant progress has been made on the molecular and cellular basis of MECP2-related disorders providing a new framework for understanding how altered epigenetic landscape can derail the formation and refinement of neuronal circuits in early postnatal life and proper neurological function. This review will summarize selected major findings from the past years and particularly highlight the integrated and multidisciplinary work done at eight NIH-funded Intellectual and Developmental Disabilities Research Centers (IDDRC) across the US. Finally, we will outline a path forward with identification of reliable biomarkers and outcome measures, longitudinal preclinical and clinical studies, reproducibility of results across centers as a synergistic effort to decode and treat the pathogenesis of the complex MeCP2 disorders.
Collapse
Affiliation(s)
- Michela Fagiolini
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Annarita Patrizi
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jocelyn LeBlanc
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lee-Way Jin
- UC Davis MIND Institute, University of California, Sacramento, CA, USA
| | - Izumi Maezawa
- UC Davis MIND Institute, University of California, Sacramento, CA, USA
| | - Sarah Sinnett
- UNC Intellectual and Developmental Disabilities Research Center, University of North Carolina, Gene Therapy Center and Dept. of Ophthalmology, Chapel Hill, NC, USA; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Steven J Gray
- UNC Intellectual and Developmental Disabilities Research Center, University of North Carolina, Gene Therapy Center and Dept. of Ophthalmology, Chapel Hill, NC, USA; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sophie Molholm
- The Cognitive Neurophysiology Laboratory, Departments of Pediatrics, Neuroscience, and Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John J Foxe
- The Cognitive Neurophysiology Laboratory, Ernest J. Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Michael V Johnston
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Sakkubai Naidu
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Mary Blue
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Ahamed Hossain
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Shilpa Kadam
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Quiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhaolan Zhou
- Department of Genetic, Epigenetic Institute, University of Pennsylvania Perelman School of Medicine, Intellectual and Developmental Disabilities Research Center, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Huda Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
33
|
Fingolimod Modulates Dendritic Architecture in a BDNF-Dependent Manner. Int J Mol Sci 2020; 21:ijms21093079. [PMID: 32349283 PMCID: PMC7247704 DOI: 10.3390/ijms21093079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/28/2022] Open
Abstract
The brain-derived neurotrophic factor (BDNF) plays crucial roles in both the developing and mature brain. Moreover, alterations in BDNF levels are correlated with the cognitive impairment observed in several neurological diseases. Among the different therapeutic strategies developed to improve endogenous BDNF levels is the administration of the BDNF-inducing drug Fingolimod, an agonist of the sphingosine-1-phosphate receptor. Fingolimod treatment was shown to rescue diverse symptoms associated with several neurological conditions (i.e., Alzheimer disease, Rett syndrome). However, the cellular mechanisms through which Fingolimod mediates its BDNF-dependent therapeutic effects remain unclear. We show that Fingolimod regulates the dendritic architecture, dendritic spine density and morphology of healthy mature primary hippocampal neurons. Moreover, the application of Fingolimod upregulates the expression of activity-related proteins c-Fos and pERK1/2 in these cells. Importantly, we show that BDNF release is required for these actions of Fingolimod. As alterations in neuronal structure underlie cognitive impairment, we tested whether Fingolimod application might prevent the abnormalities in neuronal structure typical of two neurodevelopmental disorders, namely Rett syndrome and Cdk5 deficiency disorder. We found a significant rescue in the neurite architecture of developing cortical neurons from Mecp2 and Cdkl5 mutant mice. Our study provides insights into understanding the BDNF-dependent therapeutic actions of Fingolimod.
Collapse
|
34
|
Stansley BJ, Fisher NM, Gogliotti RG, Lindsley CW, Conn PJ, Niswender CM. Contextual Fear Extinction Induces Hippocampal Metaplasticity Mediated by Metabotropic Glutamate Receptor 5. Cereb Cortex 2019; 28:4291-4304. [PMID: 29136107 DOI: 10.1093/cercor/bhx282] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 10/05/2017] [Indexed: 12/28/2022] Open
Abstract
Dysregulated fear memory can lead to a broad spectrum of anxiety disorders. The brain systems underlying fear memory are manifold, with the hippocampus being prominently involved by housing fear-related spatial memories as engrams, which are created and stored through neural changes such as synaptic plasticity. Although metabotropic glutamate (mGlu) receptors contribute significantly to both fear behavior and hippocampal synaptic plasticity, the relationship between these two phenomena has not been fully elucidated. Here, we report that contextual fear extinction induces a novel form of metaplasticity mediated by mGlu5 at the hippocampal SC-CA1 synapse. Further, blockade of mGlu5 prevents both contextual fear extinction and expression of this metaplasticity. This form of metaplasticity was absent in a mouse model of MECP2-duplication syndrome, corresponding to a complete deficit in extinction learning. These findings suggest that mGlu5-dependent metaplasticity within the hippocampus may play a critical role in extinction of contextual fear.
Collapse
Affiliation(s)
- Branden J Stansley
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Rocco G Gogliotti
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
35
|
MeCP2-E1 isoform is a dynamically expressed, weakly DNA-bound protein with different protein and DNA interactions compared to MeCP2-E2. Epigenetics Chromatin 2019; 12:63. [PMID: 31601272 PMCID: PMC6786283 DOI: 10.1186/s13072-019-0298-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 08/22/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND MeCP2-a chromatin-binding protein associated with Rett syndrome-has two main isoforms, MeCP2-E1 and MeCP2-E2, differing in a few N-terminal amino acid residues. Previous studies have shown brain region-specific expression of these isoforms which, in addition to their different cellular localization and differential expression during brain development, suggest that they may also have non-overlapping molecular mechanisms. However, differential functions of MeCP2-E1 and E2 remain largely unexplored. RESULTS Here, we show that the N-terminal domains (NTD) of MeCP2-E1 and E2 modulate the ability of the methyl-binding domain (MBD) to interact with DNA as well as influencing the turn-over rates, binding dynamics, response to neuronal depolarization, and circadian oscillations of the two isoforms. Our proteomics data indicate that both isoforms exhibit unique interacting protein partners. Moreover, genome-wide analysis using ChIP-seq provide evidence for a shared as well as a specific regulation of different sets of genes. CONCLUSIONS Our study supports the idea that Rett syndrome might arise from simultaneous impairment of cellular processes involving non-overlapping functions of MECP2 isoforms. For instance, MeCP2-E1 mutations might impact stimuli-dependent chromatin regulation, while MeCP2-E2 mutations could result in aberrant ribosomal expression. Overall, our findings provide insight into the functional complexity of MeCP2 by dissecting differential aspects of its two isoforms.
Collapse
|
36
|
Gulmez Karaca K, Brito DV, Oliveira AM. MeCP2: A Critical Regulator of Chromatin in Neurodevelopment and Adult Brain Function. Int J Mol Sci 2019; 20:ijms20184577. [PMID: 31527487 PMCID: PMC6769791 DOI: 10.3390/ijms20184577] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 01/08/2023] Open
Abstract
Methyl CpG binding protein 2 (MeCP2) was first identified as a nuclear protein with a transcriptional repressor role that recognizes DNA methylation marks. MeCP2 has a well-established function in neurodevelopment, as evidenced by the severe neurological impairments characteristic of the Rett syndrome (RTT) pathology and the MeCP2 duplication syndrome (MDS), caused by loss or gain of MeCP2 function, respectively. Research aimed at the underlying pathophysiological mechanisms of RTT and MDS has significantly advanced our understanding of MeCP2 functions in the nervous system. It has revealed, however, that MeCP2 has more varied and complex roles than previously thought. Here we review recent insights into the functions of MeCP2 in neurodevelopment and the less explored requirement for MeCP2 in adult brain function. We focus on the emerging view that MeCP2 is a global chromatin organizer. Finally, we discuss how the individual functions of MeCP2 in neurodevelopment and adulthood are linked to its role as a chromatin regulator.
Collapse
Affiliation(s)
- Kubra Gulmez Karaca
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany; (K.G.K.)
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 EN Nijmegen, The Netherlands
| | - David V.C. Brito
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany; (K.G.K.)
| | - Ana M.M. Oliveira
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany; (K.G.K.)
- Correspondence: ; Tel.: +49-(0)6221-5416510
| |
Collapse
|
37
|
Abstract
Rett syndrome (RTT) is a severe neurological disorder caused by mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2). Almost two decades of research into RTT have greatly advanced our understanding of the function and regulation of the multifunctional protein MeCP2. Here, we review recent advances in understanding how loss of MeCP2 impacts different stages of brain development, discuss recent findings demonstrating the molecular role of MeCP2 as a transcriptional repressor, assess primary and secondary effects of MeCP2 loss and examine how loss of MeCP2 can result in an imbalance of neuronal excitation and inhibition at the circuit level along with dysregulation of activity-dependent mechanisms. These factors present challenges to the search for mechanism-based therapeutics for RTT and suggest specific approaches that may be more effective than others.
Collapse
|
38
|
Krishnaraj R, Haase F, Coorey B, Luca EJ, Wong I, Boyling A, Ellaway C, Christodoulou J, Gold WA. Genome-wide transcriptomic and proteomic studies of Rett syndrome mouse models identify common signaling pathways and cellular functions as potential therapeutic targets. Hum Mutat 2019; 40:2184-2196. [PMID: 31379106 DOI: 10.1002/humu.23887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 07/27/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022]
Abstract
The discovery that Rett syndrome is caused by mutations in the MECP2 gene has provided a major breakthrough in our understanding of the disorder. However, despite this, there is still limited understanding of the underlying pathophysiology of the disorder hampering the development of curative treatments. Over the years, a number of animal models have been developed contributing to our knowledge of the role of MECP2 in development and improving our understanding of how subtle expression levels affect brain morphology and function. Transcriptomic and proteomic studies of animal models are useful in identifying perturbations in functional pathways and providing avenues for novel areas of research into disease. This review focuses on published transcriptomic and proteomic studies of mouse models of Rett syndrome with the aim of providing a summary of all the studies, the reported dysregulated genes and functional pathways that are found to be perturbed. The 36 articles identified highlighted a number of dysfunctional pathways as well as perturbed biological networks and cellular functions including synaptic dysfunction and neuronal transmission, inflammation, and mitochondrial dysfunction. These data reveal biological insights that contribute to the disease process which may be targeted to investigate curative treatments.
Collapse
Affiliation(s)
- Rahul Krishnaraj
- Genetic Metabolic Disorders Research Unit, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Florencia Haase
- Molecular Neurobiology Research Group, Kids Research, Sydney Children's Hospitals Network, Westmead, Australia
| | - Bronte Coorey
- Molecular Neurobiology Research Group, Kids Research, Sydney Children's Hospitals Network, Westmead, Australia
| | - Edward J Luca
- University Library, The University of Sydney, Sydney, New South Wales, Australia
| | - Ingar Wong
- Molecular Neurobiology Research Group, Kids Research, Sydney Children's Hospitals Network, Westmead, Australia
| | - Alexandra Boyling
- Molecular Neurobiology Research Group, Kids Research, Sydney Children's Hospitals Network, Westmead, Australia
| | - Carolyn Ellaway
- Genetic Metabolic Disorders Research Unit, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia.,Genetic Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - John Christodoulou
- Genetic Metabolic Disorders Research Unit, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia.,Genetic Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, and Department of Paediatrics, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - Wendy A Gold
- Genetic Metabolic Disorders Research Unit, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Molecular Neurobiology Research Group, Kids Research, Sydney Children's Hospitals Network, Westmead, Australia.,Discipline of Child and Adolescent Health, The University of Sydney, Sydney, New South Wales, Australia.,Kids Neuroscience Centre, The Children's Hospital at Westmead, Kids Research, Westmead, NSW, Australia
| |
Collapse
|
39
|
Vidal S, Xiol C, Pascual-Alonso A, O'Callaghan M, Pineda M, Armstrong J. Genetic Landscape of Rett Syndrome Spectrum: Improvements and Challenges. Int J Mol Sci 2019; 20:ijms20163925. [PMID: 31409060 PMCID: PMC6719047 DOI: 10.3390/ijms20163925] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 02/06/2023] Open
Abstract
Rett syndrome (RTT) is an early-onset neurodevelopmental disorder that primarily affects females, resulting in severe cognitive and physical disabilities, and is one of the most prevalent causes of intellectual disability in females. More than fifty years after the first publication on Rett syndrome, and almost two decades since the first report linking RTT to the MECP2 gene, the research community's effort is focused on obtaining a better understanding of the genetics and the complex biology of RTT and Rett-like phenotypes without MECP2 mutations. Herein, we review the current molecular genetic studies, which investigate the genetic causes of RTT or Rett-like phenotypes which overlap with other genetic disorders and document the swift evolution of the techniques and methodologies employed. This review also underlines the clinical and genetic heterogeneity of the Rett syndrome spectrum and provides an overview of the RTT-related genes described to date, many of which are involved in epigenetic gene regulation, neurotransmitter action or RNA transcription/translation. Finally, it discusses the importance of including both phenotypic and genetic diagnosis to provide proper genetic counselling from a patient's perspective and the appropriate treatment.
Collapse
Affiliation(s)
- Silvia Vidal
- Sant Joan de Déu Research Foundation, 08950 Barcelona, Spain
- Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Clara Xiol
- Sant Joan de Déu Research Foundation, 08950 Barcelona, Spain
- Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Ainhoa Pascual-Alonso
- Sant Joan de Déu Research Foundation, 08950 Barcelona, Spain
- Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - M O'Callaghan
- Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Neurology Service, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Mercè Pineda
- Sant Joan de Déu Research Foundation, 08950 Barcelona, Spain
| | - Judith Armstrong
- Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, 08950 Barcelona, Spain.
- CIBER-ER (Biomedical Network Research Center for Rare Diseases), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain.
- Molecular and Genetics Medicine Section, Hospital Sant Joan de Déu, 08950 Barcelona, Spain.
| |
Collapse
|
40
|
Lozovaya N, Nardou R, Tyzio R, Chiesa M, Pons-Bennaceur A, Eftekhari S, Bui TT, Billon-Grand M, Rasero J, Bonifazi P, Guimond D, Gaiarsa JL, Ferrari DC, Ben-Ari Y. Early alterations in a mouse model of Rett syndrome: the GABA developmental shift is abolished at birth. Sci Rep 2019; 9:9276. [PMID: 31239460 PMCID: PMC6592949 DOI: 10.1038/s41598-019-45635-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Genetic mutations of the Methyl-CpG-binding protein-2 (MECP2) gene underlie Rett syndrome (RTT). Developmental processes are often considered to be irrelevant in RTT pathogenesis but neuronal activity at birth has not been recorded. We report that the GABA developmental shift at birth is abolished in CA3 pyramidal neurons of Mecp2-/y mice and the glutamatergic/GABAergic postsynaptic currents (PSCs) ratio is increased. Two weeks later, GABA exerts strong excitatory actions, the glutamatergic/GABAergic PSCs ratio is enhanced, hyper-synchronized activity is present and metabotropic long-term depression (LTD) is impacted. One day before delivery, maternal administration of the NKCC1 chloride importer antagonist bumetanide restored these parameters but not respiratory or weight deficits, nor the onset of mortality. Results suggest that birth is a critical period in RTT with important alterations that can be attenuated by bumetanide raising the possibility of early treatment of the disorder.
Collapse
Affiliation(s)
- N Lozovaya
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France
| | - R Nardou
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France
| | - R Tyzio
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), Department of Neurobiology, Aix-Marseille University, INSERM U1249, 13273, Marseille, France
| | - M Chiesa
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), Department of Neurobiology, Aix-Marseille University, INSERM U1249, 13273, Marseille, France
| | - A Pons-Bennaceur
- Mediterranean Institute of Neurobiology (INMED), Department of Neurobiology, Aix-Marseille University, INSERM U1249, 13273, Marseille, France
| | - S Eftekhari
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), Department of Neurobiology, Aix-Marseille University, INSERM U1249, 13273, Marseille, France
| | - T-T Bui
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), Department of Neurobiology, Aix-Marseille University, INSERM U1249, 13273, Marseille, France
| | - M Billon-Grand
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France
| | - J Rasero
- Biocruces Health Research Institute, 48903, Barakaldo, Spain
| | - P Bonifazi
- Biocruces Health Research Institute, 48903, Barakaldo, Spain.,IKERBASQUE: The Basque Foundation for Science, 48013, Bilbao, Spain
| | - D Guimond
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France
| | - J-L Gaiarsa
- Mediterranean Institute of Neurobiology (INMED), Department of Neurobiology, Aix-Marseille University, INSERM U1249, 13273, Marseille, France
| | - D C Ferrari
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France
| | - Y Ben-Ari
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc scientifique et technologique de Luminy, 13288, Marseille, cedex 09, France.
| |
Collapse
|
41
|
Cosentino L, Vigli D, Franchi F, Laviola G, De Filippis B. Rett syndrome before regression: A time window of overlooked opportunities for diagnosis and intervention. Neurosci Biobehav Rev 2019; 107:115-135. [PMID: 31108160 DOI: 10.1016/j.neubiorev.2019.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 11/29/2022]
Abstract
Rett syndrome (RTT) is a rare neurological disorder primarily affecting females, causing severe cognitive, social, motor and physiological impairments for which no cure currently exists. RTT clinical diagnosis is based on the peculiar progression of the disease, since patients show an apparently normal initial development with a subsequent sudden regression at around 2 years of age. Accumulating evidences are rising doubts regarding the absence of early impairments, hence questioning the concept of regression. We reviewed the published literature addressing the pre-symptomatic stage of the disease in both patients and animal models with a particular focus on behavioral, physiological and brain abnormalities. The emerging picture delineates subtle, but reliable impairments that precede the onset of overt symptoms whose bases are likely set up already during embryogenesis. Some of the outlined alterations appear transient, suggesting compensatory mechanisms to occur in the course of development. There is urgent need for more systematic developmental analyses able to detect early pathological markers to be used as diagnostic tools and precocious targets of time-specific interventions.
Collapse
Affiliation(s)
- Livia Cosentino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Daniele Vigli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Francesca Franchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Giovanni Laviola
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| |
Collapse
|
42
|
MicroRNA-197 controls ADAM10 expression to mediate MeCP2's role in the differentiation of neuronal progenitors. Cell Death Differ 2018; 26:1863-1879. [PMID: 30560934 PMCID: PMC6748079 DOI: 10.1038/s41418-018-0257-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/22/2018] [Accepted: 12/03/2018] [Indexed: 01/17/2023] Open
Abstract
Duplication of MECP2 (Methyl-CpG-binding protein 2) causes severe mental illness called MECP2 duplication syndrome (MDS), yet the underlying mechanism remains elusive. Here we show, in Tg(MECP2) transgenic mouse brain or cultured neural progenitor cells (NPCs), that elevated MeCP2 expression promotes NPC differentiation into neurons. Ectopic expression of MeCP2 inhibits ADAM10 and thus the NOTCH pathway during NPC differentiation. In human cells, this downregulation on ADAM10 was mediated by miRNA-197, which is upregulated by MeCP2. Surprisingly, miR-197 binds to the ADAM10 3′-UTR via its 3′ side, not the canonical seed sequence on the 5′ side. In mouse cells, a noncoding RNA Gm28836 is used to replace the function of miR-197 between MeCP2 and ADAM10. Similar to MeCP2, overexpressing miR-197 also promotes NPCs differentiation into neurons. Interestingly, three rare missense mutations (H371R, E394K, and G428S) in MECP2, which we identified in a Han Chinese autism spectrum disorders (ASD) cohort showed loss-of-function effects in NPC differentiation assay. These mutations cannot upregulate miR-197. Overexpressing miR-197 together with these MeCP2 mutations could rescue the downregulation on ADAM10. Not only the inhibitor of miR-197 could reverse the effect of overexpressed MeCP2 on NPCs differentiation, but also overexpression of miR-197 could reverse the NPCs differentiation defects caused by MECP2 mutations. Our results revealed that a regulatory axis involving MeCP2, miR-197, ADAM10, and NOTCH signaling is critical for NPC differentiation, which is affected by both MeCP2 duplication and mutation.
Collapse
|
43
|
Gandaglia A, Brivio E, Carli S, Palmieri M, Bedogni F, Stefanelli G, Bergo A, Leva B, Cattaneo C, Pizzamiglio L, Cicerone M, Bianchi V, Kilstrup-Nielsen C, D’Annessa I, Di Marino D, D’Adamo P, Antonucci F, Frasca A, Landsberger N. A Novel Mecp2Y120D Knock-in Model Displays Similar Behavioral Traits But Distinct Molecular Features Compared to the Mecp2-Null Mouse Implying Precision Medicine for the Treatment of Rett Syndrome. Mol Neurobiol 2018; 56:4838-4854. [DOI: 10.1007/s12035-018-1412-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/24/2018] [Indexed: 10/27/2022]
|
44
|
Fisher NM, Seto M, Lindsley CW, Niswender CM. Metabotropic Glutamate Receptor 7: A New Therapeutic Target in Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:387. [PMID: 30405350 PMCID: PMC6206046 DOI: 10.3389/fnmol.2018.00387] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/01/2018] [Indexed: 12/27/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are characterized by a wide range of symptoms including delayed speech, intellectual disability, motor dysfunction, social deficits, breathing problems, structural abnormalities, and epilepsy. Unfortunately, current treatment strategies are limited and innovative new approaches are sorely needed to address these complex diseases. The metabotropic glutamate receptors are a class of G protein-coupled receptors that act to modulate neurotransmission across many brain structures. They have shown great promise as drug targets for numerous neurological and psychiatric diseases. Moreover, the development of subtype-selective allosteric modulators has allowed detailed studies of each receptor subtype. Here, we focus on the metabotropic glutamate receptor 7 (mGlu7) as a potential therapeutic target for NDDs. mGlu7 is expressed widely throughout the brain in regions that correspond to the symptom domains listed above and has established roles in synaptic physiology and behavior. Single nucleotide polymorphisms and mutations in the GRM7 gene have been associated with idiopathic autism and other NDDs in patients. In rodent models, existing literature suggests that decreased mGlu7 expression and/or function may lead to symptoms that overlap with those of NDDs. Furthermore, potentiation of mGlu7 activity has shown efficacy in a mouse model of Rett syndrome. In this review, we summarize current findings that provide rationale for the continued development of mGlu7 modulators as potential therapeutics.
Collapse
Affiliation(s)
- Nicole M Fisher
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Mabel Seto
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States
| | - Craig W Lindsley
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Department of Chemistry, Vanderbilt University, Nashville, TN, United States
| | - Colleen M Niswender
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, United States.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
45
|
Balakrishnan S, Mironov SL. Regenerative glutamate release in the hippocampus of Rett syndrome model mice. PLoS One 2018; 13:e0202802. [PMID: 30256804 PMCID: PMC6157837 DOI: 10.1371/journal.pone.0202802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/09/2018] [Indexed: 11/18/2022] Open
Abstract
Excess glutamate during intense neuronal activity is not instantly cleared and may accumulate in the extracellular space. This has various long-term consequences such as ectopic signaling, modulation of synaptic efficacy and excitotoxicity; the latter implicated in various neurodevelopmental and neurodegenerative diseases. In this study, the quantitative imaging of glutamate homeostasis of hippocampal slices from methyl-CpG binding protein 2 knock-out (Mecp2-/y) mice, a model of Rett syndrome (RTT), revealed unusual repetitive glutamate transients. They appeared in phase with bursts of action potentials in the CA1 neurons. Both glutamate transients and bursting activity were suppressed by the blockade of sodium, AMPA and voltage-gated calcium channels (T- and R-type), and enhanced after the inhibition of HCN channels. HCN and calcium channels in RTT and wild-type (WT) CA1 neurons displayed different voltage-dependencies and kinetics. Both channels modulated postsynaptic integration and modified the pattern of glutamate spikes in the RTT hippocampus. Spontaneous glutamate transients were much less abundant in the WT preparations, and, when observed, had smaller amplitude and frequency. The basal ambient glutamate levels in RTT were higher and transient glutamate increases (spontaneous and evoked by stimulation of Schaffer collaterals) decayed slower. Both features indicate less efficient glutamate uptake in RTT. To explain the generation of repetitive glutamate spikes, we designed a novel model of glutamate-induced glutamate release. The simulations correctly predicted the patterns of spontaneous glutamate spikes observed under different experimental conditions. We propose that pervasive spontaneous glutamate release is a hallmark of Mecp2-/y hippocampus, stemming from and modulating the hyperexcitability of neurons.
Collapse
Affiliation(s)
- Saju Balakrishnan
- CNMPB (Centre for Nanoscale Microscopy and Molecular Physiology of the Brain, DFG Research Center 103), Institute of Neuro and Sensory Physiology, Georg-August-University, Göttingen, Germany
| | - Sergej L. Mironov
- CNMPB (Centre for Nanoscale Microscopy and Molecular Physiology of the Brain, DFG Research Center 103), Institute of Neuro and Sensory Physiology, Georg-August-University, Göttingen, Germany
| |
Collapse
|
46
|
Morini R, Ferrara S, Perrucci F, Zambetti S, Pelucchi S, Marcello E, Gardoni F, Antonucci F, Matteoli M, Menna E. Lack of the Actin Capping Protein, Eps8, Affects NMDA-Type Glutamate Receptor Function and Composition. Front Mol Neurosci 2018; 11:313. [PMID: 30233314 PMCID: PMC6133960 DOI: 10.3389/fnmol.2018.00313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/15/2018] [Indexed: 11/26/2022] Open
Abstract
Actin-based remodeling underlines spine morphogenesis and plasticity and is crucially involved in the processes that constantly reshape the circuitry of the adult brain in response to external stimuli, leading to learning and memory formation and supporting cognitive functions. Hence spine morphology and synaptic strength are tightly linked and indeed abnormalities in spine number and morphology have been described in a number of neurological disorders such as autism spectrum disorders (ASDs), schizophrenia and intellectual disabilities. We have recently demonstrated that the actin regulating protein, Epidermal growth factor receptor pathway substrate 8 (Eps8), is essential for spine growth and long term potentiation. Indeed, mice lacking Eps8 display immature filopodia-like spines, which are unable to undergo potentiation, and are impaired in cognitive functions. Furthermore, reduced levels of Eps8 have been found in the brain of a cohort of patients affected by ASD compared to controls. Here we investigated whether the lack of Eps8, which is also part of the N-methyl-d-aspartate (NMDA) receptor complex, affects the functional maturation of the postsynaptic compartment. Our results demonstrate that Eps8 knock out mice (Eps8 KO) neurons display altered synaptic expression and subunit composition of NMDA receptors (i.e., increased GluN2B-, decreased GluN2A-containing receptors) and impaired GluN2B to GluN2A subunit shift. Indeed Eps8 KO neurons display increased content of GluN2B containing NMDA receptors both at the synaptic and extrasynaptic level. Furthermore, Eps8 KO neurons display an increased content of extra-synaptic GluN2B-containing receptors, suggesting that also the synaptic targeting of NMDA receptors is affected by the lack of Eps8. These data demonstrate that, besides regulation of spine morphogenesis, Eps8 also regulates the synaptic balance of NMDA receptors subunits GluN2A and GluN2B.
Collapse
Affiliation(s)
- Raffaella Morini
- Laboratory of Pharmacology and Brain Pathology, Neurocenter IRCCS Humanitas, Milan, Italy
| | - Silvia Ferrara
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università di Milano, Milan, Italy
| | - Fabio Perrucci
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Stefania Zambetti
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università di Milano, Milan, Italy
| | - Silvia Pelucchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milan, Italy.,NEUROFARBA, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Università degli Studi di Firenze, Florence, Italy
| | - Elena Marcello
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università di Milano, Milan, Italy
| | - Flavia Antonucci
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università di Milano, Milan, Italy
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Neurocenter IRCCS Humanitas, Milan, Italy.,CNR-Istituto di Neuroscienze (IN), Milan, Italy
| | - Elisabetta Menna
- Laboratory of Pharmacology and Brain Pathology, Neurocenter IRCCS Humanitas, Milan, Italy.,CNR-Istituto di Neuroscienze (IN), Milan, Italy
| |
Collapse
|
47
|
Landucci E, Brindisi M, Bianciardi L, Catania LM, Daga S, Croci S, Frullanti E, Fallerini C, Butini S, Brogi S, Furini S, Melani R, Molinaro A, Lorenzetti FC, Imperatore V, Amabile S, Mariani J, Mari F, Ariani F, Pizzorusso T, Pinto AM, Vaccarino FM, Renieri A, Campiani G, Meloni I. iPSC-derived neurons profiling reveals GABAergic circuit disruption and acetylated α-tubulin defect which improves after iHDAC6 treatment in Rett syndrome. Exp Cell Res 2018; 368:225-235. [PMID: 29730163 DOI: 10.1016/j.yexcr.2018.05.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/22/2022]
Abstract
Mutations in MECP2 gene have been identified in more than 95% of patients with classic Rett syndrome, one of the most common neurodevelopmental disorders in females. Taking advantage of the breakthrough technology of genetic reprogramming, we investigated transcriptome changes in neurons differentiated from induced Pluripotent Stem Cells (iPSCs) derived from patients with different mutations. Profiling by RNA-seq in terminally differentiated neurons revealed a prominent GABAergic circuit disruption along with a perturbation of cytoskeleton dynamics. In particular, in mutated neurons we identified a significant decrease of acetylated α-tubulin which can be reverted by treatment with selective inhibitors of HDAC6, the main α-tubulin deacetylase. These findings contribute to shed light on Rett pathogenic mechanisms and provide hints for the treatment of Rett-associated epileptic behavior as well as for the definition of new therapeutic strategies for Rett syndrome.
Collapse
Affiliation(s)
- Elisa Landucci
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Margherita Brindisi
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Laura Bianciardi
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Lorenza M Catania
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Sergio Daga
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Susanna Croci
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Elisa Frullanti
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Chiara Fallerini
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Stefania Butini
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Simone Brogi
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Simone Furini
- Department of Medical Biotechnologies, University of Siena, Strada delle Scotte 4, 53100 Siena, Italy
| | - Riccardo Melani
- Institute of Neuroscience, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy
| | - Angelo Molinaro
- Institute of Neuroscience, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy
| | | | - Valentina Imperatore
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Sonia Amabile
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Jessica Mariani
- Yale University, Child Study Center, 230 South Frontage Rd, New Haven, CT 06520, United States
| | - Francesca Mari
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy
| | - Francesca Ariani
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy
| | - Tommaso Pizzorusso
- Institute of Neuroscience, National Research Council (CNR), Via Giuseppe Moruzzi, 1, 56124 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy; BIO@SNS lab, Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56126 Pisa, Italy
| | - Anna Maria Pinto
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy
| | - Flora M Vaccarino
- Yale University, Child Study Center, 230 South Frontage Rd, New Haven, CT 06520, United States
| | - Alessandra Renieri
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy; Genetica Medica, Azienda Ospedaliera Universitaria Senese, Viale Mario Bracci 2, 53100 Siena, Italy.
| | - Giuseppe Campiani
- NatSynDrugs, Department of Biotechnology, Chemistry and Pharmacy, DoE 2018-2022 University of Siena, via Aldo Moro 2, 53100 Siena, Italy.
| | - Ilaria Meloni
- Medical Genetics, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| |
Collapse
|
48
|
Gogliotti RG, Senter RK, Fisher NM, Adams J, Zamorano R, Walker AG, Blobaum AL, Engers DW, Hopkins CR, Daniels JS, Jones CK, Lindsley CW, Xiang Z, Conn PJ, Niswender CM. mGlu 7 potentiation rescues cognitive, social, and respiratory phenotypes in a mouse model of Rett syndrome. Sci Transl Med 2018; 9:9/403/eaai7459. [PMID: 28814546 DOI: 10.1126/scitranslmed.aai7459] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 01/25/2017] [Accepted: 04/27/2017] [Indexed: 12/20/2022]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in the methyl-CpG binding protein 2 (MECP2) gene. The cognitive impairments seen in mouse models of RTT correlate with deficits in long-term potentiation (LTP) at Schaffer collateral (SC)-CA1 synapses in the hippocampus. Metabotropic glutamate receptor 7 (mGlu7) is the predominant mGlu receptor expressed presynaptically at SC-CA1 synapses in adult mice, and its activation on GABAergic interneurons is necessary for induction of LTP. We demonstrate that pathogenic mutations in MECP2 reduce mGlu7 protein expression in brain tissue from RTT patients and in MECP2-deficient mouse models. In rodents, this reduction impairs mGlu7-mediated control of synaptic transmission. We show that positive allosteric modulation of mGlu7 activity restores LTP and improves contextual fear learning, novel object recognition, and social memory. Furthermore, mGlu7 positive allosteric modulation decreases apneas in Mecp2+/- mice, suggesting that mGlu7 may be a potential therapeutic target for multiple aspects of the RTT phenotype.
Collapse
Affiliation(s)
- Rocco G Gogliotti
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Rebecca K Senter
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Nicole M Fisher
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Jeffrey Adams
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Rocio Zamorano
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Adam G Walker
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Anna L Blobaum
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Darren W Engers
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Corey R Hopkins
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - J Scott Daniels
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Zixiu Xiang
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA. .,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN 37232, USA.,Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
49
|
Mellios N, Feldman DA, Sheridan SD, Ip JP, Kwok S, Amoah SK, Rosen B, Rodriguez BA, Crawford B, Swaminathan R, Chou S, Li Y, Ziats M, Ernst C, Jaenisch R, Haggarty SJ, Sur M. MeCP2-regulated miRNAs control early human neurogenesis through differential effects on ERK and AKT signaling. Mol Psychiatry 2018; 23:1051-1065. [PMID: 28439102 PMCID: PMC5815944 DOI: 10.1038/mp.2017.86] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 02/12/2017] [Accepted: 02/28/2017] [Indexed: 12/15/2022]
Abstract
Rett syndrome (RTT) is an X-linked, neurodevelopmental disorder caused primarily by mutations in the methyl-CpG-binding protein 2 (MECP2) gene, which encodes a multifunctional epigenetic regulator with known links to a wide spectrum of neuropsychiatric disorders. Although postnatal functions of MeCP2 have been thoroughly investigated, its role in prenatal brain development remains poorly understood. Given the well-established importance of microRNAs (miRNAs) in neurogenesis, we employed isogenic human RTT patient-derived induced pluripotent stem cell (iPSC) and MeCP2 short hairpin RNA knockdown approaches to identify novel MeCP2-regulated miRNAs enriched during early human neuronal development. Focusing on the most dysregulated miRNAs, we found miR-199 and miR-214 to be increased during early brain development and to differentially regulate extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase and protein kinase B (PKB/AKT) signaling. In parallel, we characterized the effects on human neurogenesis and neuronal differentiation brought about by MeCP2 deficiency using both monolayer and three-dimensional (cerebral organoid) patient-derived and MeCP2-deficient neuronal culture models. Inhibiting miR-199 or miR-214 expression in iPSC-derived neural progenitors deficient in MeCP2 restored AKT and ERK activation, respectively, and ameliorated the observed alterations in neuronal differentiation. Moreover, overexpression of miR-199 or miR-214 in the wild-type mouse embryonic brains was sufficient to disturb neurogenesis and neuronal migration in a similar manner to Mecp2 knockdown. Taken together, our data support a novel miRNA-mediated pathway downstream of MeCP2 that influences neurogenesis via interactions with central molecular hubs linked to autism spectrum disorders.
Collapse
Affiliation(s)
- Nikolaos Mellios
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, 87131,Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139,Correspondence to and
| | - Danielle A. Feldman
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Steven D. Sheridan
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139,Chemical Neurobiology Laboratory, Center for Human Genetic Research, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Jacque P.K. Ip
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Showming Kwok
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Stephen K. Amoah
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, 87131
| | - Bess Rosen
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brian A. Rodriguez
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, 87131
| | - Benjamin Crawford
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Radha Swaminathan
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, 87131
| | - Stephanie Chou
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Yun Li
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Mark Ziats
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892
| | - Carl Ernst
- Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Center for Human Genetic Research, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139,Correspondence to and
| |
Collapse
|
50
|
Kyle SM, Vashi N, Justice MJ. Rett syndrome: a neurological disorder with metabolic components. Open Biol 2018; 8:170216. [PMID: 29445033 PMCID: PMC5830535 DOI: 10.1098/rsob.170216] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
Rett syndrome (RTT) is a neurological disorder caused by mutations in the X-linked gene methyl-CpG-binding protein 2 (MECP2), a ubiquitously expressed transcriptional regulator. Despite remarkable scientific progress since its discovery, the mechanism by which MECP2 mutations cause RTT symptoms is largely unknown. Consequently, treatment options for patients are currently limited and centred on symptom relief. Thought to be an entirely neurological disorder, RTT research has focused on the role of MECP2 in the central nervous system. However, the variety of phenotypes identified in Mecp2 mutant mouse models and RTT patients implicate important roles for MeCP2 in peripheral systems. Here, we review the history of RTT, highlighting breakthroughs in the field that have led us to present day. We explore the current evidence supporting metabolic dysfunction as a component of RTT, presenting recent studies that have revealed perturbed lipid metabolism in the brain and peripheral tissues of mouse models and patients. Such findings may have an impact on the quality of life of RTT patients as both dietary and drug intervention can alter lipid metabolism. Ultimately, we conclude that a thorough knowledge of MeCP2's varied functional targets in the brain and body will be required to treat this complex syndrome.
Collapse
Affiliation(s)
- Stephanie M Kyle
- Genetics and Genome Biology Program, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada M5G 0A4
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Neeti Vashi
- Genetics and Genome Biology Program, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada M5G 0A4
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A1
| | - Monica J Justice
- Genetics and Genome Biology Program, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada M5G 0A4
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A1
| |
Collapse
|