1
|
Sell LB, Zabel C, Grønborg SW, Shi Q, Bhat MA. A Novel Mutation in CNTNAP1 Gene Causes Disorganization of Axonal Domains, Hypomyelination and Severe Neurological Deficits. J Neurosci Res 2025; 103:e70040. [PMID: 40265789 PMCID: PMC12037115 DOI: 10.1002/jnr.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
CNTNAP1 encodes the contactin-associated protein 1 (Cntnap1) which localizes to the paranodal region in all myelinated axons and is essential for axonal domain organization and the propagation of action potentials. To date, close to 45 reported human CNTNAP1 variants have been identified that are associated with dysregulation and disorganization of the axonal domains, resulting in various forms of congenital hypomyelinating neuropathies in children. Currently, no treatments are available for neuropathies caused by CNTNAP1 variants, highlighting the importance of fully characterizing these mutations and their impact on Cntnap1 functions. To understand the importance of a novel human CNTNAP1 likely pathogenic variant that changes glycine at position 349 to valine in a child who also carries a CNTNAP1 truncation and displayed severe neurological deficits, we used CRISPR/Cas9 methodology and introduced a single nucleotide substitution in the mouse Cntnap1 gene, resulting in glycine at 350 to valine (Cntnap1G350V). Trans-allelic combination of Cntnap1G350V with a Cntnap1 null allele (Cntnap1G350V/-) mimics human pathologies, recapitulating hypomyelination neuropathies associated with CNTNAP1 mutations as well as loss of paranodal junctions and disorganization of axonal domains in myelinated axons. Expression of the wild type Cntnap1 transgene in Cntnap1G350V/- mice rescued the mutant phenotypes and restored all neurological deficits. Our studies demonstrate that GGT (glycine) to GTT (valine) change in human CNTNAP1 creates a recessive loss of function allele and lays the foundation for potential gene therapy studies aimed at treating CNTNAP1-associated hypomyelinating neuropathies in children.
Collapse
Affiliation(s)
- Lacey B. Sell
- Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Carson Zabel
- Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Sabine Weller Grønborg
- Center for Rare Diseases, Department of Pediatrics and Adolescent Medicine and Department of Genetics, University Hospital Copenhagen Rigshospitalet, Blegdamsvej 9, 2100 København, Denmark
| | - Qian Shi
- Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Manzoor A. Bhat
- Department of Cellular and Integrative Physiology, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| |
Collapse
|
2
|
D'Amore A, Sundberg M, Lin R, Lubbers ET, Winden KD, Yu L, Gawlinska K, Gawlinski D, Lopez SG, Choe Y, Wightman EV, Liang Y, Modi M, Yuskaitis CJ, Lee HHC, Rotenberg A, Sahin M. Phenotypic rescue via mTOR inhibition in neuron-specific Pten knockout mice reveals AKT and mTORC1-site specific changes. Mol Psychiatry 2025:10.1038/s41380-025-02916-2. [PMID: 39953287 DOI: 10.1038/s41380-025-02916-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/22/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Phosphatase and Tensin Homolog (PTEN) is a dual-specific protein and lipid phosphatase that regulates AKT and downstream signaling of the mechanistic target of rapamycin (mTOR). PTEN functions as a tumor suppressor gene whose mutations result in PTEN Hamartoma Tumor Syndrome (PHTS) characterized by increased cancer risk and neurodevelopmental comorbidity. Here, we generated a novel neuron-specific Pten knock-out mouse model (Syn-Cre/Pten HOM) to test the ability of pharmacologic mTOR inhibition to rescue Pten mutation-associated disease phenotypes in vivo and in vitro. We found that treatment with the mTOR inhibitor, everolimus, increased the survival of Syn-Cre/Pten HOM mice while some neurologic phenotypes persisted. Transcriptomic analyses revealed that in contrast to mice harboring a neuron-specific deletion of the Tuberous Sclerosis Complex 2 gene (Syn-Cre/Tsc2 KO), genes that are under AKT regulation were significantly increased in the Syn-Cre/Pten HOM mice. In addition, genes associated with synapse, extracellular matrix, and myelination were broadly increased in Syn-Cre/Pten HOM mouse neocortex. These findings were confirmed by immunostaining of cortical sections in vivo, which revealed excessive immunoreactivity of myelin basic protein and perineuronal nets (PNN), the specialized extracellular matrix surrounding fast-spiking parvalbumin (PV) interneurons. We also detected increased expression of Synapsin I/PSD95 positive synapses and network hyperactivity phenotypes in Syn-Cre/Pten HOM mice neurons compared to wild-type (WT) neurons in vitro. Strikingly, everolimus treatment rescued the number of synapses and network hyperactivity in the Syn-Cre/Pten HOM mice cortical neuron cultures. Taken together, our results revealed in vivo and in vitro molecular and neuronal network mechanisms underlying neurological phenotypes of PHTS. Notably, pharmacologic mTOR inhibition by everolimus led to successful downstream signaling rescue, including mTOR complex 1 (mTORC1) site-specific suppression of S6 phosphorylation, correlating with phenotypic rescue found in our novel neuron-specific Syn-Cre/Pten HOM mice.
Collapse
Affiliation(s)
- Angelica D'Amore
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Maria Sundberg
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Rui Lin
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Ella T Lubbers
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Kellen D Winden
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Lucy Yu
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Kinga Gawlinska
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Department of Clinical Pharmacy, Jagiellonian University, Medical College, Medyczna 9, PL 30-688, Krakow, Poland
| | - Dawid Gawlinski
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Sam G Lopez
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Yongho Choe
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Emma V Wightman
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Yini Liang
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Meera Modi
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Christopher J Yuskaitis
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Boston, USA
| | - Henry Hing Cheong Lee
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, USA
| | - Alexander Rotenberg
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Boston, USA
| | - Mustafa Sahin
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, USA.
| |
Collapse
|
3
|
Karalis V, Wood D, Teaney NA, Sahin M. The role of TSC1 and TSC2 proteins in neuronal axons. Mol Psychiatry 2024; 29:1165-1178. [PMID: 38212374 DOI: 10.1038/s41380-023-02402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024]
Abstract
Tuberous Sclerosis Complex 1 and 2 proteins, TSC1 and TSC2 respectively, participate in a multiprotein complex with a crucial role for the proper development and function of the nervous system. This complex primarily acts as an inhibitor of the mechanistic target of rapamycin (mTOR) kinase, and mutations in either TSC1 or TSC2 cause a neurodevelopmental disorder called Tuberous Sclerosis Complex (TSC). Neurological manifestations of TSC include brain lesions, epilepsy, autism, and intellectual disability. On the cellular level, the TSC/mTOR signaling axis regulates multiple anabolic and catabolic processes, but it is not clear how these processes contribute to specific neurologic phenotypes. Hence, several studies have aimed to elucidate the role of this signaling pathway in neurons. Of particular interest are axons, as axonal defects are associated with severe neurocognitive impairments. Here, we review findings regarding the role of the TSC1/2 protein complex in axons. Specifically, we will discuss how TSC1/2 canonical and non-canonical functions contribute to the formation and integrity of axonal structure and function.
Collapse
Affiliation(s)
- Vasiliki Karalis
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Delaney Wood
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Nicole A Teaney
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Chang C, Sell LB, Shi Q, Bhat MA. Mouse models of human CNTNAP1-associated congenital hypomyelinating neuropathy and genetic restoration of murine neurological deficits. Cell Rep 2023; 42:113274. [PMID: 37862170 PMCID: PMC10873044 DOI: 10.1016/j.celrep.2023.113274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/22/2023] Open
Abstract
The Contactin-associated protein 1 (Cntnap1) mouse mutants fail to establish proper axonal domains in myelinated axons. Human CNTNAP1 mutations are linked to hypomyelinating neuropathy-3, which causes severe neurological deficits. To understand the human neuropathology and to model human CNTNAP1C323R and CNTNAP1R764C mutations, we generated Cntnap1C324R and Cntnap1R765C mouse mutants, respectively. Both Cntnap1 mutants show weight loss, reduced nerve conduction, and progressive motor dysfunction. The paranodal ultrastructure shows everted myelin loops and the absence of axo-glial junctions. Biochemical analysis reveals that these Cntnap1 mutant proteins are nearly undetectable in the paranodes, have reduced surface expression and stability, and are retained in the neuronal soma. Postnatal transgenic expression of Cntnap1 in the mutant backgrounds rescues the phenotypes and restores the organization of axonal domains with improved motor function. This study uncovers the mechanistic impact of two human CNTNAP1 mutations in a mouse model and provides proof of concept for gene therapy for CNTNAP1 patients.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Lacey B Sell
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; IBMS Neuroscience Graduate Program, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Qian Shi
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; IBMS Neuroscience Graduate Program, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; IBMS Neuroscience Graduate Program, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
5
|
Lin DS, Huang YW, Lee TH, Chang L, Huang ZD, Wu TY, Wang TJ, Ho CS. Rapamycin Alleviates Protein Aggregates, Reduces Neuroinflammation, and Rescues Demyelination in Globoid Cell Leukodystrophy. Cells 2023; 12:cells12070993. [PMID: 37048066 PMCID: PMC10093124 DOI: 10.3390/cells12070993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
We have shown in vivo and in vitro previously that psychosine causes dysfunction of autophagy and the ubiquitin-proteasome system underlying the pathogenesis of globoid cell leukodystrophy (GLD), a devastating lysosomal storage disease complicated by global demyelination. Here, we investigated the therapeutic efficacy of the mTOR inhibitor rapamycin in twitcher mice, a murine model of infantile GLD, in biochemical, histochemical, and clinical aspects. Administration of rapamycin to twitcher mice inhibited mTOR signaling in the brains, and significantly reduced the accumulation of insoluble ubiquitinated protein and the formation of ubiquitin aggregates. The astrocytes and microglia reactivity were attenuated in that reactive astrocytes, ameboid microglia, and globoid cells were reduced in the brains of rapamycin-treated twitcher mice. Furthermore, rapamycin improved the cortical myelination, neurite density, and rescued the network complexity in the cortex of twitcher mice. The therapeutic action of rapamycin on the pathology of the twitcher mice's brains prolonged the longevity of treated twitcher mice. Overall, these findings validate the therapeutic efficacy of rapamycin and highlight enhancing degradation of aggregates as a therapeutic strategy to modulate neuroinflammation, demyelination, and disease progression of GLD and other leukodystrophies associated with intracellular aggregates.
Collapse
Affiliation(s)
- Dar-Shong Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
| | - Yu-Wen Huang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Tsung-Han Lee
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Lung Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
| | - Zon-Darr Huang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Tsu-Yen Wu
- Department of Medical Research, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Tuan-Jen Wang
- Department of Laboratory Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan
| | - Che-Sheng Ho
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
- Department of Neurology, MacKay Children's Hospital, Taipei 10449, Taiwan
| |
Collapse
|
6
|
Barnes-Vélez JA, Aksoy Yasar FB, Hu J. Myelin lipid metabolism and its role in myelination and myelin maintenance. Innovation (N Y) 2023; 4:100360. [PMID: 36588745 PMCID: PMC9800635 DOI: 10.1016/j.xinn.2022.100360] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Myelin is a specialized cell membrane indispensable for rapid nerve conduction. The high abundance of membrane lipids is one of myelin's salient features that contribute to its unique role as an insulator that electrically isolates nerve fibers across their myelinated surface. The most abundant lipids in myelin include cholesterol, glycosphingolipids, and plasmalogens, each playing critical roles in myelin development as well as function. This review serves to summarize the role of lipid metabolism in myelination and myelin maintenance, as well as the molecular determinants of myelin lipid homeostasis, with an emphasis on findings from genetic models. In addition, the implications of myelin lipid dysmetabolism in human diseases are highlighted in the context of hereditary leukodystrophies and neuropathies as well as acquired disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Joseph A. Barnes-Vélez
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054-1901, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Science, Houston, TX 77225-0334, USA
- University of Puerto Rico Medical Sciences Campus, School of Medicine, San Juan, PR 00936-5067, USA
| | - Fatma Betul Aksoy Yasar
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054-1901, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Science, Houston, TX 77225-0334, USA
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054-1901, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Science, Houston, TX 77225-0334, USA
| |
Collapse
|
7
|
Raptor downregulation rescues neuronal phenotypes in mouse models of Tuberous Sclerosis Complex. Nat Commun 2022; 13:4665. [PMID: 35945201 PMCID: PMC9363483 DOI: 10.1038/s41467-022-31961-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 07/08/2022] [Indexed: 12/16/2022] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a neurodevelopmental disorder caused by mutations in the TSC1 or TSC2 genes, which encode proteins that negatively regulate mTOR complex 1 (mTORC1) signaling. Current treatment strategies focus on mTOR inhibition with rapamycin and its derivatives. While effective at improving some aspects of TSC, chronic rapamycin inhibits both mTORC1 and mTORC2 and is associated with systemic side-effects. It is currently unknown which mTOR complex is most relevant for TSC-related brain phenotypes. Here we used genetic strategies to selectively reduce neuronal mTORC1 or mTORC2 activity in mouse models of TSC. We find that reduction of the mTORC1 component Raptor, but not the mTORC2 component Rictor, rebalanced mTOR signaling in Tsc1 knock-out neurons. Raptor reduction was sufficient to improve several TSC-related phenotypes including neuronal hypertrophy, macrocephaly, impaired myelination, network hyperactivity, and premature mortality. Raptor downregulation represents a promising potential therapeutic intervention for the neurological manifestations of TSC.
Collapse
|
8
|
Shi Q, Chang C, Saliba A, Bhat MA. Microglial mTOR Activation Upregulates Trem2 and Enhances β-Amyloid Plaque Clearance in the 5XFAD Alzheimer's Disease Model. J Neurosci 2022; 42:5294-5313. [PMID: 35672148 PMCID: PMC9270922 DOI: 10.1523/jneurosci.2427-21.2022] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway plays a major role in key cellular processes including metabolism and differentiation; however, the role of mTOR in microglia and its importance in Alzheimer's disease (AD) have remained largely uncharacterized. We report that selective loss of Tsc1, a negative regulator of mTOR, in microglia in mice of both sexes, caused mTOR activation and upregulation of Trem2 with enhanced β-Amyloid (Aβ) clearance, reduced spine loss, and improved cognitive function in the 5XFAD AD mouse model. Combined loss of Tsc1 and Trem2 in microglia led to reduced Aβ clearance and increased Aβ plaque burden revealing that Trem2 functions downstream of mTOR. Tsc1 mutant microglia showed increased phagocytosis with upregulation of CD68 and Lamp1 lysosomal proteins. In vitro studies using Tsc1-deficient microglia revealed enhanced endocytosis of the lysosomal tracker indicator Green DND-26 suggesting increased lysosomal activity. Incubation of Tsc1-deficient microglia with fluorescent-labeled Aβ revealed enhanced Aβ uptake and clearance, which was blunted by rapamycin, an mTOR inhibitor. In vivo treatment of mice of relevant genotypes in the 5XFAD background with rapamycin, affected microglial activity, decreased Trem2 expression and reduced Aβ clearance causing an increase in Aβ plaque burden. Prolonged treatment with rapamycin caused even further reduction of mTOR activity, reduction in Trem2 expression, and increase in Aβ levels. Together, our findings reveal that mTOR signaling in microglia is critically linked to Trem2 regulation and lysosomal biogenesis, and that the upregulation of Trem2 in microglia through mTOR activation could be exploited toward better therapeutic avenues to Aβ-related AD pathologies.SIGNIFICANCE STATEMENT Mechanistic target of rapamycin (mTOR) signaling pathway is a key regulator for major cellular metabolic processes. However, the link between mTOR signaling and Alzheimer's disease (AD) is not well understood. In this study, we provide compelling in vivo evidence that mTOR activation in microglia would benefit β-Amyloid (Aβ)-related AD pathologies, as it upregulates Trem2, a key receptor for Aβ plaque uptake. Inhibition of mTOR pathway with rapamycin, a well-established immunosuppressant, downregulated Trem2 in microglia and reduced Aβ plaque clearance indicating that mTOR inactivation may be detrimental in Aβ-associated AD patients. This finding will have a significant public health impact and benefit, regarding the usage of rapamycin in AD patients, which we believe will aggravate the Aβ-related AD pathologies.
Collapse
Affiliation(s)
- Qian Shi
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Cheng Chang
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Afaf Saliba
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229
| |
Collapse
|
9
|
Garcia-Martin G, Alcover-Sanchez B, Wandosell F, Cubelos B. Pathways Involved in Remyelination after Cerebral Ischemia. Curr Neuropharmacol 2022; 20:751-765. [PMID: 34151767 PMCID: PMC9878953 DOI: 10.2174/1570159x19666210610093658] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022] Open
Abstract
Brain ischemia, also known as ischemic stroke, occurs when there is a lack of blood supply into the brain. When an ischemic insult appears, both neurons and glial cells can react in several ways that will determine the severity and prognosis. This high heterogeneity of responses has been a major obstacle in developing effective treatments or preventive methods for stroke. Although white matter pathophysiology has not been deeply assessed in stroke, its remodelling can greatly influence the clinical outcome and the disability degree. Oligodendrocytes, the unique cell type implied in CNS myelination, are sensible to ischemic damage. Loss of myelin sheaths can compromise axon survival, so new Oligodendrocyte Precursor Cells are required to restore brain function. Stroke can, therefore, enhance oligodendrogenesis to regenerate those new oligodendrocytes that will ensheath the damaged axons. Given that myelination is a highly complex process that requires coordination of multiple pathways such as Sonic Hedgehog, RTKs or Wnt/β-catenin, we will analyse new research highlighting their importance after brain ischemia. In addition, oligodendrocytes are not isolated cells inside the brain, but rather form part of a dynamic environment of interactions between neurons and glial cells. For this reason, we will put some context into how microglia and astrocytes react against stroke and influence oligodendrogenesis to highlight the relevance of remyelination in the ischemic brain. This will help to guide future studies to develop treatments focused on potentiating the ability of the brain to repair the damage.
Collapse
Affiliation(s)
- Gonzalo Garcia-Martin
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Berta Alcover-Sanchez
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Francisco Wandosell
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain,Address correspondence to this author at the Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Tel: 34-91-1964561; Fax: 34-91-1964420; E-mail:
| |
Collapse
|
10
|
EWAS of Monozygotic Twins Implicate a Role of mTOR Pathway in Pathogenesis of Tic Spectrum Disorder. Genes (Basel) 2021; 12:genes12101510. [PMID: 34680906 PMCID: PMC8535383 DOI: 10.3390/genes12101510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 11/17/2022] Open
Abstract
Tic spectrum disorder (TSD) is an umbrella term which includes Gilles de la Tourette syndrome (GTS) and chronic tic disorder (CTD). They are considered highly heritable, yet the genetic components remain largely unknown. In this study we aimed to investigate disease-associated DNA methylation differences to identify genes and pathways which may be implicated in TSD aetiology. For this purpose, we performed an exploratory analysis of the genome-wide DNA methylation patterns in whole blood samples of 16 monozygotic twin pairs, of which eight were discordant and six concordant for TSD, while two pairs were asymptomatic. Although no sites reached genome-wide significance, we identified several sites and regions with a suggestive significance, which were located within or in the vicinity of genes with biological functions associated with neuropsychiatric disorders. The two top genes identified (TSC1 and CRYZ/TYW3) and the enriched pathways and components (phosphoinosides and PTEN pathways, and insulin receptor substrate binding) are related to, or have been associated with, the PI3K/AKT/mTOR pathway. Genes in this pathway have previously been associated with GTS, and mTOR signalling has been implicated in a range of neuropsychiatric disorders. It is thus possible that altered mTOR signalling plays a role in the complex pathogenesis of TSD.
Collapse
|
11
|
Deficiency of Microglial Autophagy Increases the Density of Oligodendrocytes and Susceptibility to Severe Forms of Seizures. eNeuro 2021; 8:ENEURO.0183-20.2021. [PMID: 33472865 PMCID: PMC7890520 DOI: 10.1523/eneuro.0183-20.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/11/2020] [Accepted: 01/02/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive activation of mTOR in microglia impairs CNS homeostasis and causes severe epilepsy. Autophagy constitutes an important part of mTOR signaling. The contribution of microglial autophagy to CNS homeostasis and epilepsy remains to be determined. Here, we report that ATG7KO mice deficient for autophagy in microglia display a marked increase of myelination markers, a higher density of mature oligodendrocytes (ODCs), and altered lengths of the nodes of Ranvier. Moreover, we found that deficiency of microglial autophagy (ATG7KO) leads to increased seizure susceptibility in three seizure models (pilocarpine, kainic acid, and amygdala kindling). We demonstrated that ATG7KO mice develop severe generalized seizures and display nearly 100% mortality to convulsions induced by pilocarpine and kainic acid. In the amygdala kindling model, we observed significant facilitation of contralateral propagation of seizures, a process underlying the development of generalized seizures. Taken together, our results reveal impaired microglial autophagy as a novel mechanism underlying altered homeostasis of ODCs and increased susceptibility to severe and fatal generalized seizures.
Collapse
|
12
|
R-Ras GTPases Signaling Role in Myelin Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21165911. [PMID: 32824627 PMCID: PMC7460555 DOI: 10.3390/ijms21165911] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/18/2022] Open
Abstract
Myelination is required for fast and efficient synaptic transmission in vertebrates. In the central nervous system, oligodendrocytes are responsible for creating myelin sheaths that isolate and protect axons, even throughout adulthood. However, when myelin is lost, the failure of remyelination mechanisms can cause neurodegenerative myelin-associated pathologies. From oligodendrocyte progenitor cells to mature myelinating oligodendrocytes, myelination is a highly complex process that involves many elements of cellular signaling, yet many of the mechanisms that coordinate it, remain unknown. In this review, we will focus on the three major pathways involved in myelination (PI3K/Akt/mTOR, ERK1/2-MAPK, and Wnt/β-catenin) and recent advances describing the crosstalk elements which help to regulate them. In addition, we will review the tight relation between Ras GTPases and myelination processes and discuss its potential as novel elements of crosstalk between the pathways. A better understanding of the crosstalk elements orchestrating myelination mechanisms is essential to identify new potential targets to mitigate neurodegeneration.
Collapse
|
13
|
Dong J, Shin N, Chen S, Lei J, Burd I, Wang X. Is there a definite relationship between placental mTOR signaling and fetal growth? Biol Reprod 2020; 103:471-486. [PMID: 32401303 DOI: 10.1093/biolre/ioaa070] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/22/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023] Open
Abstract
Fetal growth restriction and overgrowth are common obstetrical complications that result in adverse perinatal outcomes and long-term health risks later in life, including neurodevelopmental dysfunction and adult metabolic syndrome. The placenta plays a critical role in the nutrition transfer from mother to fetus and even exerts adaptive mechanism when the fetus is under poor developmental conditions. The mammalian/mechanistic target of rapamycin (mTOR) signaling serves as a critical hub of cell growth, survival, and metabolism in response to nutrients, growth factors, energy, and stress signals. Placental mTOR signaling regulates placental function, including oxygen and nutrient transport. Therefore, placental mTOR signaling is hypothesized to have a positive relationship with fetal growth. In this review, we summarize that most studies support the current evidence that there is connection between placental mTOR signaling and abnormal fetal growth; however, but more studies should be performed following a vigorous and unanimous method for assessment to determine placental mTOR activity.
Collapse
Affiliation(s)
- Jie Dong
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Na Shin
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shuqiang Chen
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaohong Wang
- Reproductive Medical Center, Department of Gynecology and Obstetrics, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
14
|
Li W, Su D, Zhai Q, Chi H, She X, Gao X, Wang K, Yang H, Wang R, Cui B. Proteomes analysis reveals the involvement of autophagy in AD-like neuropathology induced by noise exposure and ApoE4. ENVIRONMENTAL RESEARCH 2019; 176:108537. [PMID: 31228807 DOI: 10.1016/j.envres.2019.108537] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Noise is one of the most important environmental health hazards for humans. Environmental noise or apolipoprotein ε4 (ApoE4) can cause typical Alzheimer's disease (AD)-like pathological changes, which is characterized by progressive cognitive decline and neurodegenerative lesions. Gene-environment interactions may accelerate cognitive decline and increase AD risk. However, there is limited experimental evidence regarding the underlying mechanisms of noise-ApoE4 interactions and AD, which may be closely related to AD development. METHODS In this study, we investigated the combined effects of chronic noise exposure and the ApoE4 gene activation on hippocampus by using proteomics and differentially expressed proteins were found through performed gene ontology function and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. In addition, we assessed the changes in adult hippocampal neurogenesis and potential underlying mechanism for AD-like neuropathology. RESULTS Relative to control rats, combined exposure of noise and ApoE4 synergistically increased the characteristic pathological amyloid β-protein of AD-like neuropathology changes in hippocampus. The research identifies a total of 4147 proteins and 15 differentially expressed proteins in hippocampus. Furthermore, comparison of several of the diverse key pathways studied (e.g., PI3K/AKT, insulin, calpain-CDK5, and mammalian target of rapamycin (mTOR) signaling pathways) help to articulate the different mechanisms involved in combined effects of noise and ApoE4 on AD-like pathology. We verified four selected proteins, namely, eukaryotic translation elongation factor 1 epsilon 1, glycine amidinotransferase, nucleoredoxin, and tuberous sclerosis 1 proteins. Validation data shows significant effects of chronic noise and ApoE4 on the expression of four selected proteins, eukaryotic translation elongation factor 1 epsilon 1, glycine amidinotransferase, nucleoredoxin, and tuberous sclerosis 1 proteins, and mTOR and autophagy-related proteins, which share significant interaction effect of chronic noise and ApoE4. CONCLUSION Gene-environment interactions between chronic noise and ApoE4 activate the mTOR signaling, decrease autophagy, and facilitate AD-like changes in the hippocampus. Thus, our findings may help elucidate the role of gene-environment interactions in AD development.
Collapse
Affiliation(s)
- Wenlong Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Donghong Su
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Qingfeng Zhai
- School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Huimin Chi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; School of Public Health and Management, Weifang Medical University, Weifang, China
| | - Xiaojun She
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiujie Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kun Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Honglian Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Rui Wang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Bo Cui
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.
| |
Collapse
|