1
|
Gonçalez JL, Shen J, Li W. Molecular Mechanisms of Rett Syndrome: Emphasizing the Roles of Monoamine, Immunity, and Mitochondrial Dysfunction. Cells 2024; 13:2077. [PMID: 39768168 PMCID: PMC11674639 DOI: 10.3390/cells13242077] [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: 11/08/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Rett syndrome (RTT), which predominantly affects females, arises in most cases from mutations in the Methyl-CpG-binding Protein-2 (MECP2) gene. When MeCP2 is impaired, it disrupts the regulation of numerous genes, causing the production of dysfunctional proteins associated with various multi-systemic issues in RTT. In this review, we explore the current insights into molecular signaling related to monoamines, immune response, and mitochondrial function, and their implications for the pathophysiology of RTT. Research has shown that monoamines-such as dopamine, norepinephrine, epinephrine, serotonin, and histamine-exhibit alterations in RTT, contributing to a range of neurological symptoms. Furthermore, the immune system in RTT individuals demonstrates dysfunction through the abnormal activity of microglia, macrophages, lymphocytes, and non-immune cells, leading to the atypical release of inflammatory mediators and disruptions in the NF-κB signaling pathway. Moreover, mitochondria, essential for energy production and calcium storage, also show dysfunction in this condition. The delicate balance of producing and scavenging reactive oxygen species-termed redox balance-is disrupted in RTT. Targeting these molecular pathways presents a promising avenue for developing effective therapies.
Collapse
Affiliation(s)
- Julia Lopes Gonçalez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.L.G.); (J.S.)
- Graduate Program in Behavioral Neuroscience, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jenny Shen
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.L.G.); (J.S.)
| | - Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.L.G.); (J.S.)
| |
Collapse
|
2
|
Cosentino L, Urbinati C, Lanzillotta C, De Rasmo D, Valenti D, Pellas M, Quattrini MC, Piscitelli F, Kostrzewa M, Di Domenico F, Pietraforte D, Bisogno T, Signorile A, Vacca RA, De Filippis B. Pharmacological inhibition of the CB1 cannabinoid receptor restores abnormal brain mitochondrial CB1 receptor expression and rescues bioenergetic and cognitive defects in a female mouse model of Rett syndrome. Mol Autism 2024; 15:39. [PMID: 39300547 PMCID: PMC11414047 DOI: 10.1186/s13229-024-00617-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Defective mitochondria and aberrant brain mitochondrial bioenergetics are consistent features in syndromic intellectual disability disorders, such as Rett syndrome (RTT), a rare neurologic disorder that severely affects mainly females carrying mutations in the X-linked MECP2 gene. A pool of CB1 cannabinoid receptors (CB1R), the primary receptor subtype of the endocannabinoid system in the brain, is located on brain mitochondrial membranes (mtCB1R), where it can locally regulate energy production, synaptic transmission and memory abilities through the inhibition of the intra-mitochondrial protein kinase A (mtPKA). In the present study, we asked whether an overactive mtCB1R-mtPKA signaling might underlie the brain mitochondrial alterations in RTT and whether its modulation by systemic administration of the CB1R inverse agonist rimonabant might improve bioenergetics and cognitive defects in mice modeling RTT. METHODS Rimonabant (0.3 mg/kg/day, intraperitoneal injections) was administered daily to symptomatic female mice carrying a truncating mutation of the Mecp2 gene and its effects on brain mitochondria functionality, systemic oxidative status, and memory function were assessed. RESULTS mtCB1R is overexpressed in the RTT mouse brain. Subchronic treatment with rimonabant normalizes mtCB1R expression in RTT mouse brains, boosts mtPKA signaling, and restores the defective brain mitochondrial bioenergetics, abnormal peripheral redox homeostasis, and impaired cognitive abilities in RTT mice. LIMITATIONS The lack of selectivity of the rimonabant treatment towards mtCB1R does not allow us to exclude that the beneficial effects exerted by the treatment in the RTT mouse model may be ascribed more broadly to the modulation of CB1R activity and distribution among intracellular compartments, rather than to a selective effect on mtCB1R-mediated signaling. The low sample size of few experiments is a further limitation that has been addressed replicating the main findings under different experimental conditions. CONCLUSIONS The present data identify mtCB1R overexpression as a novel molecular alteration in the RTT mouse brain that may underlie defective brain mitochondrial bioenergetics and cognitive dysfunction.
Collapse
Affiliation(s)
- Livia Cosentino
- Center for Behavioral Sciences and Mental Health, Italian National Institute of Health, Rome, Italy
| | - Chiara Urbinati
- Center for Behavioral Sciences and Mental Health, Italian National Institute of Health, Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | - Domenico De Rasmo
- Institute of Biomembranes Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Daniela Valenti
- Institute of Biomembranes Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Mattia Pellas
- Center for Behavioral Sciences and Mental Health, Italian National Institute of Health, Rome, Italy
| | | | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
| | - Magdalena Kostrzewa
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Rome, Italy
| | | | - Tiziana Bisogno
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Anna Signorile
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy.
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Italian National Institute of Health, Rome, Italy.
| |
Collapse
|
3
|
Handy NB, Xu Y, Moon D, Sowizral JJ, Moon E, Ho M, Wilson BA. Hierarchical determinants in cytotoxic necrotizing factor (CNF) toxins driving Rho G-protein deamidation versus transglutamination. mBio 2024; 15:e0122124. [PMID: 38920360 PMCID: PMC11253639 DOI: 10.1128/mbio.01221-24] [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: 04/27/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
The cytotoxic necrotizing factor (CNF) family of AB-type bacterial protein toxins catalyze two types of modification on their Rho GTPase substrates: deamidation and transglutamination. It has been established that E. coli CNF1 and its close homolog proteins catalyze primarily deamidation and Bordetella dermonecrotic toxin (DNT) catalyzes primarily transglutamination. The rapidly expanding microbial genome sequencing data have revealed that there are at least 13 full-length variants of CNF1 homologs. CNFx from E. coli strain GN02091 is the most distant from all other members of the CNF family with 50%-55% sequence identity at the protein level and 0.45-0.52 nucleotide substitutions per site at the DNA level. CNFx modifies RhoA, Rac1, and Cdc42, and like CNF1, activates downstream SRE-dependent mitogenic signaling pathways in human HEK293T cells, but at a 1,000-fold higher EC50 value. Unlike other previously characterized CNF toxins, CNFx modifies Rho proteins primarily through transglutamination, as evidenced by gel-shift assay and confirmed by MALDI mass spectral analysis, when coexpressed with Rho-protein substrates in E. coli BL21 cells or through direct treatment of HEK293T cells. A comparison of CNF1 and CNFx sequences identified two critical active-site residues corresponding to positions 832 and 862 in CNF1. Reciprocal site-specific mutations at these residues in each toxin revealed hierarchical rules that define the preference for deamidase versus a transglutaminase activity in CNFs. An additional unique Cys residue at the C-terminus of CNFx was also discovered to be critical for retarding cargo delivery.IMPORTANCECytotoxic necrotizing factor (CNF) toxins not only play important virulence roles in pathogenic E. coli and other bacterial pathogens, but CNF-like genes have also been found in an expanding number of genomes from clinical isolates. Harnessing the power of evolutionary relationships among the CNF toxins enabled the deciphering of the hierarchical active-site determinants that define whether they modify their Rho GTPase substrates through deamidation or transglutamination. With our finding that a distant CNF variant (CNFx) unlike other known CNFs predominantly transglutaminates its Rho GTPase substrates, the paradigm of "CNFs deamidate and DNTs transglutaminate" could finally be attributed to two critical amino acid residues within the active site other than the previously identified catalytic Cys-His dyad residues. The significance of our approach and research findings is that they can be applied to deciphering enzyme reaction determinants and substrate specificities for other bacterial proteins in the development of precision therapeutic strategies.
Collapse
Affiliation(s)
- Nicholas B. Handy
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yiting Xu
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Damee Moon
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jacob J. Sowizral
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Eric Moon
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Mengfei Ho
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Brenda A. Wilson
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
4
|
Villablanca C, Vidal R, Gonzalez-Billault C. Are cytoskeleton changes observed in astrocytes functionally linked to aging? Brain Res Bull 2023; 196:59-67. [PMID: 36935053 DOI: 10.1016/j.brainresbull.2023.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/22/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
Astrocytes are active participants in the performance of the Central Nervous System (CNS) in both health and disease. During aging, astrocytes are susceptible to reactive astrogliosis, a molecular state characterized by functional changes in response to pathological situations, and cellular senescence, characterized by loss of cell division, apoptosis resistance, and gain of proinflammatory functions. This results in two different states of astrocytes, which can produce proinflammatory phenotypes with harmful consequences in chronic conditions. Reactive astrocytes and senescent astrocytes share morpho-functional features that are dependent on the organization of the cytoskeleton. However, such changes in the cytoskeleton have yet to receive the necessary attention to explain their role in the alterations of astrocytes that are associated with aging and pathologies. In this review, we summarize all the available findings that connect changes in the cytoskeleton of the astrocytes with aging. In addition, we discuss future avenues that we believe will guide such a novel topic.
Collapse
Affiliation(s)
- Cristopher Villablanca
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - René Vidal
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Institute for Nutrition and Food Technologies, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
5
|
Sun J, Osenberg S, Irwin A, Ma LH, Lee N, Xiang Y, Li F, Wan YW, Park IH, Maletic-Savatic M, Ballas N. Mutations in the transcriptional regulator MeCP2 severely impact key cellular and molecular signatures of human astrocytes during maturation. Cell Rep 2023; 42:111942. [PMID: 36640327 PMCID: PMC10857774 DOI: 10.1016/j.celrep.2022.111942] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 06/12/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Mutations in the MECP2 gene underlie a spectrum of neurodevelopmental disorders, most commonly Rett syndrome (RTT). We ask whether MECP2 mutations interfere with human astrocyte developmental maturation, thereby affecting their ability to support neurons. Using human-based models, we show that RTT-causing MECP2 mutations greatly impact the key role of astrocytes in regulating overall brain bioenergetics and that these metabolic aberrations are likely mediated by dysfunctional mitochondria. During post-natal maturation, astrocytes rely on neurons to induce their complex stellate morphology and transcriptional changes. While MECP2 mutations cause cell-intrinsic aberrations in the astrocyte transcriptional landscape, surprisingly, they do not affect the neuron-induced astrocyte gene expression. Notably, however, astrocytes are unable to develop complex mature morphology due to cell- and non-cell-autonomous aberrations caused by MECP2 mutations. Thus, MECP2 mutations critically impact key cellular and molecular features of human astrocytes and, hence, their ability to interact and support the structural and functional maturation of neurons.
Collapse
Affiliation(s)
- Jialin Sun
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Sivan Osenberg
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA; Departments of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Austin Irwin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Li-Hua Ma
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nigel Lee
- Departments of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Yangfei Xiang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Feng Li
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA; Center for Drug Discovery and Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ying-Wooi Wan
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mirjana Maletic-Savatic
- Departments of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA.
| | - Nurit Ballas
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
6
|
Fuchs C, Cosentino L, Urbinati C, Talamo MC, Medici G, Quattrini MC, Mottolese N, Pietraforte D, Fuso A, Ciani E, De Filippis B. Treatment with FRAX486 rescues neurobehavioral and metabolic alterations in a female mouse model of CDKL5 deficiency disorder. CNS Neurosci Ther 2022; 28:1718-1732. [PMID: 35932179 PMCID: PMC9532911 DOI: 10.1111/cns.13907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/21/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction CDKL5 deficiency disorder (CDD) is a rare neurodevelopmental condition, primarily affecting girls for which no cure currently exists. Neuronal morphogenesis and plasticity impairments as well as metabolic dysfunctions occur in CDD patients. The present study explored the potential therapeutic value for CDD of FRAX486, a brain‐penetrant molecule that was reported to selectively inhibit group I p21‐activated kinases (PAKs), serine/threonine kinases critically involved in the regulation of neuronal morphology and glucose homeostasis. Methods The effects of treatment with FRAX486 on CDD‐related alterations were assessed in vitro (100 nM for 48 h) on primary hippocampal cultures from Cdkl5‐knockout male mice (Cdkl5‐KO) and in vivo (20 mg/Kg, s.c. for 5 days) on Cdkl5‐KO heterozygous females (Cdkl5‐Het). Results The in vitro treatment with FRAX486 completely rescued the abnormal neuronal maturation and the number of PSD95‐positive puncta in Cdkl5‐KO mouse neurons. In vivo, FRAX486 normalized the general health status, the hyperactive profile and the fear learning defects of fully symptomatic Cdkl5‐Het mice. Systemically, FRAX486 treatment normalized the levels of reactive oxidizing species in the whole blood and the fasting‐induced hypoglycemia displayed by Cdkl5‐Het mice. In the hippocampus of Cdkl5‐Het mice, treatment with FRAX486 rescued spine maturation and PSD95 expression and restored the abnormal PAKs phosphorylation at sites which are critical for their activation (P‐PAK‐Ser144/141/139) or for the control cytoskeleton remodeling (P‐PAK1‐Thr212). Conclusions Present results provide evidence that PAKs may represent innovative therapeutic targets for CDD.
Collapse
Affiliation(s)
- Claudia Fuchs
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | - Livia Cosentino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Chiara Urbinati
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Cristina Talamo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Giorgio Medici
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | | | - Nicola Mottolese
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | | | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Science, University of Bologna, Bologna, Italy
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
7
|
Albizzati E, Florio E, Miramondi F, Sormonta I, Landsberger N, Frasca A. Identification of Region-Specific Cytoskeletal and Molecular Alterations in Astrocytes of Mecp2 Deficient Animals. Front Neurosci 2022; 16:823060. [PMID: 35242007 PMCID: PMC8886113 DOI: 10.3389/fnins.2022.823060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that represents the most common genetic cause of severe intellectual disability in females. Most patients carry mutations in the X-linked MECP2 gene, coding for the methyl-CpG-binding protein 2 (MeCP2), originally isolated as an epigenetic transcriptional factor able to bind methylated DNA and repress transcription. Recent data implicated a role for glia in RTT, showing that astrocytes express Mecp2 and that its deficiency affects their ability to support neuronal maturation by non-cell autonomous mechanisms. To date, some molecular, structural and functional alterations have been attributed to Mecp2 null astrocytes, but how they evolve over time and whether they follow a spatial heterogeneity are two aspects which deserve further investigations. In this study, we assessed cytoskeletal features of astrocytes in Mecp2 deficient brains by analyzing their arbor complexity and processes in reconstructed GFAP+ cells at different ages, corresponding to peculiar stages of the disorder, and in different cerebral regions (motor and somatosensory cortices and CA1 layer of hippocampus). Our findings demonstrate the presence of defects in Mecp2 null astrocytes that worsen along disease progression and strictly depend on the brain area, highlighting motor and somatosensory cortices as the most affected regions. Of relevance, astrocyte cytoskeleton is impaired also in the somatosensory cortex of symptomatic heterozygous animals, with Mecp2 + astrocytes showing slightly more pronounced defects with respect to the Mecp2 null cells, emphasizing the importance of non-cell autonomous effects. We reported a temporal correlation between the progressive thinning of layer I and the atrophy of astrocytes, suggesting that their cytoskeletal dysfunctions might contribute to cortical defects. Considering the reciprocal link between morphology and function in astrocytes, we analyzed the effect of Mecp2 deficiency on the expression of selected astrocyte-enriched genes, which describe typical astrocytic features. qRT-PCR data corroborated our results, reporting an overall decrement of gene expression, which is area and age-dependent. In conclusion, our data show that Mecp2 deficiency causes structural and molecular alterations in astrocytes, which progress along with the severity of symptoms and diversely occur in the different cerebral regions, highlighting the importance of considering heterogeneity when studying astrocytes in RTT.
Collapse
Affiliation(s)
- Elena Albizzati
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Elena Florio
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Federica Miramondi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Irene Sormonta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicoletta Landsberger
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelisa Frasca
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
8
|
Li W. Excitation and Inhibition Imbalance in Rett Syndrome. Front Neurosci 2022; 16:825063. [PMID: 35250460 PMCID: PMC8894599 DOI: 10.3389/fnins.2022.825063] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
A loss of the excitation/inhibition (E/I) balance in the neural circuit has emerged as a common neuropathological feature in many neurodevelopmental disorders. Rett syndrome (RTT), a prevalent neurodevelopmental disorder that affects 1:10,000-15,000 women globally, is caused by loss-of-function mutations in the Methyl-CpG-binding Protein-2 (Mecp2) gene. E/I imbalance is recognized as the leading cellular and synaptic hallmark that is fundamental to diverse RTT neurological symptoms, including stereotypic hand movements, impaired motor coordination, breathing irregularities, seizures, and learning/memory dysfunctions. E/I balance in RTT is not homogeneously altered but demonstrates brain region and cell type specificity instead. In this review, I elaborate on the current understanding of the loss of E/I balance in a range of brain areas at molecular and cellular levels. I further describe how the underlying cellular mechanisms contribute to the disturbance of the proper E/I ratio. Last, I discuss current pharmacologic innervations for RTT and their role in modifying the E/I balance.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
9
|
Fabbri A, Bracci L. Immunomodulatory properties of CNF1 toxin from E. coli: implications for colorectal carcinogenesis. Am J Cancer Res 2022; 12:651-660. [PMID: 35261793 PMCID: PMC8899975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/27/2022] [Indexed: 06/14/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer death worldwide. The risk of developing CRC is influenced by both environmental and genetic factors. Recently, chronic inflammation and gut microbiota modifications have been associated with increased CRC risk. Escherichia coli belongs to the commensal intestinal flora and can become highly pathogenic following the acquisition of genes coding for virulence factors, such as the cytotoxic necrotizing factor type 1 (CNF1). Numerous reports highlight that, besides exerting direct effects on epithelial cells, CNF1 can also act on immune cells, modulating their responses and possibly contributing to disease development. In the present review, we summarized the key studies addressing the immunomodulatory functions of CNF1 and discussed the contribution that CNF1 can bring about to CRC through the creation of a pro-inflammatory microenvironment.
Collapse
Affiliation(s)
- Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di SanitàViale Regina Elena 299, Rome, Italy
| | - Laura Bracci
- Departement of Oncology and Molecular Medicine, Istituto Superiore di SanitàViale Regina Elena 299, Rome, Italy
| |
Collapse
|
10
|
The Cytotoxic Necrotizing Factors (CNFs)-A Family of Rho GTPase-Activating Bacterial Exotoxins. Toxins (Basel) 2021; 13:toxins13120901. [PMID: 34941738 PMCID: PMC8709095 DOI: 10.3390/toxins13120901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/17/2022] Open
Abstract
The cytotoxic necrotizing factors (CNFs) are a family of Rho GTPase-activating single-chain exotoxins that are produced by several Gram-negative pathogenic bacteria. Due to the pleiotropic activities of the targeted Rho GTPases, the CNFs trigger multiple signaling pathways and host cell processes with diverse functional consequences. They influence cytokinesis, tissue integrity, cell barriers, and cell death, as well as the induction of inflammatory and immune cell responses. This has an enormous influence on host-pathogen interactions and the severity of the infection. The present review provides a comprehensive insight into our current knowledge of the modular structure, cell entry mechanisms, and the mode of action of this class of toxins, and describes their influence on the cell, tissue/organ, and systems levels. In addition to their toxic functions, possibilities for their use as drug delivery tool and for therapeutic applications against important illnesses, including nervous system diseases and cancer, have also been identified and are discussed.
Collapse
|
11
|
Scuteri D, Rombolà L, Natoli S, Pisani A, Bonsi P, Hamamura K, Bagetta G, Tonin P, Corasaniti MT. Exploitation of Thermal Sensitivity and Hyperalgesia in a Mouse Model of Dystonia. Life (Basel) 2021; 11:life11090985. [PMID: 34575134 PMCID: PMC8468866 DOI: 10.3390/life11090985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/02/2022] Open
Abstract
Neuropathic pain is characterized by mechanical allodynia and thermal hyperalgesia to heat, and it affects some 20% of European population. Patients suffering from several neurologic diseases experience neuropathic pain, often finding no relief in therapy. Transgenic mice expressing the gene encoding the human mutant (hMT) or the human wild-type (hWT) torsin A represent a preclinical model of DYT1 dystonia which is the most common form of early-onset inherited dystonia. Baseline thermal sensitivity and hyperalgesia to heat have never been studied in models of dystonia. Therefore, the aim of this research has been to characterize thermal sensitivity in baseline conditions and hyperalgesia to heat after the induction of neuropathic pain through the spinal nerve ligation (SNL) model in mice overexpressing human wild-type and mutated torsin A in comparison to non-transgenic C57BL/6 mice. According to our results, the paw withdrawal latency time to heat in the Hargreaves’ test is significantly lower in the hMT mice (Kruskal–Wallis test = 6.933; p = 0.0312*; hMT vs. hWT p = 0.0317*). On the other hand, no significant differences in SNL-induced thermal hyperalgesia was found among the three strains (Friedman test = 4.933; p = 0.1019). Future studies are needed to better understand the role of torsin A in sensory processing of heat stimuli.
Collapse
Affiliation(s)
- Damiana Scuteri
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
- Regional Center for Serious Brain Injuries, S. Anna Institute, 88900 Crotone, Italy;
- Correspondence: (D.S.); (G.B.); Tel.: +39-0984/493462 (D.S. & G.B.)
| | - Laura Rombolà
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Silvia Natoli
- Department of Clinical Science and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.P.); (P.B.)
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Paola Bonsi
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.P.); (P.B.)
| | - Kengo Hamamura
- Laboratory of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Daiichi University of Pharmacy, Fukuoka 815-8511, Japan;
| | - Giacinto Bagetta
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
- Correspondence: (D.S.); (G.B.); Tel.: +39-0984/493462 (D.S. & G.B.)
| | - Paolo Tonin
- Regional Center for Serious Brain Injuries, S. Anna Institute, 88900 Crotone, Italy;
| | | |
Collapse
|
12
|
Mao Y, Evans EE, Mishra V, Balch L, Eberhardt A, Zauderer M, Gold WA. Anti-Semaphorin 4D Rescues Motor, Cognitive, and Respiratory Phenotypes in a Rett Syndrome Mouse Model. Int J Mol Sci 2021; 22:ijms22179465. [PMID: 34502373 PMCID: PMC8431088 DOI: 10.3390/ijms22179465] [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: 07/22/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 01/09/2023] Open
Abstract
Rett syndrome is a neurodevelopmental disorder caused by mutations of the methyl-CpG binding protein 2 gene. Abnormal physiological functions of glial cells contribute to pathogenesis of Rett syndrome. Semaphorin 4D (SEMA4D) regulates processes central to neuroinflammation and neurodegeneration including cytoskeletal structures required for process extension, communication, and migration of glial cells. Blocking SEMA4D-induced gliosis may preserve normal glial and neuronal function and rescue neurological dysfunction in Rett syndrome. We evaluated the pre-clinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody in the Rett syndrome Mecp2T158A transgenic mouse model and investigated the contribution of glial cells as a proposed mechanism of action in treated mice and in primary glial cultures isolated from Mecp2T158A/y mutant mice. SEMA4D is upregulated in neurons while glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1-positive cells are upregulated in Mecp2T158A/y mice. Anti-SEMA4D treatment ameliorates Rett syndrome-specific symptoms and improves behavioural functions in both pre-symptomatic and symptomatic cohorts of hemizygous Mecp2T158A/y male mice. Anti-SEMA4D also reduces astrocyte and microglia activation in vivo. In vitro experiments demonstrate an abnormal cytoskeletal structure in mutant astrocytes in the presence of SEMA4D, while anti-SEMA4D antibody treatment blocks SEMA4D–Plexin B1 signaling and mitigates these abnormalities. These results suggest that anti-SEMA4D immunotherapy may be an effective treatment option to alleviate symptoms and improve cognitive and motor function in Rett syndrome.
Collapse
Affiliation(s)
- Yilin Mao
- Molecular Neurobiology Research Laboratory, Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia;
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Elizabeth E. Evans
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Vikas Mishra
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Leslie Balch
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Allison Eberhardt
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Maurice Zauderer
- Vaccinex Inc., Rochester, NY 14620, USA; (E.E.E.); (V.M.); (L.B.); (A.E.); (M.Z.)
| | - Wendy A. Gold
- Molecular Neurobiology Research Laboratory, Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Westmead, NSW 2145, Australia;
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Molecular Neurobiology Research Laboratory, The Children’s Medical Research Institute, Westmead, NSW 2145, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
13
|
Urbinati C, Cosentino L, Germinario EAP, Valenti D, Vigli D, Ricceri L, Laviola G, Fiorentini C, Vacca RA, Fabbri A, De Filippis B. Treatment with the Bacterial Toxin CNF1 Selectively Rescues Cognitive and Brain Mitochondrial Deficits in a Female Mouse Model of Rett Syndrome Carrying a MeCP2-Null Mutation. Int J Mol Sci 2021; 22:6739. [PMID: 34201747 PMCID: PMC8269120 DOI: 10.3390/ijms22136739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/25/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Rett syndrome (RTT) is a rare neurological disorder caused by mutations in the X-linked MECP2 gene and a major cause of intellectual disability in females. No cure exists for RTT. We previously reported that the behavioural phenotype and brain mitochondria dysfunction are widely rescued by a single intracerebroventricular injection of the bacterial toxin CNF1 in a RTT mouse model carrying a truncating mutation of the MeCP2 gene (MeCP2-308 mice). Given the heterogeneity of MECP2 mutations in RTT patients, we tested the CNF1 therapeutic efficacy in a mouse model carrying a null mutation (MeCP2-Bird mice). CNF1 selectively rescued cognitive defects, without improving other RTT-related behavioural alterations, and restored brain mitochondrial respiratory chain complex activity in MeCP2-Bird mice. To shed light on the molecular mechanisms underlying the differential CNF1 effects on the behavioural phenotype, we compared treatment effects on relevant signalling cascades in the brain of the two RTT models. CNF1 provided a significant boost of the mTOR activation in MeCP2-308 hippocampus, which was not observed in the MeCP2-Bird model, possibly explaining the differential effects of CNF1. These results demonstrate that CNF1 efficacy depends on the mutation beared by MeCP2-mutated mice, stressing the need of testing potential therapeutic approaches across RTT models.
Collapse
Affiliation(s)
- Chiara Urbinati
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Livia Cosentino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Elena Angela Pia Germinario
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.A.P.G.); (A.F.)
| | - Daniela Valenti
- Bioenergetics and Molecular Biotechnologies, Institute of Biomembranes, National Council of Research, 70126 Bari, Italy; (D.V.); (R.A.V.)
| | - Daniele Vigli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Laura Ricceri
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Giovanni Laviola
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Carla Fiorentini
- Association for Research on Integrative Oncology Therapies (ARTOI), 00165 Rome, Italy;
| | - Rosa Anna Vacca
- Bioenergetics and Molecular Biotechnologies, Institute of Biomembranes, National Council of Research, 70126 Bari, Italy; (D.V.); (R.A.V.)
| | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.A.P.G.); (A.F.)
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| |
Collapse
|
14
|
Liaci C, Camera M, Caslini G, Rando S, Contino S, Romano V, Merlo GR. Neuronal Cytoskeleton in Intellectual Disability: From Systems Biology and Modeling to Therapeutic Opportunities. Int J Mol Sci 2021; 22:ijms22116167. [PMID: 34200511 PMCID: PMC8201358 DOI: 10.3390/ijms22116167] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Intellectual disability (ID) is a pathological condition characterized by limited intellectual functioning and adaptive behaviors. It affects 1–3% of the worldwide population, and no pharmacological therapies are currently available. More than 1000 genes have been found mutated in ID patients pointing out that, despite the common phenotype, the genetic bases are highly heterogeneous and apparently unrelated. Bibliomic analysis reveals that ID genes converge onto a few biological modules, including cytoskeleton dynamics, whose regulation depends on Rho GTPases transduction. Genetic variants exert their effects at different levels in a hierarchical arrangement, starting from the molecular level and moving toward higher levels of organization, i.e., cell compartment and functions, circuits, cognition, and behavior. Thus, cytoskeleton alterations that have an impact on cell processes such as neuronal migration, neuritogenesis, and synaptic plasticity rebound on the overall establishment of an effective network and consequently on the cognitive phenotype. Systems biology (SB) approaches are more focused on the overall interconnected network rather than on individual genes, thus encouraging the design of therapies that aim to correct common dysregulated biological processes. This review summarizes current knowledge about cytoskeleton control in neurons and its relevance for the ID pathogenesis, exploiting in silico modeling and translating the implications of those findings into biomedical research.
Collapse
Affiliation(s)
- Carla Liaci
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Mattia Camera
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Giovanni Caslini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Simona Rando
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Salvatore Contino
- Department of Engineering, University of Palermo, Viale delle Scienze Ed. 8, 90128 Palermo, Italy;
| | - Valentino Romano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze Ed. 16, 90128 Palermo, Italy;
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
- Correspondence: ; Tel.: +39-0116706449; Fax: +39-0116706432
| |
Collapse
|
15
|
Ren B, Dunaevsky A. Modeling Neurodevelopmental and Neuropsychiatric Diseases with Astrocytes Derived from Human-Induced Pluripotent Stem Cells. Int J Mol Sci 2021; 22:1692. [PMID: 33567562 PMCID: PMC7915337 DOI: 10.3390/ijms22041692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Accumulating studies demonstrate the morphological and functional diversity of astrocytes, a subtype of glial cells in the central nervous system. Animal models are instrumental in advancing our understanding of the role of astrocytes in brain development and their contribution to neurological disease; however, substantial interspecies differences exist between rodent and human astrocytes, underscoring the importance of studying human astrocytes. Human pluripotent stem cell differentiation approaches allow the study of patient-specific astrocytes in the etiology of neurological disorders. In this review, we summarize the structural and functional properties of astrocytes, including the unique features of human astrocytes; demonstrate the necessity of the stem cell platform; and discuss how this platform has been applied to the research of neurodevelopmental and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Baiyan Ren
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Department of Neurological Science, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Anna Dunaevsky
- Department of Neurological Science, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
16
|
Gomathi M, Padmapriya S, Balachandar V. Drug Studies on Rett Syndrome: From Bench to Bedside. J Autism Dev Disord 2020; 50:2740-2764. [PMID: 32016693 DOI: 10.1007/s10803-020-04381-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Drug studies on Rett syndrome (RTT) have drastically increased over the past few decades. This review aims to provide master data on bench-to-bedside drug studies involving RTT. A comprehensive literature review was performed by searching in PUBMED, MEDLINE and Google Scholar, international, national and regional clinical trial registries and pharmaceutical companies using the keywords "Rett syndrome treatment and/or drug or compound or molecule". Seventy drugs were investigated in non-clinical (N = 65 animal/cell line-based studies; N = 5 iPSC-based study) and clinical trials (N = 34) for ameliorating the symptoms of RTT. Though there is good progress in both clinical and non-clinical studies, none of these drugs entered phase III/IV for being launched as a therapeutic agent for RTT.
Collapse
Affiliation(s)
- Mohan Gomathi
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | | | - Vellingiri Balachandar
- Human Molecular Genetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India.
| |
Collapse
|
17
|
Vigli D, Cosentino L, Pellas M, De Filippis B. Chronic Treatment with Cannabidiolic Acid (CBDA) Reduces Thermal Pain Sensitivity in Male Mice and Rescues the Hyperalgesia in a Mouse Model of Rett Syndrome. Neuroscience 2020; 453:113-123. [PMID: 33010341 DOI: 10.1016/j.neuroscience.2020.09.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023]
Abstract
Rett syndrome (RTT) is a rare neurologic disorder, characterized by severe behavioural and physiological symptoms. RTT is caused by mutations in the MECP2 gene in about 95% of cases and to date no cure is available. Recent evidence suggests that non-euphoric phytocannabinoids (pCBs) extracted from Cannabis sativa may represent innovative therapeutic molecules for RTT, with the cannabinoid cannabidivarin having beneficial effects on behavioural and brain molecular alterations in RTT mouse models. The present study evaluated the potential therapeutic efficacy for RTT of cannabidiolic acid (CBDA; 0.2, 2, 20 mg/kg through intraperitoneal injections for 14 days), a pCB that has proved to be effective for the treatment of nausea and anxiety in rodents. This study demonstrates that systemic treatment with the low dose of CBDA has anti-nociceptive effects and reduces the thermal hyperalgesia in 8 month-old MeCP2-308 male mice, a validated RTT mouse model. CBDA did not affect other behavioural or molecular parameters. These results provide support to the antinociceptive effects of CBDA and stress the need for further studies aimed at clarifying the mechanisms underlying the abnormal pain perception in RTT.
Collapse
Affiliation(s)
- Daniele Vigli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Livia Cosentino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Mattia Pellas
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
18
|
Zuliani I, Urbinati C, Valenti D, Quattrini MC, Medici V, Cosentino L, Pietraforte D, Di Domenico F, Perluigi M, Vacca RA, De Filippis B. The Anti-Diabetic Drug Metformin Rescues Aberrant Mitochondrial Activity and Restrains Oxidative Stress in a Female Mouse Model of Rett Syndrome. J Clin Med 2020; 9:jcm9061669. [PMID: 32492904 PMCID: PMC7355965 DOI: 10.3390/jcm9061669] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/25/2022] Open
Abstract
Metformin is the first-line therapy for diabetes, even in children, and a promising attractive candidate for drug repurposing. Mitochondria are emerging as crucial targets of metformin action both in the periphery and in the brain. The present study evaluated whether treatment with metformin may rescue brain mitochondrial alterations and contrast the increased oxidative stress in a validated mouse model of Rett syndrome (RTT), a rare neurologic disorder of monogenic origin characterized by severe behavioral and physiological symptoms. No cure for RTT is available. In fully symptomatic RTT mice (12 months old MeCP2-308 heterozygous female mice), systemic treatment with metformin (100 mg/kg ip for 10 days) normalized the reduced mitochondrial ATP production and ATP levels in the whole-brain, reduced brain oxidative damage, and rescued the increased production of reactive oxidizing species in blood. A 10-day long treatment with metformin also boosted pathways related to mitochondrial biogenesis and antioxidant defense in the brain of metformin-treated RTT mice. This treatment regimen did not improve general health status and motor dysfunction in RTT mice at an advanced stage of the disease. Present results provide evidence that systemic treatment with metformin may represent a novel, repurposable therapeutic strategy for RTT.
Collapse
Affiliation(s)
- Ilaria Zuliani
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (F.D.D.); (M.P.)
| | - Chiara Urbinati
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (V.M.); (L.C.)
| | - Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, 70126 Bari, Italy; (D.V.); (R.A.V.)
| | | | - Vanessa Medici
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (V.M.); (L.C.)
| | - Livia Cosentino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (V.M.); (L.C.)
| | | | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (F.D.D.); (M.P.)
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy; (I.Z.); (F.D.D.); (M.P.)
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, 70126 Bari, Italy; (D.V.); (R.A.V.)
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (V.M.); (L.C.)
- Correspondence:
| |
Collapse
|
19
|
Travaglione S, Loizzo S, Vona R, Ballan G, Rivabene R, Giordani D, Guidotti M, Dupuis ML, Maroccia Z, Baiula M, Rimondini R, Campana G, Fiorentini C. The Bacterial Toxin CNF1 Protects Human Neuroblastoma SH-SY5Y Cells against 6-Hydroxydopamine-Induced Cell Damage: The Hypothesis of CNF1-Promoted Autophagy as an Antioxidant Strategy. Int J Mol Sci 2020; 21:ijms21093390. [PMID: 32403292 PMCID: PMC7247702 DOI: 10.3390/ijms21093390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/29/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
Several chronic neuroinflammatory diseases, including Parkinson’s disease (PD), have the so-called ‘redox imbalance’ in common, a dynamic system modulated by various factors. Among them, alteration of the mitochondrial functionality can cause overproduction of reactive oxygen species (ROS) with the consequent induction of oxidative DNA damage and apoptosis. Considering the failure of clinical trials with drugs that eliminate ROS directly, research currently focuses on approaches that counteract redox imbalance, thus restoring normal physiology in a neuroinflammatory condition. Herein, we used SH-SY5Y cells treated with 6-hydroxydopamine (6-OHDA), a neurotoxin broadly employed to generate experimental models of PD. Cells were pre-treated with the Rho-modulating Escherichia coli cytotoxic necrotizing factor 1 (CNF1), before the addition of 6-OHDA. Then, cell viability, mitochondrial morphology and dynamics, redox profile as well as autophagic markers expression were assessed. We found that CNF1 preserves cell viability and counteracts oxidative stress induced by 6-OHDA. These effects are accompanied by modulation of the mitochondrial network and an increase in macroautophagic markers. Our results confirm the Rho GTPases as suitable pharmacological targets to counteract neuroinflammatory diseases and evidence the potentiality of CNF1, whose beneficial effects on pathological animal models have been already proven to act against oxidative stress through an autophagic strategy.
Collapse
Affiliation(s)
- Sara Travaglione
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
- Correspondence: ; Tel.: +39-06-49903692
| | - Stefano Loizzo
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Rosa Vona
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Giulia Ballan
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Roberto Rivabene
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Danila Giordani
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Marco Guidotti
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Maria Luisa Dupuis
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Zaira Maroccia
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
| | - Monica Baiula
- University of Bologna, 40126 Bologna, Italy; (M.B.); (R.Rim); (G.C.)
| | - Roberto Rimondini
- University of Bologna, 40126 Bologna, Italy; (M.B.); (R.Rim); (G.C.)
| | - Gabriele Campana
- University of Bologna, 40126 Bologna, Italy; (M.B.); (R.Rim); (G.C.)
| | - Carla Fiorentini
- Istituto Superiore di Sanità, 00161 Rome, Italy; (S.L.); (R.V.); (G.B.); (R.Riv); (D.G.); (M.G.); (M.L.D.); (Z.M.); or
- Association for Research on Integrative Oncology Therapies (ARTOI), 00165 Rome, Italy
| |
Collapse
|
20
|
Colarusso A, Maroccia Z, Parrilli E, Germinario EAP, Fortuna A, Loizzo S, Ricceri L, Tutino ML, Fiorentini C, Fabbri A. Cnf1 Variants Endowed with the Ability to Cross the Blood-Brain Barrier: A New Potential Therapeutic Strategy for Glioblastoma. Toxins (Basel) 2020; 12:toxins12050291. [PMID: 32375387 PMCID: PMC7290510 DOI: 10.3390/toxins12050291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023] Open
Abstract
Among gliomas, primary tumors originating from glial cells, glioblastoma (GBM) identified as WHO grade IV glioma, is the most common and aggressive malignant brain tumor. We have previously shown that the Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1) is remarkably effective as an anti-neoplastic agent in a mouse model of glioma, reducing the tumor volume, increasing survival, and maintaining the functional properties of peritumoral neurons. However, being unable to cross the blood–brain barrier (BBB), CNF1 requires injection directly into the brain, which is a very invasive administration route. Thus, to overcome this pitfall, we designed a CNF1 variant characterized by the presence of an N-terminal BBB-crossing tag. The variant was produced and we verified whether its activity was comparable to that of wild-type CNF1 in GBM cells. We investigated the signaling pathways engaged in the cell response to CNF1 variants to provide preliminary data to the subsequent studies in experimental animals. CNF1 may represent a novel avenue for GBM therapy, particularly because, besides blocking tumor growth, it also preserves the healthy surrounding tissue, maintaining its architecture and functionality. This renders CNF1 the most interesting candidate for the treatment of brain tumors, among other potentially effective bacterial toxins.
Collapse
Affiliation(s)
- Andrea Colarusso
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario M. S. Angelo, Via Cintia, 80126 Napoli, Italy; (A.C.); (E.P.); (M.L.T.)
| | - Zaira Maroccia
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (Z.M.); (E.A.P.G.); (A.F.); (S.L.); (L.R.); (C.F.)
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario M. S. Angelo, Via Cintia, 80126 Napoli, Italy; (A.C.); (E.P.); (M.L.T.)
| | - Elena Angela Pia Germinario
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (Z.M.); (E.A.P.G.); (A.F.); (S.L.); (L.R.); (C.F.)
| | - Andrea Fortuna
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (Z.M.); (E.A.P.G.); (A.F.); (S.L.); (L.R.); (C.F.)
| | - Stefano Loizzo
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (Z.M.); (E.A.P.G.); (A.F.); (S.L.); (L.R.); (C.F.)
| | - Laura Ricceri
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (Z.M.); (E.A.P.G.); (A.F.); (S.L.); (L.R.); (C.F.)
| | - Maria Luisa Tutino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario M. S. Angelo, Via Cintia, 80126 Napoli, Italy; (A.C.); (E.P.); (M.L.T.)
| | - Carla Fiorentini
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (Z.M.); (E.A.P.G.); (A.F.); (S.L.); (L.R.); (C.F.)
- Association for Research on Integrative Oncological Therapies (ARTOI), 00165 Rome, Italy
| | - Alessia Fabbri
- Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (Z.M.); (E.A.P.G.); (A.F.); (S.L.); (L.R.); (C.F.)
- Correspondence: ; Tel.: +39-06-4990-2939
| |
Collapse
|
21
|
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
|
22
|
Aldosary M, Al-Bakheet A, Al-Dhalaan H, Almass R, Alsagob M, Al-Younes B, AlQuait L, Mustafa OM, Bulbul M, Rahbeeni Z, Alfadhel M, Chedrawi A, Al-Hassnan Z, AlDosari M, Al-Zaidan H, Al-Muhaizea MA, AlSayed MD, Salih MA, AlShammari M, Faiyaz-Ul-Haque M, Chishti MA, Al-Harazi O, Al-Odaib A, Kaya N, Colak D. Rett Syndrome, a Neurodevelopmental Disorder, Whole-Transcriptome, and Mitochondrial Genome Multiomics Analyses Identify Novel Variations and Disease Pathways. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:160-171. [PMID: 32105570 DOI: 10.1089/omi.2019.0192] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder reported worldwide in diverse populations. RTT is diagnosed primarily in females, with clinical findings manifesting early in life. Despite the variable rates across populations, RTT has an estimated prevalence of ∼1 in 10,000 live female births. Among 215 Saudi Arabian patients with neurodevelopmental and autism spectrum disorders, we identified 33 patients with RTT who were subsequently examined by genome-wide transcriptome and mitochondrial genome variations. To the best of our knowledge, this is the first in-depth molecular and multiomics analyses of a large cohort of Saudi RTT cases with a view to informing the underlying mechanisms of this disease that impact many patients and families worldwide. The patients were unrelated, except for 2 affected sisters, and comprised of 25 classic and eight atypical RTT cases. The cases were screened for methyl-CpG binding protein 2 (MECP2), CDKL5, FOXG1, NTNG1, and mitochondrial DNA (mtDNA) variants, as well as copy number variations (CNVs) using a genome-wide experimental strategy. We found that 15 patients (13 classic and two atypical RTT) have MECP2 mutations, 2 of which were novel variants. Two patients had novel FOXG1 and CDKL5 variants (both atypical RTT). Whole mtDNA sequencing of the patients who were MECP2 negative revealed two novel mtDNA variants in two classic RTT patients. Importantly, the whole-transcriptome analysis of our RTT patients' blood and further comparison with previous expression profiling of brain tissue from patients with RTT revealed 77 significantly dysregulated genes. The gene ontology and interaction network analysis indicated potentially critical roles of MAPK9, NDUFA5, ATR, SMARCA5, RPL23, SRSF3, and mitochondrial dysfunction, oxidative stress response and MAPK signaling pathways in the pathogenesis of RTT genes. This study expands our knowledge on RTT disease networks and pathways as well as presents novel mutations and mtDNA alterations in RTT in a population sample that was not previously studied.
Collapse
Affiliation(s)
- Mazhor Aldosary
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - AlBandary Al-Bakheet
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Hesham Al-Dhalaan
- Department of Neuroscience, and King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Rawan Almass
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Maysoon Alsagob
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Banan Al-Younes
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Laila AlQuait
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Osama Mufid Mustafa
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mustafa Bulbul
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Zuhair Rahbeeni
- Department of Medical Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Majid Alfadhel
- King Abdullah International Medical Research Centre, King Saud bin Abdulaziz University for Health Sciences, Genetics Division, Department of Pediatrics, King Abdullah Specialized Children Hospital, Riyadh, Saudi Arabia
| | - Aziza Chedrawi
- Department of Neuroscience, and King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Zuhair Al-Hassnan
- Department of Medical Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mohammed AlDosari
- Center for Pediatric Neurosciences, Cleveland Clinic, Cleveland, Ohio
| | - Hamad Al-Zaidan
- Department of Medical Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mohammad A Al-Muhaizea
- Department of Neuroscience, and King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Moeenaldeen D AlSayed
- Department of Medical Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mustafa A Salih
- Division of Pediatric Neurology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mai AlShammari
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | | | - Mohammad Azhar Chishti
- Department of Biochemistry, King Khalid Hospital, King Saud University, Riyadh, Saudi Arabia
| | - Olfat Al-Harazi
- Department of Biostatistics, Epidemiology, and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ali Al-Odaib
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Namik Kaya
- Department of Genetics, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Dilek Colak
- Department of Biostatistics, Epidemiology, and Scientific Computing, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
23
|
Pillet LE, Cresto N, Saillour Y, Ghézali G, Bemelmans AP, Livet J, Bienvenu T, Rouach N, Billuart P. The intellectual disability protein Oligophrenin-1 controls astrocyte morphology and migration. Glia 2020; 68:1729-1742. [PMID: 32073702 DOI: 10.1002/glia.23801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023]
Abstract
Astrocytes are involved in several aspects of neuronal development and properties which are altered in intellectual disability (ID). Oligophrenin-1 is a RhoGAP protein implicated in actin cytoskeleton regulation, and whose mutations are associated with X-linked ID. Oligophrenin-1 is expressed in neurons, where its functions have been widely reported at the synapse, as well as in glial cells. However, its roles in astrocytes are still largely unexplored. Using in vitro and in vivo models of oligophrenin1 disruption in astrocytes, we found that oligophrenin1 regulates at the molecular level the RhoA/ROCK/MLC2 pathway in astroglial cells. We also showed at the cellular level that oligophrenin1 modulates astrocyte morphology and migration both in vitro and in vivo, and is involved in glial scar formation. Altogether, these data suggest that oligophrenin1 deficiency alters not only neuronal but also astrocytic functions, which might contribute to the development of ID.
Collapse
Affiliation(s)
- Laure-Elise Pillet
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France.,Doctoral School N°562, Paris Descartes University, Paris, France.,Institut Cochin, INSERM UMR 1016, CNRS UMR 8104, Université de Paris, Paris, France
| | - Noémie Cresto
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Yoann Saillour
- Institut Cochin, INSERM UMR 1016, CNRS UMR 8104, Université de Paris, Paris, France
| | - Grégory Ghézali
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Alexis-Pierre Bemelmans
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale, Institut de biologie François Jacob, MIRCen, and CNRS UMR 9199, Université Paris-Sud, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Jean Livet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Thierry Bienvenu
- Institut Cochin, INSERM UMR 1016, CNRS UMR 8104, Université de Paris, Paris, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, France
| | - Pierre Billuart
- Institut Cochin, INSERM UMR 1016, CNRS UMR 8104, Université de Paris, Paris, France
| |
Collapse
|
24
|
Kadam SD, Sullivan BJ, Goyal A, Blue ME, Smith-Hicks C. Rett Syndrome and CDKL5 Deficiency Disorder: From Bench to Clinic. Int J Mol Sci 2019; 20:ijms20205098. [PMID: 31618813 PMCID: PMC6834180 DOI: 10.3390/ijms20205098] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022] Open
Abstract
Rett syndrome (RTT) and CDKL5 deficiency disorder (CDD) are two rare X-linked developmental brain disorders with overlapping but distinct phenotypic features. This review examines the impact of loss of methyl-CpG-binding protein 2 (MeCP2) and cyclin-dependent kinase-like 5 (CDKL5) on clinical phenotype, deficits in synaptic- and circuit-homeostatic mechanisms, seizures, and sleep. In particular, we compare the overlapping and contrasting features between RTT and CDD in clinic and in preclinical studies. Finally, we discuss lessons learned from recent clinical trials while reviewing the findings from pre-clinical studies.
Collapse
Affiliation(s)
- Shilpa D Kadam
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Brennan J Sullivan
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
| | - Archita Goyal
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
| | - Mary E Blue
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Constance Smith-Hicks
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, MD 21205, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
25
|
Kahanovitch U, Patterson KC, Hernandez R, Olsen ML. Glial Dysfunction in MeCP2 Deficiency Models: Implications for Rett Syndrome. Int J Mol Sci 2019; 20:ijms20153813. [PMID: 31387202 PMCID: PMC6696322 DOI: 10.3390/ijms20153813] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023] Open
Abstract
Rett syndrome (RTT) is a rare, X-linked neurodevelopmental disorder typically affecting females, resulting in a range of symptoms including autistic features, intellectual impairment, motor deterioration, and autonomic abnormalities. RTT is primarily caused by the genetic mutation of the Mecp2 gene. Initially considered a neuronal disease, recent research shows that glial dysfunction contributes to the RTT disease phenotype. In the following manuscript, we review the evidence regarding glial dysfunction and its effects on disease etiology.
Collapse
Affiliation(s)
- Uri Kahanovitch
- School of Neuroscience, Virginia Polytechnic and State University, Life Sciences I Building Room 212, 970 Washington St. SW, Blacksburg, VA 24061, USA
| | - Kelsey C Patterson
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd., Birmingham, AL 35294, USA
| | - Raymundo Hernandez
- School of Neuroscience, Virginia Polytechnic and State University, Life Sciences I Building Room 212, 970 Washington St. SW, Blacksburg, VA 24061, USA
- Graduate Program in Translational Biology Medicine and Health, Virginia Tech, Roanoke, VL 24014, USA
| | - Michelle L Olsen
- School of Neuroscience, Virginia Polytechnic and State University, Life Sciences I Building Room 212, 970 Washington St. SW, Blacksburg, VA 24061, USA.
| |
Collapse
|
26
|
Matteucci A, Ricceri L, Fabbri A, Fortuna A, Travaglione S, Guidotti M, Martinelli A, Villa M, Pricci F, Maroccia Z, Campana G, Malchiodi-Albedi F, Fiorentini C, Loizzo S. Eye Drop Instillation of the Rac1 Modulator CNF1 Attenuates Retinal Gliosis and Ameliorates Visual Performance in a Rat Model of Hypertensive Retinopathy. Neuroscience 2019; 411:119-129. [PMID: 31128161 DOI: 10.1016/j.neuroscience.2019.05.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022]
Abstract
In hypertensive retinopathy, the retinal damage due to high blood pressure is accompanied by increased expression of Glial Fibrillary Acidic Protein (GFAP), which indicates a role of neuroinflammatory processes in such a retinopathy. Proteins belonging to the Rho GTPase family, particularly Rac1, are involved in the activation of Müller glia and in the progression of photoreceptor degeneration, and may thus represent a novel candidate for therapeutic intervention following central nervous system inflammation. In this paper, we have observed that topical administration as eye drops of Cytotoxic Necrotizing Factor 1 (CNF1), a Rho GTPase modulator, surprisingly improves electrophysiological and behavioral visual performances in aged spontaneously hypertensive rats. Furthermore, such functional improvement is accompanied by a reduction of Rac1 activity and retinal GFAP expression. Our results suggest that Rac1 inhibition through CNF1 topical administration may represent a new strategy to target retinal gliosis.
Collapse
Affiliation(s)
- Andrea Matteucci
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Laura Ricceri
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessia Fabbri
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Andrea Fortuna
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Sara Travaglione
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marco Guidotti
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Andrea Martinelli
- National Centre for Animal Research and Welfare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marika Villa
- Department of Cardiovascular, Dysmetabolic and Endocrine-Metabolic Diseases, and Ageing, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Flavia Pricci
- Department of Cardiovascular, Dysmetabolic and Endocrine-Metabolic Diseases, and Ageing, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Zaira Maroccia
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Gabriele Campana
- Department of Pharmacy and Biotechnology, University of Bologna, Via Zamboni, 33, Bologna, Italy
| | - Fiorella Malchiodi-Albedi
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Carla Fiorentini
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy; Association for Research on Integrative Oncology Therapies (ARTOI), Rome, Italy
| | - Stefano Loizzo
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
27
|
The bacterial protein CNF1 as a new strategy against Plasmodium falciparum cytoadherence. PLoS One 2019; 14:e0213529. [PMID: 30845261 PMCID: PMC6405130 DOI: 10.1371/journal.pone.0213529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/22/2019] [Indexed: 11/19/2022] Open
Abstract
Plasmodium falciparum severe malaria causes more than 400,000 deaths every year. One feature of P. falciparum-parasitized erythrocytes (pRBC) leading to cerebral malaria (CM), the most dangerous form of severe malaria, is cytoadherence to endothelium and blockage of the brain microvasculature. Preventing ligand-receptor interactions involved in this process could inhibit pRBC sequestration and insurgence of severe disease whilst reversing existing cytoadherence could be a saving life adjunct therapy. Increasing evidence indicate the endothelial Rho signaling as a crucial player in malaria parasite cytoadherence. Therefore, we have used the cytotoxic necrotizing factor 1 (CNF1), an Escherichia coli protein able to modulate the activity of Cdc42, Rac, and Rho, three subfamilies of the Rho GTPases family, to study interactions between infected erythrocytes and cerebral endothelium in co-culture models. The main results are that CNF1 not only prevents cytoadherence but, more importantly, induces the detachment of pRBCs from endothelia monolayers. We first observed that CNF1 does affect neither parasite growth, nor the morphology and concentration of knobs that characterize the parasitized erythrocyte surface, as viewed by scanning electron microscopy. On the other hand, flow cytometry experiments show that cytoadherence reversion induced by CNF1 occurs in parallel with a decreased ICAM-1 receptor expression on the cell surface, suggesting the involvement of a toxin-promoted endocytic activity in such a response. Furthermore, since the endothelial barrier functionality is compromised by P. falciparum, we conducted a permeability assay on endothelial cells, revealing the CNF1 capacity to restore the brain endothelial barrier integrity. Then, using pull-down assays and inhibitory studies, we demonstrated, for the first time, that CNF1 is able not only to prevent but also to cause the parasite detachment by simultaneously activating Rho, Rac and Cdc42 in endothelial cells. All in all our findings indicate that CNF1 may represent a potential novel therapeutic strategy for preventing neurological complications of CM.
Collapse
|
28
|
Treating Rett syndrome: from mouse models to human therapies. Mamm Genome 2019; 30:90-110. [PMID: 30820643 PMCID: PMC6606665 DOI: 10.1007/s00335-019-09793-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/09/2019] [Indexed: 02/06/2023]
Abstract
Rare diseases are very difficult to study mechanistically and to develop therapies for because of the scarcity of patients. Here, the rare neuro-metabolic disorder Rett syndrome (RTT) is discussed as a prototype for precision medicine, demonstrating how mouse models have led to an understanding of the development of symptoms. RTT is caused by mutations in the X-linked gene methyl-CpG-binding protein 2 (MECP2). Mecp2-mutant mice are being used in preclinical studies that target the MECP2 gene directly, or its downstream pathways. Importantly, this work may improve the health of RTT patients. Clinical presentation may vary widely among individuals based on their mutation, but also because of the degree of X chromosome inactivation and the presence of modifier genes. Because it is a complex disorder involving many organ systems, it is likely that recovery of RTT patients will involve a combination of treatments. Precision medicine is warranted to provide the best efficacy to individually treat RTT patients.
Collapse
|
29
|
Ramsey J, Martin EC, Purcell OM, Lee KM, MacLean AG. Self-injurious behaviours in rhesus macaques: Potential glial mechanisms. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2018; 62:1008-1017. [PMID: 30450801 PMCID: PMC6385863 DOI: 10.1111/jir.12558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/20/2018] [Accepted: 09/28/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Self-injurious behaviour (SIB) can be classified as intentional, direct injuring of body tissue usually without suicidal intent. In its non-suicidal form it is commonly seen as a clinical sign of borderline personality disorder, autism, PTSD, depression, and anxiety affecting a wide range of ages and conditions. In rhesus macaques SIB is most commonly manifested through hair plucking, self-biting, self-hitting, and head banging. SIB in the form of self-biting is observed in approximately 5-15% of individually housed monkeys. Recently, glial cells are becoming recognised as key players in regulating behaviours. METHOD The goal of this study was to determine the role of glial activation, including astrocytes, in macaques that had displayed SIB. To this end, we performed immunohistochemistry and next generation sequence of brain tissues from rhesus macaques with SIB. RESULTS Our studies showed increased vimentin, but not nestin, expression on astrocytes of macaques displaying SIB. Initial RNA Seq analyses indicate activation of pathways involved in tissue remodelling, neuroinflammation and cAMP signalling. CONCLUSIONS Glia are most probably activated in primates with self-injury, and are therefore potential novel targets for therapeutics.
Collapse
Affiliation(s)
- Joseph Ramsey
- Tulane Program in Neuroscience, Tulane University, New Orleans, LA 70112
| | - Elizabeth C. Martin
- Center for Stem Cell Research and Regenerative Medicine, School of Medicine, Tulane University, New Orleans, LA 70112
| | - Olivia M. Purcell
- Tulane Program in Neuroscience, Tulane University, New Orleans, LA 70112
| | - Kim M. Lee
- Tulane National Primate Research Center, Covington, LA 70433
- Tulane Program in Biomedical Science, Tulane Medical School, New Orleans, LA 70112
| | - Andrew G. MacLean
- Tulane Program in Neuroscience, Tulane University, New Orleans, LA 70112
- Tulane National Primate Research Center, Covington, LA 70433
- Tulane Program in Biomedical Science, Tulane Medical School, New Orleans, LA 70112
- Department of Microbiology & Immunology, Tulane Medical School, New Orleans, LA 70112
- Tulane Center for Aging, Tulane University New Orleans, LA 70112
| |
Collapse
|
30
|
Vigli D, Cosentino L, Raggi C, Laviola G, Woolley-Roberts M, De Filippis B. Chronic treatment with the phytocannabinoid Cannabidivarin (CBDV) rescues behavioural alterations and brain atrophy in a mouse model of Rett syndrome. Neuropharmacology 2018; 140:121-129. [DOI: 10.1016/j.neuropharm.2018.07.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/30/2018] [Accepted: 07/24/2018] [Indexed: 12/26/2022]
|
31
|
Fabbri A, Travaglione S, Maroccia Z, Guidotti M, Pierri CL, Primiano G, Servidei S, Loizzo S, Fiorentini C. The Bacterial Protein CNF1 as a Potential Therapeutic Strategy against Mitochondrial Diseases: A Pilot Study. Int J Mol Sci 2018; 19:E1825. [PMID: 29933571 PMCID: PMC6073533 DOI: 10.3390/ijms19071825] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 06/18/2018] [Indexed: 02/07/2023] Open
Abstract
The Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1), which acts on the Rho GTPases that are key regulators of the actin cytoskeleton, is emerging as a potential therapeutic tool against certain neurological diseases characterized by cellular energy homeostasis impairment. In this brief communication, we show explorative results on the toxin’s effect on fibroblasts derived from a patient affected by myoclonic epilepsy with ragged-red fibers (MERRF) that carries a mutation in the m.8344A>G gene of mitochondrial DNA. We found that, in the patient’s cells, besides rescuing the wild-type-like mitochondrial morphology, CNF1 administration is able to trigger a significant increase in cellular content of ATP and of the mitochondrial outer membrane marker Tom20. These results were accompanied by a profound F-actin reorganization in MERRF fibroblasts, which is a typical CNF1-induced effect on cell cytoskeleton. These results point at a possible role of the actin organization in preventing or limiting the cell damage due to mitochondrial impairment and at CNF1 treatment as a possible novel strategy against mitochondrial diseases still without cure.
Collapse
Affiliation(s)
- Alessia Fabbri
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Sara Travaglione
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Zaira Maroccia
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Marco Guidotti
- Department of Veterinary Public Health and Food Safety, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Ciro Leonardo Pierri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, via Orabona, 4, 70124 Bari, Italy.
| | - Guido Primiano
- Unità di Neurofisiopatologia, Area Neuroscienze, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168 Rome, Italy.
| | - Serenella Servidei
- Unità di Neurofisiopatologia, Area Neuroscienze, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli, 8, 00168 Rome, Italy.
| | - Stefano Loizzo
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| | - Carla Fiorentini
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy.
| |
Collapse
|
32
|
Rho GTPases in Intellectual Disability: From Genetics to Therapeutic Opportunities. Int J Mol Sci 2018; 19:ijms19061821. [PMID: 29925821 PMCID: PMC6032284 DOI: 10.3390/ijms19061821] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/14/2018] [Accepted: 06/16/2018] [Indexed: 12/22/2022] Open
Abstract
Rho-class small GTPases are implicated in basic cellular processes at nearly all brain developmental steps, from neurogenesis and migration to axon guidance and synaptic plasticity. GTPases are key signal transducing enzymes that link extracellular cues to the neuronal responses required for the construction of neuronal networks, as well as for synaptic function and plasticity. Rho GTPases are highly regulated by a complex set of activating (GEFs) and inactivating (GAPs) partners, via protein:protein interactions (PPI). Misregulated RhoA, Rac1/Rac3 and cdc42 activity has been linked with intellectual disability (ID) and other neurodevelopmental conditions that comprise ID. All genetic evidences indicate that in these disorders the RhoA pathway is hyperactive while the Rac1 and cdc42 pathways are consistently hypoactive. Adopting cultured neurons for in vitro testing and specific animal models of ID for in vivo examination, the endophenotypes associated with these conditions are emerging and include altered neuronal networking, unbalanced excitation/inhibition and altered synaptic activity and plasticity. As we approach a clearer definition of these phenotype(s) and the role of hyper- and hypo-active GTPases in the construction of neuronal networks, there is an increasing possibility that selective inhibitors and activators might be designed via PPI, or identified by screening, that counteract the misregulation of small GTPases and result in alleviation of the cognitive condition. Here we review all knowledge in support of this possibility.
Collapse
|
33
|
Tantillo E, Colistra A, Vannini E, Cerri C, Pancrazi L, Baroncelli L, Costa M, Caleo M. Bacterial Toxins and Targeted Brain Therapy: New Insights from Cytotoxic Necrotizing Factor 1 (CNF1). Int J Mol Sci 2018; 19:ijms19061632. [PMID: 29857515 PMCID: PMC6032336 DOI: 10.3390/ijms19061632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/07/2018] [Accepted: 05/12/2018] [Indexed: 01/17/2023] Open
Abstract
Pathogenic bacteria produce toxins to promote host invasion and, therefore, their survival. The extreme potency and specificity of these toxins confer to this category of proteins an exceptionally strong potential for therapeutic exploitation. In this review, we deal with cytotoxic necrotizing factor (CNF1), a cytotoxin produced by Escherichia coli affecting fundamental cellular processes, including cytoskeletal dynamics, cell cycle progression, transcriptional regulation, cell survival and migration. First, we provide an overview of the mechanisms of action of CNF1 in target cells. Next, we focus on the potential use of CNF1 as a pharmacological treatment in central nervous system’s diseases. CNF1 appears to impact neuronal morphology, physiology, and plasticity and displays an antineoplastic activity on brain tumors. The ability to preserve neural functionality and, at the same time, to trigger senescence and death of proliferating glioma cells, makes CNF1 an encouraging new strategy for the treatment of brain tumors.
Collapse
Affiliation(s)
- Elena Tantillo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Fondazione Pisana per la Scienza Onlus (FPS), via Ferruccio Giovannini 13, San Giuliano Terme, 56017 Pisa, Italy.
| | - Antonella Colistra
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Departement of Biology, University of Pisa, via Luca Ghini 13, 56126 Pisa, Italy.
| | - Eleonora Vannini
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Chiara Cerri
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
- Fondazione Umberto Veronesi, Piazza Velasca 5, 20122 Milano, Italy.
| | - Laura Pancrazi
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Laura Baroncelli
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Mario Costa
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Matteo Caleo
- CNR Neuroscience Institute, via G. Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
34
|
Maroccia Z, Loizzo S, Travaglione S, Frank C, Fabbri A, Fiorentini C. New therapeutics from Nature: The odd case of the bacterial cytotoxic necrotizing factor 1. Biomed Pharmacother 2018; 101:929-937. [PMID: 29635902 DOI: 10.1016/j.biopha.2018.02.140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 02/19/2018] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
Natural products may represent a rich source of new drugs. The enthusiasm toward this topic has recently been fueled by the 2015 Nobel Prize in Physiology or Medicine, awarded for the discovery of avermectin and artemisinin, natural products from Bacteria and Plantae, respectively, which have targeted one of the major global health issues, the parasitic diseases. Specifically, bacteria either living in the environment or colonizing our body may produce compounds of unexpected biomedical value with the potentiality to be employed as therapeutic drugs. In this review, the fascinating history of CNF1, a protein toxin produced by pathogenic strains of Escherichia coli, is divulged. Even if produced by bacteria responsible for a variety of diseases, CNF1 can behave as a promising benefactor to mankind. By modulating the Rho GTPases, this bacterial product plays a key role in organizing the actin cytoskeleton, enhancing synaptic plasticity and brain energy level, rescuing cognitive deficits, reducing glioma growth in experimental animals. These abilities strongly suggest the need to proceed with the studies on this odd drug in order to pave the way toward clinical trials.
Collapse
Affiliation(s)
- Zaira Maroccia
- Italian Centre for Global Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161 Rome, Italy
| | - Stefano Loizzo
- Italian Centre for Global Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161 Rome, Italy
| | - Sara Travaglione
- Italian Centre for Global Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161 Rome, Italy
| | - Claudio Frank
- Italian Centre for Rare Diseases, Istituto Superiore di Sanità, viale Regina Elena 299, 00161 Rome, Italy
| | - Alessia Fabbri
- Italian Centre for Global Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161 Rome, Italy
| | - Carla Fiorentini
- Italian Centre for Global Health, Istituto Superiore di Sanità, viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
35
|
Colarusso A, Caterino M, Fabbri A, Fiorentini C, Vergara A, Sica F, Parrilli E, Tutino ML. High yield purification and first structural characterization of the full-length bacterial toxin CNF1. Biotechnol Prog 2017; 34:150-159. [PMID: 29063721 DOI: 10.1002/btpr.2574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/12/2017] [Indexed: 11/10/2022]
Abstract
The Cytotoxic Necrotizing Factor 1 (CNF1) is a bacterial toxin secreted by certain Escherichia coli strains causing severe pathologies, making it a protein of pivotal interest in toxicology. In parallel, the CNF1 capability to influence important neuronal processes, like neuronal arborization, astrocytic support, and efficient ATP production, has been efficiently used in the treatment of neurological diseases, making it a promising candidate for therapy. Nonetheless, there are still some unsolved issues about the CNF1 mechanism of action and structuration probably caused by the difficulty to achieve sufficient amounts of the full-length protein for further studies. Here, we propose an efficient strategy for the production and purification of this toxin as a his-tagged recombinant protein from E. coli extracts (CNF1-H8). CNF1-H8 was expressed at the low temperature of 15°C to diminish its characteristic degradation. Then, its purification was achieved using an immobilized metal affinity chromatography (IMAC) and a size exclusion chromatography so as to collect up to 8 mg of protein per liter of culture in a highly pure form. Routine dynamic light scattering (DLS) experiments showed that the recombinant protein preparations were homogeneous and preserved this state for a long time. Furthermore, CNF1-H8 functionality was confirmed by testing its activity on purified RhoA and on HEp-2 cultured cells. Finally, a first structural characterization of the full-length toxin in terms of secondary structure and thermal stability was performed by circular dichroism (CD). These studies demonstrate that our system can be used to produce high quantities of pure recombinant protein for a detailed structural analysis. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 34:150-159, 2018.
Collapse
Affiliation(s)
- Andrea Colarusso
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| | - Marco Caterino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| | - Alessia Fabbri
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Roma, 00161, Italy
| | - Carla Fiorentini
- Italian Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Roma, 00161, Italy
| | - Alessandro Vergara
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy.,CEINGE Biotecnologie Avanzate scarl, Via G. Salvatore, Napoli, 80100, Italy.,Institute of Biostructures and Biomaging, CNR, Napoli, Italia Via Mezzocannone 16, Napoli, 80134, Italy
| | - Filomena Sica
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy.,Institute of Biostructures and Biomaging, CNR, Napoli, Italia Via Mezzocannone 16, Napoli, 80134, Italy
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| | - Maria Luisa Tutino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Via Cinthia, Napoli, 80126, Italy
| |
Collapse
|
36
|
Jin XR, Chen XS, Xiao L. MeCP2 Deficiency in Neuroglia: New Progress in the Pathogenesis of Rett Syndrome. Front Mol Neurosci 2017; 10:316. [PMID: 29046627 PMCID: PMC5632713 DOI: 10.3389/fnmol.2017.00316] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 09/19/2017] [Indexed: 01/24/2023] Open
Abstract
Rett syndrome (RTT) is an X-linked neurodevelopmental disease predominantly caused by mutations of the methyl-CpG-binding protein 2 (MeCP2) gene. Generally, RTT has been attributed to neuron-centric dysfunction. However, increasing evidence has shown that glial abnormalities are also involved in the pathogenesis of RTT. Mice that are MeCP2-null specifically in glial cells showed similar behavioral and/or neuronal abnormalities as those found in MeCP2-null mice, a mouse model of RTT. MeCP2 deficiency in astrocytes impacts the expression of glial intermediate filament proteins such as fibrillary acidic protein (GFAP) and S100 and induces neuron toxicity by disturbing glutamate metabolism or enhancing microtubule instability. MeCP2 deficiency in oligodendrocytes (OLs) results in down-regulation of myelin gene expression and impacts myelination. While MeCP2-deficient microglia cells fail in response to environmental stimuli, release excessive glutamate, and aggravate impairment of the neuronal circuit. In this review, we mainly focus on the progress in determining the role of MeCP2 in glial cells involved in RTT, which may provide further insight into a therapeutic intervention for RTT.
Collapse
Affiliation(s)
- Xu-Rui Jin
- Department of Histology and Embryology, Faculty of Basic Medicine, Collaborative Program for Brain Research, Third Military Medical University, Chongqing, China.,The Cadet Brigade of Clinic Medicine, Third Military Medical University, Chongqing, China
| | - Xing-Shu Chen
- Department of Histology and Embryology, Faculty of Basic Medicine, Collaborative Program for Brain Research, Third Military Medical University, Chongqing, China
| | - Lan Xiao
- Department of Histology and Embryology, Faculty of Basic Medicine, Collaborative Program for Brain Research, Third Military Medical University, Chongqing, China
| |
Collapse
|
37
|
Niebert S, van Belle GJ, Vogelgesang S, Manzke T, Niebert M. The Serotonin Receptor Subtype 5b Specifically Interacts with Serotonin Receptor Subtype 1A. Front Mol Neurosci 2017; 10:299. [PMID: 28983239 PMCID: PMC5613149 DOI: 10.3389/fnmol.2017.00299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/06/2017] [Indexed: 11/13/2022] Open
Abstract
Previously, we described the dysregulation of serotonin (5-HT) receptor subtype 5b (5-ht5b) in a mouse model of Rett syndrome (RTT). 5-ht5b has not been extensively studied, so we set out to characterize it in more detail. Unlike common cell surface receptors, 5-ht5b displays no membrane expression, while receptor clusters are located in endosomes. This unusual subcellular localization is at least in part controlled by glycosylation of the N-terminus, with 5-ht5b possessing fewer glycosylation sites than related receptors. We analyzed whether the localization to endosomes has any functional relevance and found that 5-ht5b receptors can specifically interact with 5-HT1A receptors and retain them in endosomal compartments. This interaction reduces 5-HT1A surface expression and is mediated by interactions between the fourth and fifth trans-membrane domain (TMD). This possibly represents a mechanism by which 5-ht5b receptors regulate the activity of other 5-HT receptor.
Collapse
Affiliation(s)
- Sabine Niebert
- Department of Maxillofacial Surgery, University Medical CenterGöttingen, Germany
| | - Gijsbert J van Belle
- Institute of Cardiovascular Physiology, University Medical CenterGöttingen, Germany
| | - Steffen Vogelgesang
- Institute of Neuro- and Sensory Physiology, University Medical CenterGöttingen, Germany.,Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical CenterGöttingen, Germany
| | - Till Manzke
- Institute of Neuro- and Sensory Physiology, University Medical CenterGöttingen, Germany.,Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical CenterGöttingen, Germany
| | - Marcus Niebert
- Institute of Neuro- and Sensory Physiology, University Medical CenterGöttingen, Germany.,Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical CenterGöttingen, Germany
| |
Collapse
|
38
|
Torres RF, Hidalgo C, Kerr B. Mecp2 Mediates Experience-Dependent Transcriptional Upregulation of Ryanodine Receptor Type-3. Front Mol Neurosci 2017; 10:188. [PMID: 28659760 PMCID: PMC5468404 DOI: 10.3389/fnmol.2017.00188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/29/2017] [Indexed: 12/03/2022] Open
Abstract
Mecp2 is a DNA methylation reader that plays a critical role in experience-dependent plasticity. Increasing evidence supports a role for epigenetic modifications in activity-induced gene expression. Hence, candidate genes related to such phenomena are of great interest. Ryanodine receptors are intracellular calcium channels that contribute to hippocampal synaptic plasticity, dendritic spine remodeling, and participate in learning and memory processes. Here we exposed mice to the enriched environment (EE) paradigm, which through increased stimulation induces experience dependent-plasticity, to explore a role for methyl-cytosines, and Mecp2 in directing Ryanodine receptor 3 (Ryr3) transcriptional activity. EE induced a hippocampal-specific increase in the methylation of discrete cytosines located at a Ryr3 isoform promoter; chromatin immunoprecipitation experiments revealed that EE increased Mecp2 binding to this Ryr3 isoform promoter. Interestingly, the experimental paradigm induced robust Ryr3 upregulation, accompanied by miR132-dependent suppression of p250GAP, a pathway driving synaptogenesis. In contrast to WT mice, Mecp2-null mice showed diminished levels of Ryr3 and displayed impaired EE-induced Ryr3 upregulation, compromising miR132 dependent suppression of p250GAP and experience-dependent structural plasticity. Based on these results, we propose that Mecp2 acts as a transcriptional activator of Ryr3, contributing to experience-dependent plasticity.
Collapse
Affiliation(s)
- Rodrigo F Torres
- Laboratory of Biology, Centro de Estudios CientíficosValdivia, Chile
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Centro de Estudios Moleculares de la Célula, Department of Neuroscience and Physiology and Biophysics Program, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de ChileSantiago, Chile
| | - Bredford Kerr
- Laboratory of Biology, Centro de Estudios CientíficosValdivia, Chile
| |
Collapse
|
39
|
Valenti D, de Bari L, Vigli D, Lacivita E, Leopoldo M, Laviola G, Vacca RA, De Filippis B. Stimulation of the brain serotonin receptor 7 rescues mitochondrial dysfunction in female mice from two models of Rett syndrome. Neuropharmacology 2017; 121:79-88. [PMID: 28419872 DOI: 10.1016/j.neuropharm.2017.04.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/21/2017] [Accepted: 04/14/2017] [Indexed: 02/07/2023]
Abstract
Rett syndrome (RTT) is a rare neurodevelopmental disorder, characterized by severe behavioral and physiological symptoms. Mutations in the methyl CpG binding protein 2 gene (MECP2) cause more than 95% of classic cases, and currently there is no cure for this devastating disorder. Recently we have demonstrated that neurobehavioral and brain molecular alterations can be rescued in a RTT mouse model, by pharmacological stimulation of the brain serotonin receptor 7 (5-HT7R). This member of the serotonin receptor family, crucially involved in the regulation of brain structural plasticity and cognitive processes, can be stimulated by systemic repeated treatment with LP-211, a brain-penetrant selective agonist. The present study extends previous findings by demonstrating that LP-211 treatment (0.25 mg/kg, once per day for 7 days) rescues mitochondrial respiratory chain impairment, oxidative phosphorylation deficiency and the reduced energy status in the brain of heterozygous female mice from two highly validated mouse models of RTT (MeCP2-308 and MeCP2-Bird mice). Moreover, LP-211 treatment completely restored the radical species overproduction by brain mitochondria in the MeCP2-308 model and partially recovered the oxidative imbalance in the more severely affected MeCP2-Bird model. These results provide the first evidence that RTT brain mitochondrial dysfunction can be rescued targeting the brain 5-HT7R and add compelling preclinical evidence of the potential therapeutic value of LP-211 as a pharmacological approach for this devastating neurodevelopmental disorder.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy.
| | - Lidia de Bari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Daniele Vigli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Enza Lacivita
- Dept. Pharmacy, University of Bari "A. Moro", via Orabona 4, 70125 Bari, Italy
| | - Marcello Leopoldo
- Dept. Pharmacy, University of Bari "A. Moro", via Orabona 4, 70125 Bari, Italy
| | - Giovanni Laviola
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, Bari, Italy
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
40
|
Guadagni V, Cerri C, Piano I, Novelli E, Gargini C, Fiorentini C, Caleo M, Strettoi E. The bacterial toxin CNF1 as a tool to induce retinal degeneration reminiscent of retinitis pigmentosa. Sci Rep 2016; 6:35919. [PMID: 27775019 PMCID: PMC5075935 DOI: 10.1038/srep35919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/04/2016] [Indexed: 12/02/2022] Open
Abstract
Retinitis pigmentosa (RP) comprises a group of inherited pathologies characterized by progressive photoreceptor degeneration. In rodent models of RP, expression of defective genes and retinal degeneration usually manifest during the first weeks of postnatal life, making it difficult to distinguish consequences of primary genetic defects from abnormalities in retinal development. Moreover, mouse eyes are small and not always adequate to test pharmacological and surgical treatments. An inducible paradigm of retinal degeneration potentially extensible to large animals is therefore desirable. Starting from the serendipitous observation that intraocular injections of a Rho GTPase activator, the bacterial toxin Cytotoxic Necrotizing Factor 1 (CNF1), lead to retinal degeneration, we implemented an inducible model recapitulating most of the key features of Retinitis Pigmentosa. The model also unmasks an intrinsic vulnerability of photoreceptors to the mechanism of CNF1 action, indicating still unexplored molecular pathways potentially leading to the death of these cells in inherited forms of retinal degeneration.
Collapse
Affiliation(s)
- Viviana Guadagni
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Chiara Cerri
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy.,Accademia dei Lincei, Rome, 00165, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, 56126, Italy
| | - Elena Novelli
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Claudia Gargini
- Department of Pharmacy, University of Pisa, Pisa, 56126, Italy
| | | | - Matteo Caleo
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| | - Enrica Strettoi
- Neuroscience Institute, Italian National Research Council (CNR), Pisa, 56124, Italy
| |
Collapse
|
41
|
Therapeutic effects of the Rho GTPase modulator CNF1 in a model of Parkinson’s disease. Neuropharmacology 2016; 109:357-365. [DOI: 10.1016/j.neuropharm.2016.06.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 05/25/2016] [Accepted: 06/17/2016] [Indexed: 11/23/2022]
|
42
|
Romano E, Cosentino L, Laviola G, De Filippis B. Genes and sex hormones interaction in neurodevelopmental disorders. Neurosci Biobehav Rev 2016; 67:9-24. [DOI: 10.1016/j.neubiorev.2016.02.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/01/2016] [Indexed: 12/14/2022]
|
43
|
Veeraragavan S, Wan YW, Connolly DR, Hamilton SM, Ward CS, Soriano S, Pitcher MR, McGraw CM, Huang SG, Green JR, Yuva LA, Liang AJ, Neul JL, Yasui DH, LaSalle JM, Liu Z, Paylor R, Samaco RC. Loss of MeCP2 in the rat models regression, impaired sociability and transcriptional deficits of Rett syndrome. Hum Mol Genet 2016; 25:3284-3302. [PMID: 27365498 PMCID: PMC5179927 DOI: 10.1093/hmg/ddw178] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/18/2016] [Accepted: 06/08/2016] [Indexed: 01/31/2023] Open
Abstract
Mouse models of the transcriptional modulator Methyl-CpG-Binding Protein 2 (MeCP2) have advanced our understanding of Rett syndrome (RTT). RTT is a 'prototypical' neurodevelopmental disorder with many clinical features overlapping with other intellectual and developmental disabilities (IDD). Therapeutic interventions for RTT may therefore have broader applications. However, the reliance on the laboratory mouse to identify viable therapies for the human condition may present challenges in translating findings from the bench to the clinic. In addition, the need to identify outcome measures in well-chosen animal models is critical for preclinical trials. Here, we report that a novel Mecp2 rat model displays high face validity for modelling psychomotor regression of a learned skill, a deficit that has not been shown in Mecp2 mice. Juvenile play, a behavioural feature that is uniquely present in rats and not mice, is also impaired in female Mecp2 rats. Finally, we demonstrate that evaluating the molecular consequences of the loss of MeCP2 in both mouse and rat may result in higher predictive validity with respect to transcriptional changes in the human RTT brain. These data underscore the similarities and differences caused by the loss of MeCP2 among divergent rodent species which may have important implications for the treatment of individuals with disease-causing MECP2 mutations. Taken together, these findings demonstrate that the Mecp2 rat model is a complementary tool with unique features for the study of RTT and highlight the potential benefit of cross-species analyses in identifying potential disease-relevant preclinical outcome measures.
Collapse
Affiliation(s)
- Surabi Veeraragavan
- Department of Molecular and Human Genetics
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Daniel R Connolly
- Department of Molecular and Human Genetics
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | | | - Christopher S Ward
- Department of Pediatrics, Section of Neurology
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Sirena Soriano
- Department of Molecular and Human Genetics
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Meagan R Pitcher
- Program in Translational Biology and Molecular Medicine
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Christopher M McGraw
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Sharon G Huang
- Department of Molecular and Human Genetics
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | | | - Lisa A Yuva
- Department of Molecular and Human Genetics
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Agnes J Liang
- Department of Molecular and Human Genetics
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Jeffrey L Neul
- Department of Pediatrics, Section of Neurology
- Program in Translational Biology and Molecular Medicine
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Dag H Yasui
- Rowe Program in Human Genetics, University of California Davis, Davis, CA, USA
| | - Janine M LaSalle
- Rowe Program in Human Genetics, University of California Davis, Davis, CA, USA
| | - Zhandong Liu
- Department of Pediatrics, Section of Neurology
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | | | - Rodney C Samaco
- Department of Molecular and Human Genetics
- Program in Translational Biology and Molecular Medicine
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
44
|
Robillard KN, Lee KM, Chiu KB, MacLean AG. Glial cell morphological and density changes through the lifespan of rhesus macaques. Brain Behav Immun 2016; 55:60-69. [PMID: 26851132 PMCID: PMC4899176 DOI: 10.1016/j.bbi.2016.01.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 01/04/2016] [Accepted: 01/12/2016] [Indexed: 11/18/2022] Open
Abstract
How aging impacts the central nervous system (CNS) is an area of intense interest. Glial morphology is known to affect neuronal and immune function as well as metabolic and homeostatic balance. Activation of glia, both astrocytes and microglia, occurs at several stages during development and aging. The present study analyzed changes in glial morphology and density through the entire lifespan of rhesus macaques, which are physiologically and anatomically similar to humans. We observed apparent increases in gray matter astrocytic process length and process complexity as rhesus macaques matured from juveniles through adulthood. These changes were not attributed to cell enlargement because they were not accompanied by proportional changes in soma or process volume. There was a decrease in white matter microglial process length as rhesus macaques aged. Aging was shown to have a significant effect on gray matter microglial density, with a significant increase in aged macaques compared with adults. Overall, we observed significant changes in glial morphology as macaques age indicative of astrocytic activation with subsequent increase in microglial density in aged macaques.
Collapse
Affiliation(s)
- Katelyn N Robillard
- Tulane National Primate Research Center, Covington, LA, United States; Southeastern Louisiana University, Hammond, LA, United States
| | - Kim M Lee
- Tulane National Primate Research Center, Covington, LA, United States; Tulane Program in Biomedical Sciences, Tulane University School of Medicine, New Orleans, LA, United States
| | - Kevin B Chiu
- Tulane National Primate Research Center, Covington, LA, United States
| | - Andrew G MacLean
- Tulane National Primate Research Center, Covington, LA, United States; Tulane Program in Biomedical Sciences, Tulane University School of Medicine, New Orleans, LA, United States; Tulane Program in Neuroscience, Tulane University, New Orleans, LA, United States; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States.
| |
Collapse
|
45
|
Impairments in dendrite morphogenesis as etiology for neurodevelopmental disorders and implications for therapeutic treatments. Neurosci Biobehav Rev 2016; 68:946-978. [PMID: 27143622 DOI: 10.1016/j.neubiorev.2016.04.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 02/08/2023]
Abstract
Dendrite morphology is pivotal for neural circuitry functioning. While the causative relationship between small-scale dendrite morphological abnormalities (shape, density of dendritic spines) and neurodevelopmental disorders is well established, such relationship remains elusive for larger-scale dendrite morphological impairments (size, shape, branching pattern of dendritic trees). Here, we summarize published data on dendrite morphological irregularities in human patients and animal models for neurodevelopmental disorders, with focus on autism and schizophrenia. We next discuss high-risk genes for these disorders and their role in dendrite morphogenesis. We finally overview recent developments in therapeutic attempts and we discuss how they relate to dendrite morphology. We find that both autism and schizophrenia are accompanied by dendritic arbor morphological irregularities, and that majority of their high-risk genes regulate dendrite morphogenesis. Thus, we present a compelling argument that, along with smaller-scale morphological impairments in dendrites (spines and synapse), irregularities in larger-scale dendrite morphology (arbor shape, size) may be an important part of neurodevelopmental disorders' etiology. We suggest that this should not be ignored when developing future therapeutic treatments.
Collapse
|
46
|
Cell-to-cell propagation of the bacterial toxin CNF1 via extracellular vesicles: potential impact on the therapeutic use of the toxin. Toxins (Basel) 2015; 7:4610-21. [PMID: 26556375 PMCID: PMC4663523 DOI: 10.3390/toxins7114610] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/21/2015] [Accepted: 11/02/2015] [Indexed: 11/21/2022] Open
Abstract
Eukaryotic cells secrete extracellular vesicles (EVs), either constitutively or in a regulated manner, which represent an important mode of intercellular communication. EVs serve as vehicles for transfer between cells of membrane and cytosolic proteins, lipids and RNA. Furthermore, certain bacterial protein toxins, or possibly their derived messages, can be transferred cell to cell via EVs. We have herein demonstrated that eukaryotic EVs represent an additional route of cell-to-cell propagation for the Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1). Our results prove that EVs from CNF1 pre-infected epithelial cells can induce cytoskeleton changes, Rac1 and NF-κB activation comparable to that triggered by CNF1. The observation that the toxin is detectable inside EVs derived from CNF1-intoxicated cells strongly supports the hypothesis that extracellular vesicles can offer to the toxin a novel route to travel from cell to cell. Since anthrax and tetanus toxins have also been reported to engage in the same process, we can hypothesize that EVs represent a common mechanism exploited by bacterial toxins to enhance their pathogenicity.
Collapse
|
47
|
Travaglione S, Ballan G, Fortuna A, Ferri A, Guidotti M, Campana G, Fiorentini C, Loizzo S. CNF1 Enhances Brain Energy Content and Counteracts Spontaneous Epileptiform Phenomena in Aged DBA/2J Mice. PLoS One 2015; 10:e0140495. [PMID: 26457896 PMCID: PMC4601759 DOI: 10.1371/journal.pone.0140495] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/25/2015] [Indexed: 11/19/2022] Open
Abstract
Epilepsy, one of the most common conditions affecting the brain, is characterized by neuroplasticity and brain cell energy defects. In this work, we demonstrate the ability of the Escherichia coli protein toxin cytotoxic necrotizing factor 1 (CNF1) to counteract epileptiform phenomena in inbred DBA/2J mice, an animal model displaying genetic background with an high susceptibility to induced- and spontaneous seizures. Via modulation of the Rho GTPases, CNF1 regulates actin dynamics with a consequent increase in spine density and length in pyramidal neurons of rat visual cortex, and influences the mitochondrial homeostasis with remarkable changes in the mitochondrial network architecture. In addition, CNF1 improves cognitive performances and increases ATP brain content in mouse models of Rett syndrome and Alzheimer's disease. The results herein reported show that a single dose of CNF1 induces a remarkable amelioration of the seizure phenotype, with a significant augmentation in neuroplasticity markers and in cortex mitochondrial ATP content. This latter effect is accompanied by a decrease in the expression of mitochondrial fission proteins, suggesting a role of mitochondrial dynamics in the CNF1-induced beneficial effects on this epileptiform phenotype. Our results strongly support the crucial role of brain energy homeostasis in the pathogenesis of certain neurological diseases, and suggest that CNF1 could represent a putative new therapeutic tool for epilepsy.
Collapse
Affiliation(s)
- Sara Travaglione
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Giulia Ballan
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Andrea Fortuna
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Alberto Ferri
- Institute of Cellular Biology and Neurobiology, CNR, Via del Fosso di Fiorano 64/65, 00143, Roma, Italy
| | - Marco Guidotti
- Department of Veterinary Public Health and Food Safety, Viale Regina Elena 299, 00161, Roma, Italy
| | - Gabriele Campana
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Carla Fiorentini
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
| | - Stefano Loizzo
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Roma, Italy
- * E-mail:
| |
Collapse
|
48
|
Deficient Purposeful Use of Forepaws in Female Mice Modelling Rett Syndrome. Neural Plast 2015; 2015:326184. [PMID: 26185689 PMCID: PMC4491574 DOI: 10.1155/2015/326184] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/21/2015] [Accepted: 05/24/2015] [Indexed: 12/31/2022] Open
Abstract
Rett syndrome (RTT) is a rare neurodevelopmental disorder, characterized by severe behavioural and physiological symptoms. Mutations in the methyl CpG binding protein 2 gene (MECP2) cause more than 95% of classic cases. Motor abnormalities represent a significant part of the spectrum of RTT symptoms. In the present study we investigated motor coordination and fine motor skill domains in MeCP2-308 female mice, a validated RTT model. This was complemented by the in vivo magnetic resonance spectroscopy (MRS) analysis of metabolic profile in behaviourally relevant brain areas. MeCP2-308 heterozygous female mice (Het, 10-12 months of age) were impaired in tasks validated for the assessment of purposeful and coordinated forepaw use (Morag test and Capellini handling task). A fine-grain analysis of spontaneous behaviour in the home-cage also revealed an abnormal handling pattern when interacting with the nesting material, reduced motivation to explore the environment, and increased time devoted to feeding in Het mice. The brain MRS evaluation highlighted decreased levels of bioenergetic metabolites in the striatal area in Het mice compared to controls. Present results confirm behavioural and brain alterations previously reported in MeCP2-308 males and identify novel endpoints on which the efficacy of innovative therapeutic strategies for RTT may be tested.
Collapse
|
49
|
De Filippis B, Valenti D, de Bari L, De Rasmo D, Musto M, Fabbri A, Ricceri L, Fiorentini C, Laviola G, Vacca RA. Mitochondrial free radical overproduction due to respiratory chain impairment in the brain of a mouse model of Rett syndrome: protective effect of CNF1. Free Radic Biol Med 2015; 83:167-77. [PMID: 25708779 DOI: 10.1016/j.freeradbiomed.2015.02.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 11/19/2022]
Abstract
Rett syndrome (RTT) is a pervasive neurodevelopmental disorder mainly caused by mutations in the X-linked MECP2 gene associated with severe intellectual disability, movement disorders, and autistic-like behaviors. Its pathogenesis remains mostly not understood and no effective therapy is available. High circulating levels of oxidative stress markers in patients and the occurrence of oxidative brain damage in MeCP2-deficient mouse models suggest the involvement of oxidative stress in RTT pathogenesis. However, the molecular mechanism and the origin of the oxidative stress have not been elucidated. Here we demonstrate that a redox imbalance arises from aberrant mitochondrial functionality in the brain of MeCP2-308 heterozygous female mice, a condition that more closely recapitulates that of RTT patients. The marked increase in the rate of hydrogen peroxide generation in the brain of RTT mice seems mainly produced by the dysfunctional complex II of the mitochondrial respiratory chain. In addition, both membrane potential generation and mitochondrial ATP synthesis are decreased in RTT mouse brains when succinate, the complex II respiratory substrate, is used as an energy source. Respiratory chain impairment is brain area specific, owing to a decrease in either cAMP-dependent phosphorylation or protein levels of specific complex subunits. Further, we investigated whether the treatment of RTT mice with the bacterial protein CNF1, previously reported to ameliorate the neurobehavioral phenotype and brain bioenergetic markers in an RTT mouse model, exerts specific effects on brain mitochondrial function and consequently on hydrogen peroxide production. In RTT brains treated with CNF1, we observed the reactivation of respiratory chain complexes, the rescue of mitochondrial functionality, and the prevention of brain hydrogen peroxide overproduction. These results provide definitive evidence of mitochondrial reactive oxygen species overproduction in RTT mouse brain and highlight CNF1 efficacy in counteracting RTT-related mitochondrial defects.
Collapse
Affiliation(s)
- Bianca De Filippis
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, 00161 Roma, Italy.
| | - Daniela Valenti
- Institute of Biomembranes and Bioenergetics, National Council of Research, Bari, Italy
| | - Lidia de Bari
- Institute of Biomembranes and Bioenergetics, National Council of Research, Bari, Italy
| | - Domenico De Rasmo
- Institute of Biomembranes and Bioenergetics, National Council of Research, Bari, Italy
| | - Mattia Musto
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Alessia Fabbri
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Laura Ricceri
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Carla Fiorentini
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Giovanni Laviola
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes and Bioenergetics, National Council of Research, Bari, Italy.
| |
Collapse
|
50
|
Modulation of Rho GTPases rescues brain mitochondrial dysfunction, cognitive deficits and aberrant synaptic plasticity in female mice modeling Rett syndrome. Eur Neuropsychopharmacol 2015; 25:889-901. [PMID: 25890884 DOI: 10.1016/j.euroneuro.2015.03.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 02/03/2015] [Accepted: 03/22/2015] [Indexed: 11/20/2022]
Abstract
Rho GTPases are molecules critically involved in neuronal plasticity and cognition. We have previously reported that modulation of brain Rho GTPases by the bacterial toxin CNF1 rescues the neurobehavioral phenotype in MeCP2-308 male mice, a model of Rett syndrome (RTT). RTT is a rare X-linked neurodevelopmental disorder and a genetic cause of intellectual disability, for which no effective therapy is available. Mitochondrial dysfunction has been proposed to be involved in the mechanism of the disease pathogenesis. Here we demonstrate that modulation of Rho GTPases by CNF1 rescues the reduced mitochondrial ATP production via oxidative phosphorylation in the brain of MeCP2-308 heterozygous female mice, the condition which more closely recapitulates that of RTT patients. In RTT mouse brain, CNF1 also restores the alterations in the activity of the mitochondrial respiratory chain (MRC) complexes and of ATP synthase, the molecular machinery responsible for the majority of cell energy production. Such effects were achieved through the upregulation of the protein content of those MRC complexes subunits, which were defective in RTT mouse brain. Restored mitochondrial functionality was accompanied by the rescue of deficits in cognitive function (spatial reference memory in the Barnes maze), synaptic plasticity (long-term potentiation) and Tyr1472 phosphorylation of GluN2B, which was abnormally enhanced in the hippocampus of RTT mice. Present findings bring into light previously unknown functional mitochondrial alterations in the brain of female mice modeling RTT and provide the first evidence that RTT brain mitochondrial dysfunction can be rescued by modulation of Rho GTPases.
Collapse
|