Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 409
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(8): 1689-1707, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37059069

ABSTRACT

The nervous system governs both ontogeny and oncology. Regulating organogenesis during development, maintaining homeostasis, and promoting plasticity throughout life, the nervous system plays parallel roles in the regulation of cancers. Foundational discoveries have elucidated direct paracrine and electrochemical communication between neurons and cancer cells, as well as indirect interactions through neural effects on the immune system and stromal cells in the tumor microenvironment in a wide range of malignancies. Nervous system-cancer interactions can regulate oncogenesis, growth, invasion and metastatic spread, treatment resistance, stimulation of tumor-promoting inflammation, and impairment of anti-cancer immunity. Progress in cancer neuroscience may create an important new pillar of cancer therapy.


Subject(s)
Neoplasms , Neurosciences , Humans , Immune System , Neoplasms/pathology , Neurons/pathology , Tumor Microenvironment
2.
Cell ; 181(2): 219-222, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302564

ABSTRACT

Mounting evidence indicates that the nervous system plays a central role in cancer pathogenesis. In turn, cancers and cancer therapies can alter nervous system form and function. This Commentary seeks to describe the burgeoning field of "cancer neuroscience" and encourage multidisciplinary collaboration for the study of cancer-nervous system interactions.


Subject(s)
Neoplasms/metabolism , Nervous System/metabolism , Humans , Neurosciences
3.
Cell ; 181(7): 1445-1449, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32533917

ABSTRACT

The COVID19 crisis has magnified the issues plaguing academic science, but it has also provided the scientific establishment with an unprecedented opportunity to reset. Shoring up the foundation of academic science will require a concerted effort between funding agencies, universities, and the public to rethink how we support scientists, with a special emphasis on early career researchers.


Subject(s)
Career Mobility , Research Personnel/trends , Research/trends , Achievement , Biomedical Research , Humans , Research Personnel/education , Science/education , Science/trends , Universities
4.
Cell ; 176(1-2): 2-4, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30633904

ABSTRACT

With the advent of more successful chemotherapies, the number of cancer survivors continues to increase. Unfortunately, many of these patients will exhibit long-term sequelae from their treatments, including serious cognitive deficits that impair daily function. In this issue of Cell, Gibson et al. (2019) demonstrate that chemotherapy-related cognitive impairment is orchestrated by microglia.


Subject(s)
Cognition Disorders , Cognitive Dysfunction , Humans , Methotrexate
5.
Cell ; 170(5): 821-822, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28841413

ABSTRACT

The lateral ventricle (LV) is a preferential location for brain tumor spread; however, the instructive cues responsible for this unique tropism were previously unknown. In this issue, Qin et al. elucidate the underlying mechanism, demonstrating that LV-neural progenitors secrete a pleiotrophin (PTN)-containing complex, which attracts glioma cells through ROCK/Rho activation.


Subject(s)
Carrier Proteins , Glioma , Brain , Brain Neoplasms , Cytokines , Humans , Tropism
6.
Cell ; 164(3): 550-63, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824661

ABSTRACT

Therapy development for adult diffuse glioma is hindered by incomplete knowledge of somatic glioma driving alterations and suboptimal disease classification. We defined the complete set of genes associated with 1,122 diffuse grade II-III-IV gliomas from The Cancer Genome Atlas and used molecular profiles to improve disease classification, identify molecular correlations, and provide insights into the progression from low- to high-grade disease. Whole-genome sequencing data analysis determined that ATRX but not TERT promoter mutations are associated with increased telomere length. Recent advances in glioma classification based on IDH mutation and 1p/19q co-deletion status were recapitulated through analysis of DNA methylation profiles, which identified clinically relevant molecular subsets. A subtype of IDH mutant glioma was associated with DNA demethylation and poor outcome; a group of IDH-wild-type diffuse glioma showed molecular similarity to pilocytic astrocytoma and relatively favorable survival. Understanding of cohesive disease groups may aid improved clinical outcomes.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Transcriptome , Adult , Brain Neoplasms/metabolism , Cell Proliferation , Cluster Analysis , DNA Helicases/genetics , DNA Methylation , Epigenesis, Genetic , Glioma/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Middle Aged , Mutation , Nuclear Proteins/genetics , Promoter Regions, Genetic , Signal Transduction , Telomerase/genetics , Telomere , X-linked Nuclear Protein
7.
Nature ; 594(7862): 277-282, 2021 06.
Article in English | MEDLINE | ID: mdl-34040258

ABSTRACT

Neurons have recently emerged as essential cellular constituents of the tumour microenvironment, and their activity has been shown to increase the growth of a diverse number of solid tumours1. Although the role of neurons in tumour progression has previously been demonstrated2, the importance of neuronal activity to tumour initiation is less clear-particularly in the setting of cancer predisposition syndromes. Fifteen per cent of individuals with the neurofibromatosis 1 (NF1) cancer predisposition syndrome (in which tumours arise in close association with nerves) develop low-grade neoplasms of the optic pathway (known as optic pathway gliomas (OPGs)) during early childhood3,4, raising  the possibility that postnatal light-induced activity of the optic nerve drives tumour initiation. Here we use an authenticated mouse model of OPG driven by mutations in the neurofibromatosis 1 tumour suppressor gene (Nf1)5 to demonstrate that stimulation of optic nerve activity increases optic glioma growth, and that decreasing visual experience via light deprivation prevents tumour formation and maintenance. We show that the initiation of Nf1-driven OPGs (Nf1-OPGs) depends on visual experience during a developmental period in which Nf1-mutant mice are susceptible to tumorigenesis. Germline Nf1 mutation in retinal neurons results in aberrantly increased shedding of neuroligin 3 (NLGN3) within the optic nerve in response to retinal neuronal activity. Moreover, genetic Nlgn3 loss or pharmacological inhibition of NLGN3 shedding blocks the formation and progression of Nf1-OPGs. Collectively, our studies establish an obligate role for neuronal activity in the development of some types of brain tumours, elucidate a therapeutic strategy to reduce OPG incidence or mitigate tumour progression, and underscore the role of Nf1mutation-mediated dysregulation of neuronal signalling pathways in mouse models of the NF1 cancer predisposition syndrome.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, Neurofibromatosis 1 , Mutation , Neurofibromin 1/genetics , Neurons/metabolism , Optic Nerve Glioma/genetics , Optic Nerve Glioma/pathology , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Transformation, Neoplastic/radiation effects , Female , Germ-Line Mutation , Humans , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/radiation effects , Optic Nerve/cytology , Optic Nerve/radiation effects , Photic Stimulation , Retina/cytology , Retina/radiation effects
8.
Genes Dev ; 32(7-8): 491-496, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29632086

ABSTRACT

Pediatric low-grade gliomas (LGGs) frequently do not engraft in immunocompromised mice, limiting their use as an experimental platform. In contrast, murine Neurofibromatosis-1 (Nf1) optic LGG stem cells (o-GSCs) form glioma-like lesions in wild-type, but not athymic, mice following transplantation. Here, we show that the inability of athymic mice to support o-GSC engraftment results from impaired microglia/macrophage function, including reduced expression of Ccr2 and Ccl5, both of which are required for o-GSC engraftment and Nf1 optic glioma growth. Impaired Ccr2 and Ccl5 expression in athymic microglia/macrophages was restored by T-cell exposure, establishing T-cell-microglia/macrophage interactions as critical stromal determinants that support NF1 LGG growth.


Subject(s)
Glioma/immunology , Microglia/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Animals , Cells, Cultured , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokine CCL5/physiology , Gene Expression , Genes, Neurofibromatosis 1 , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Microglia/metabolism , Microglia/pathology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
9.
Hum Mol Genet ; 32(24): 3342-3352, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37712888

ABSTRACT

Single nucleotide variants in the general population are common genomic alterations, where the majority are presumed to be silent polymorphisms without known clinical significance. Using human induced pluripotent stem cell (hiPSC) cerebral organoid modeling of the 1.4 megabase Neurofibromatosis type 1 (NF1) deletion syndrome, we previously discovered that the cytokine receptor-like factor-3 (CRLF3) gene, which is co-deleted with the NF1 gene, functions as a major regulator of neuronal maturation. Moreover, children with NF1 and the CRLF3L389P variant have greater autism burden, suggesting that this gene might be important for neurologic function. To explore the functional consequences of this variant, we generated CRLF3L389P-mutant hiPSC lines and Crlf3L389P-mutant genetically engineered mice. While this variant does not impair protein expression, brain structure, or mouse behavior, CRLF3L389P-mutant human cerebral organoids and mouse brains exhibit impaired neuronal maturation and dendrite formation. In addition, Crlf3L389P-mutant mouse neurons have reduced dendrite lengths and branching, without any axonal deficits. Moreover, Crlf3L389P-mutant mouse hippocampal neurons have decreased firing rates and synaptic current amplitudes relative to wild type controls. Taken together, these findings establish the CRLF3L389P variant as functionally deleterious and suggest that it may be a neurodevelopmental disease modifier.


Subject(s)
Induced Pluripotent Stem Cells , Child , Humans , Animals , Mice , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Brain/metabolism , Receptors, Cytokine/metabolism , Nucleotides/metabolism
10.
Genes Dev ; 31(12): 1177-1179, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28765159

ABSTRACT

Converging evidence from numerous laboratories has revealed that malignant brain cancers are complex ecological systems composed of distinct cellular and acellular elements that collectively dictate glioblastoma biology. Our understanding of the individual contributions of each of these components is vital to the design of effective therapies against these cancers. In this issue of Genes & Development, Zanca and colleagues (pp. 1212-1227) demonstrate that one subpopulation of glioblastoma cells expressing a mutant epidermal growth factor receptor (EGFRvIII) is responsible for the survival of non-EGFRvIII-expressing tumor cells as well as for evading molecularly targeted therapy.


Subject(s)
Brain Neoplasms/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Brain Neoplasms/physiopathology , Brain Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Glioblastoma/physiopathology , Glioblastoma/therapy , Humans , Molecular Targeted Therapy , Mutation
11.
Childs Nerv Syst ; 2024 May 05.
Article in English | MEDLINE | ID: mdl-38704493

ABSTRACT

Neurofibromatosis type 1 (NF1) is an autosomal dominant cancer predisposition syndrome characterized by the development of both central and peripheral nervous system tumors. Low-grade glioma (LGG) is the most prevalent central nervous system tumor occurring in children with NF1, arising most frequently within the optic pathway, followed by the brainstem. Historically, treatment of NF1-LGG has been limited to conventional cytotoxic chemotherapy and surgery. Despite treatment with chemotherapy, a subset of children with NF1-LGG fail initial therapy, have a continued decline in function, or recur. The recent development of several preclinical models has allowed for the identification of novel, molecularly targeted therapies. At present, exploration of these novel precision-based therapies is ongoing in the preclinical setting and through larger, collaborative clinical trials. Herein, we review the approach to surveillance and management of NF1-LGG in children and discuss upcoming novel therapies and treatment protocols.

12.
Genes Dev ; 30(12): 1383-8, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27340174

ABSTRACT

As a critical regulator of cell growth, the mechanistic target of rapamycin (mTOR) protein operates as part of two molecularly and functionally distinct complexes. Herein, we demonstrate that mTOR complex molecular composition varies in different somatic tissues. In astrocytes and neural stem cells, we identified G-protein-coupled receptor kinase-interacting protein 1 (GIT1) as a novel mTOR-binding protein, creating a unique mTOR complex lacking Raptor and Rictor. Moreover, GIT1 binding to mTOR is regulated by AKT activation and is essential for mTOR-mediated astrocyte survival. Together, these data reveal that mTOR complex function is partly dictated by its molecuflar composition in different cell types.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Astrocytes/cytology , Astrocytes/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Survival/genetics , Cells, Cultured , Enzyme Activation , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Protein Binding , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics
13.
Cell ; 135(3): 549-60, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18984165

ABSTRACT

We uncovered a role for ERK signaling in GABA release, long-term potentiation (LTP), and learning, and show that disruption of this mechanism accounts for the learning deficits in a mouse model for learning disabilities in neurofibromatosis type I (NF1). Our results demonstrate that neurofibromin modulates ERK/synapsin I-dependent GABA release, which in turn modulates hippocampal LTP and learning. An Nf1 heterozygous null mutation, which results in enhanced ERK and synapsin I phosphorylation, increased GABA release in the hippocampus, and this was reversed by pharmacological downregulation of ERK signaling. Importantly, the learning deficits associated with the Nf1 mutation were rescued by a subthreshold dose of a GABA(A) antagonist. Accordingly, Cre deletions of Nf1 showed that only those deletions involving inhibitory neurons caused hippocampal inhibition, LTP, and learning abnormalities. Importantly, our results also revealed lasting increases in GABA release triggered by learning, indicating that the mechanisms uncovered here are of general importance for learning.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Neurofibromatosis 1 , Learning , Long-Term Potentiation , Neurofibromin 1/metabolism , Signal Transduction , gamma-Aminobutyric Acid/metabolism , Animals , Female , Hippocampus/metabolism , Learning Disabilities/physiopathology , Male , Mice , Neurofibromatosis 1/physiopathology , Neurofibromin 1/genetics , Phosphorylation , Synapsins/metabolism
14.
Childs Nerv Syst ; 39(3): 787-791, 2023 03.
Article in English | MEDLINE | ID: mdl-36107223

ABSTRACT

PURPOSE: Nearly a quarter of neurofibromatosis type 1 (NF 1)- associated diencephalic low-grade tumors are refractory to chemotherapy. Addition of alternative treatment options with laser interstitial thermal therapy will have a positive impact on the outcome of these patients. METHODS: We report on two illustrated cases of pediatric NF1- associated, chemoresistant, WHO grade 1 pilocytic astrocytomas treated with laser interstitial thermal therapy (LITT). RESULTS: Both tumors responded favorably to LITT. CONCLUSION: LITT should be considered as a treatment option for chemoresistant deep-seated NF1-associated low-grade gliomas.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Laser Therapy , Neurofibromatosis 1 , Humans , Child , Brain Neoplasms/complications , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Neurofibromatosis 1/complications , Neurofibromatosis 1/diagnostic imaging , Neurofibromatosis 1/therapy , Glioma/complications , Glioma/diagnostic imaging , Glioma/therapy , Magnetic Resonance Imaging , Astrocytoma/diagnostic imaging , Astrocytoma/therapy , Lasers
15.
Proc Natl Acad Sci U S A ; 117(46): 29101-29112, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33127758

ABSTRACT

Patients with amyotrophic lateral sclerosis (ALS) can have abnormal TDP-43 aggregates in the nucleus and cytosol of their surviving neurons and glia. Although accumulating evidence indicates that astroglial dysfunction contributes to motor neuron degeneration in ALS, the normal function of TDP-43 in astrocytes are largely unknown, and the role of astroglial TDP-43 loss to ALS pathobiology remains to be clarified. Herein, we show that TDP-43-deleted astrocytes exhibit a cell-autonomous increase in GFAP immunoreactivity without affecting astrocyte or microglia proliferation. At the transcriptomic level, TDP-43-deleted astrocytes resemble A1-reactive astrocytes and induce microglia to increase C1q expression. These astrocytic changes do not cause loss of motor neurons in the spinal cord or denervation at the neuromuscular junction. In contrast, there is a selective reduction of mature oligodendrocytes, but not oligodendrocyte precursor cells, suggesting triglial dysfunction mediated by TDP-43 loss in astrocytes. Moreover, mice with astroglial TDP-43 deletion develop motor, but not sensory, deficits. Taken together, our results demonstrate that TDP-43 is required to maintain the protective functions of astrocytes relevant to the development of motor deficits in mice.


Subject(s)
Astrocytes/metabolism , DNA-Binding Proteins/metabolism , Phenotype , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Motor Neurons/metabolism , Neuromuscular Junction/metabolism , Oligodendroglia/metabolism , Transcriptome
16.
Genes Dev ; 29(16): 1677-82, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26272820

ABSTRACT

Neurofibromatosis type 1 (NF1) is a common neurodevelopmental disorder caused by impaired function of the neurofibromin RAS regulator. Using a combination of Nf1 genetically engineered mice and pharmacological/genetic inhibition approaches, we report that neurofibromin differentially controls neural stem cell (NSC) proliferation and multilineage differentiation through the selective use of the PI3K/AKT and RAF/MEK pathways. While PI3K/AKT governs neurofibromin-regulated NSC proliferation, multilineage differentiation is MEK-dependent. Moreover, whereas MEK-regulated multilineage differentiation requires Smad3-induced Jagged-1 expression and Notch activation, MEK/Smad3-regulated Hes1 induction is only responsible for astrocyte and neuronal differentiation. Collectively, these findings establish distinct roles for the RAS effector pathways in regulating brain NSC function.


Subject(s)
Cell Differentiation , Neural Stem Cells/cytology , Neurofibromatosis 1/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Astrocytes/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins/genetics , Cell Lineage , Cell Proliferation , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurofibromatosis 1/genetics , Neurons/cytology , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Notch/metabolism , Serrate-Jagged Proteins , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transcription Factor HES-1 , ras Proteins/genetics
17.
Genet Med ; 24(9): 1967-1977, 2022 09.
Article in English | MEDLINE | ID: mdl-35674741

ABSTRACT

PURPOSE: Neurofibromatosis type 2 (NF2) and schwannomatosis (SWN) are genetically distinct tumor predisposition syndromes with overlapping phenotypes. We sought to update the diagnostic criteria for NF2 and SWN by incorporating recent advances in genetics, ophthalmology, neuropathology, and neuroimaging. METHODS: We used a multistep process, beginning with a Delphi method involving global disease experts and subsequently involving non-neurofibromatosis clinical experts, patients, and foundations/patient advocacy groups. RESULTS: We reached consensus on the minimal clinical and genetic criteria for diagnosing NF2 and SWN. These criteria incorporate mosaic forms of these conditions. In addition, we recommend updated nomenclature for these disorders to emphasize their phenotypic overlap and to minimize misdiagnosis with neurofibromatosis type 1. CONCLUSION: The updated criteria for NF2 and SWN incorporate clinical features and genetic testing, with a focus on using molecular data to differentiate the 2 conditions. It is likely that continued refinement of these new criteria will be necessary as investigators study the diagnostic properties of the revised criteria and identify new genes associated with SWN. In the revised nomenclature, the term "neurofibromatosis 2" has been retired to improve diagnostic specificity.


Subject(s)
Neurilemmoma , Neurofibromatoses , Neurofibromatosis 1 , Neurofibromatosis 2 , Skin Neoplasms , Consensus , Humans , Neurilemmoma/diagnosis , Neurilemmoma/genetics , Neurilemmoma/pathology , Neurofibromatoses/diagnosis , Neurofibromatoses/genetics , Neurofibromatosis 1/genetics , Neurofibromatosis 2/diagnosis , Neurofibromatosis 2/genetics , Skin Neoplasms/genetics
18.
J Pediatr ; 248: 94-99.e1, 2022 09.
Article in English | MEDLINE | ID: mdl-35561805

ABSTRACT

OBJECTIVE: To evaluate sociodemographic and medical predictors of patient return to a neurofibromatosis subspecialty clinic. STUDY DESIGN: Data were collected from the Washington University Neurofibromatosis Clinical Program electronic medical records. A total of 713 subjects with initial visits to the Washington University Neurofibromatosis Clinical Program between July 1, 2005 and December 18, 2020 were included. Variables collected included sex, race, ethnicity, age, date of first visit, place of residence, diagnosis, insurance payer, physician recommendation for return, and subject return. Return rates for each demographic group were calculated. Bivariate analyses were performed to inform variable inclusion in the model, and a binary logistic regression model was calculated to predict subject return. RESULTS: The overall return rate was 76%. The binary logistic regression model was statistically significant (χ29 = 131.094; P < .001) and showed that subjects who self-identified as Black and/or African American, presented with or received a diagnosis of café-au-lait macules at their initial visit, were from a rural area, were older, or who lived farther from the Washington University Neurofibromatosis Clinical Program were less likely to return to clinic. CONCLUSIONS: These findings support the implementation of tailored communication and monitoring interventions to improve the care for children with neurofibromatosis type 1.


Subject(s)
Neurofibromatoses , Neurofibromatosis 1 , Ambulatory Care Facilities , Cafe-au-Lait Spots/diagnosis , Child , Humans , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/therapy , Washington
19.
EMBO J ; 36(24): 3650-3665, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29127155

ABSTRACT

The tumor suppressors Tsc1 and Tsc2 form the tuberous sclerosis complex (TSC), a regulator of mTOR activity. Tsc1 stabilizes Tsc2; however, the precise mechanism involved remains elusive. The molecular chaperone heat-shock protein 90 (Hsp90) is an essential component of the cellular homeostatic machinery in eukaryotes. Here, we show that Tsc1 is a new co-chaperone for Hsp90 that inhibits its ATPase activity. The C-terminal domain of Tsc1 (998-1,164 aa) forms a homodimer and binds to both protomers of the Hsp90 middle domain. This ensures inhibition of both subunits of the Hsp90 dimer and prevents the activating co-chaperone Aha1 from binding the middle domain of Hsp90. Conversely, phosphorylation of Aha1-Y223 increases its affinity for Hsp90 and displaces Tsc1, thereby providing a mechanism for equilibrium between binding of these two co-chaperones to Hsp90. Our findings establish an active role for Tsc1 as a facilitator of Hsp90-mediated folding of kinase and non-kinase clients-including Tsc2-thereby preventing their ubiquitination and proteasomal degradation.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Tumor Suppressor Proteins/metabolism , HEK293 Cells , HSP90 Heat-Shock Proteins/genetics , Humans , Phosphorylation , Phosphotransferases/metabolism , Proteasome Endopeptidase Complex , Protein Folding , Proteolysis , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Ubiquitination
20.
Genet Med ; 23(8): 1506-1513, 2021 08.
Article in English | MEDLINE | ID: mdl-34012067

ABSTRACT

PURPOSE: By incorporating major developments in genetics, ophthalmology, dermatology, and neuroimaging, to revise the diagnostic criteria for neurofibromatosis type 1 (NF1) and to establish diagnostic criteria for Legius syndrome (LGSS). METHODS: We used a multistep process, beginning with a Delphi method involving global experts and subsequently involving non-NF experts, patients, and foundations/patient advocacy groups. RESULTS: We reached consensus on the minimal clinical and genetic criteria for diagnosing and differentiating NF1 and LGSS, which have phenotypic overlap in young patients with pigmentary findings. Criteria for the mosaic forms of these conditions are also recommended. CONCLUSION: The revised criteria for NF1 incorporate new clinical features and genetic testing, whereas the criteria for LGSS were created to differentiate the two conditions. It is likely that continued refinement of these new criteria will be necessary as investigators (1) study the diagnostic properties of the revised criteria, (2) reconsider criteria not included in this process, and (3) identify new clinical and other features of these conditions. For this reason, we propose an initiative to update periodically the diagnostic criteria for NF1 and LGSS.


Subject(s)
Neurofibromatosis 1 , Cafe-au-Lait Spots/genetics , Consensus , Genetic Testing , Humans , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL