Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 699
Filter
Add more filters

Publication year range
1.
Cell ; 187(8): 1955-1970.e23, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38503282

ABSTRACT

Characterizing somatic mutations in the brain is important for disentangling the complex mechanisms of aging, yet little is known about mutational patterns in different brain cell types. Here, we performed whole-genome sequencing (WGS) of 86 single oligodendrocytes, 20 mixed glia, and 56 single neurons from neurotypical individuals spanning 0.4-104 years of age and identified >92,000 somatic single-nucleotide variants (sSNVs) and small insertions/deletions (indels). Although both cell types accumulate somatic mutations linearly with age, oligodendrocytes accumulated sSNVs 81% faster than neurons and indels 28% slower than neurons. Correlation of mutations with single-nucleus RNA profiles and chromatin accessibility from the same brains revealed that oligodendrocyte mutations are enriched in inactive genomic regions and are distributed across the genome similarly to mutations in brain cancers. In contrast, neuronal mutations are enriched in open, transcriptionally active chromatin. These stark differences suggest an assortment of active mutagenic processes in oligodendrocytes and neurons.


Subject(s)
Aging , Brain , Neurons , Oligodendroglia , Humans , Aging/genetics , Aging/pathology , Chromatin/genetics , Chromatin/metabolism , Mutation , Neurons/metabolism , Neurons/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Single-Cell Gene Expression Analysis , Whole Genome Sequencing , Brain/metabolism , Brain/pathology , Polymorphism, Single Nucleotide , INDEL Mutation , Biological Specimen Banks , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology
2.
Cell ; 175(1): 10-13, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30217361

ABSTRACT

This year's Lasker Clinical Research Award goes to James Baird Glen for the discovery and development of the anesthetic propofol. Patients benefit from its fast onset and rapid systemic clearance, eliminating the prolonged sedation effects experienced with earlier agents. In just 30 years, propofol has been adopted around the world for safe and controlled induction of anesthesia.


Subject(s)
Propofol/pharmacology , Propofol/therapeutic use , Anesthesia/history , Anesthesia/methods , Awards and Prizes , History, 21st Century , Humans , Propofol/history
3.
Annu Rev Biochem ; 86: 1-19, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28125288

ABSTRACT

After an undergraduate degree in biology at Harvard, I started graduate school at The Rockefeller Institute for Medical Research in New York City in July 1965. I was attracted to the chemical side of biochemistry and joined Fritz Lipmann's large, hierarchical laboratory to study enzyme mechanisms. That work led to postdoctoral research with Robert Abeles at Brandeis, then a center of what, 30 years later, would be called chemical biology. I spent 15 years on the Massachusetts Institute of Technology faculty, in both the Chemistry and Biology Departments, and then 26 years on the Harvard Medical School Faculty. My research interests have been at the intersection of chemistry, biology, and medicine. One unanticipated major focus has been investigating the chemical logic and enzymatic machinery of natural product biosynthesis, including antibiotics and antitumor agents. In this postgenomic era it is now recognized that there may be from 105 to 106 biosynthetic gene clusters as yet uncharacterized for potential new therapeutic agents.


Subject(s)
Anti-Bacterial Agents/metabolism , Antineoplastic Agents/metabolism , Biochemistry/history , Biological Products/metabolism , Biomedical Research/history , Drug Industry/history , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , Biochemistry/trends , Biological Products/chemistry , Biomedical Research/trends , Drug Industry/trends , Gene Expression Regulation , History, 20th Century , History, 21st Century , Humans , Ligases/genetics , Ligases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Vancomycin Resistance/genetics , Workforce
4.
Cell ; 185(25): 4861, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36493757
5.
Cell ; 166(5): 1147-1162.e15, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565344

ABSTRACT

Alternative splicing is prevalent in the mammalian brain. To interrogate the functional role of alternative splicing in neural development, we analyzed purified neural progenitor cells (NPCs) and neurons from developing cerebral cortices, revealing hundreds of differentially spliced exons that preferentially alter key protein domains-especially in cytoskeletal proteins-and can harbor disease-causing mutations. We show that Ptbp1 and Rbfox proteins antagonistically govern the NPC-to-neuron transition by regulating neuron-specific exons. Whereas Ptbp1 maintains apical progenitors partly through suppressing a poison exon of Flna in NPCs, Rbfox proteins promote neuronal differentiation by switching Ninein from a centrosomal splice form in NPCs to a non-centrosomal isoform in neurons. We further uncover an intronic human mutation within a PTBP1-binding site that disrupts normal skipping of the FLNA poison exon in NPCs and causes a brain-specific malformation. Our study indicates that dynamic control of alternative splicing governs cell fate in cerebral cortical development.


Subject(s)
Alternative Splicing , Cerebral Cortex/embryology , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/cytology , Animals , Centrosome/metabolism , Cerebral Cortex/abnormalities , Cerebral Cortex/cytology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Exons , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Mice , Neural Stem Cells/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Protein Domains , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing Factors
6.
Cell ; 167(2): 341-354.e12, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27667684

ABSTRACT

Comparative analyses have identified genomic regions potentially involved in human evolution but do not directly assess function. Human accelerated regions (HARs) represent conserved genomic loci with elevated divergence in humans. If some HARs regulate human-specific social and behavioral traits, then mutations would likely impact cognitive and social disorders. Strikingly, rare biallelic point mutations-identified by whole-genome and targeted "HAR-ome" sequencing-showed a significant excess in individuals with ASD whose parents share common ancestry compared to familial controls, suggesting a contribution in 5% of consanguineous ASD cases. Using chromatin interaction sequencing, massively parallel reporter assays (MPRA), and transgenic mice, we identified disease-linked, biallelic HAR mutations in active enhancers for CUX1, PTBP2, GPC4, CDKL5, and other genes implicated in neural function, ASD, or both. Our data provide genetic evidence that specific HARs are essential for normal development, consistent with suggestions that their evolutionary changes may have altered social and/or cognitive behavior. PAPERCLIP.


Subject(s)
Autism Spectrum Disorder/genetics , Cognition , Genetic Predisposition to Disease , Neurogenesis/genetics , Point Mutation , Social Behavior , Alleles , Animals , Cerebral Cortex/metabolism , Gene Dosage , Genetic Variation , Genome, Human , Homeodomain Proteins/genetics , Humans , Introns , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Quantitative Trait Loci , Regulatory Elements, Transcriptional , Repressor Proteins/genetics , Transcription Factors
7.
Nature ; 604(7907): 714-722, 2022 04.
Article in English | MEDLINE | ID: mdl-35444284

ABSTRACT

Dementia in Alzheimer's disease progresses alongside neurodegeneration1-4, but the specific events that cause neuronal dysfunction and death remain poorly understood. During normal ageing, neurons progressively accumulate somatic mutations5 at rates similar to those of dividing cells6,7 which suggests that genetic factors, environmental exposures or disease states might influence this accumulation5. Here we analysed single-cell whole-genome sequencing data from 319 neurons from the prefrontal cortex and hippocampus of individuals with Alzheimer's disease and neurotypical control individuals. We found that somatic DNA alterations increase in individuals with Alzheimer's disease, with distinct molecular patterns. Normal neurons accumulate mutations primarily in an age-related pattern (signature A), which closely resembles 'clock-like' mutational signatures that have been previously described in healthy and cancerous cells6-10. In neurons affected by Alzheimer's disease, additional DNA alterations are driven by distinct processes (signature C) that highlight C>A and other specific nucleotide changes. These changes potentially implicate nucleotide oxidation4,11, which we show is increased in Alzheimer's-disease-affected neurons in situ. Expressed genes exhibit signature-specific damage, and mutations show a transcriptional strand bias, which suggests that transcription-coupled nucleotide excision repair has a role in the generation of mutations. The alterations in Alzheimer's disease affect coding exons and are predicted to create dysfunctional genetic knockout cells and proteostatic stress. Our results suggest that known pathogenic mechanisms in Alzheimer's disease may lead to genomic damage to neurons that can progressively impair function. The aberrant accumulation of DNA alterations in neurodegeneration provides insight into the cascade of molecular and cellular events that occurs in the development of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Neurons , Aging , Alzheimer Disease/genetics , Alzheimer Disease/pathology , DNA , Exons , Genomics , Hippocampus/cytology , Humans , Mutation Rate , Neurons/pathology , Nucleotides , Prefrontal Cortex/cytology , Whole Genome Sequencing
8.
Am J Hum Genet ; 111(5): 863-876, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38565148

ABSTRACT

Copy number variants (CNVs) are significant contributors to the pathogenicity of rare genetic diseases and, with new innovative methods, can now reliably be identified from exome sequencing. Challenges still remain in accurate classification of CNV pathogenicity. CNV calling using GATK-gCNV was performed on exomes from a cohort of 6,633 families (15,759 individuals) with heterogeneous phenotypes and variable prior genetic testing collected at the Broad Institute Center for Mendelian Genomics of the Genomics Research to Elucidate the Genetics of Rare Diseases consortium and analyzed using the seqr platform. The addition of CNV detection to exome analysis identified causal CNVs for 171 families (2.6%). The estimated sizes of CNVs ranged from 293 bp to 80 Mb. The causal CNVs consisted of 140 deletions, 15 duplications, 3 suspected complex structural variants (SVs), 3 insertions, and 10 complex SVs, the latter two groups being identified by orthogonal confirmation methods. To classify CNV variant pathogenicity, we used the 2020 American College of Medical Genetics and Genomics/ClinGen CNV interpretation standards and developed additional criteria to evaluate allelic and functional data as well as variants on the X chromosome to further advance the framework. We interpreted 151 CNVs as likely pathogenic/pathogenic and 20 CNVs as high-interest variants of uncertain significance. Calling CNVs from existing exome data increases the diagnostic yield for individuals undiagnosed after standard testing approaches, providing a higher-resolution alternative to arrays at a fraction of the cost of genome sequencing. Our improvements to the classification approach advances the systematic framework to assess the pathogenicity of CNVs.


Subject(s)
DNA Copy Number Variations , Exome Sequencing , Exome , Rare Diseases , Humans , DNA Copy Number Variations/genetics , Rare Diseases/genetics , Rare Diseases/diagnosis , Exome/genetics , Male , Female , Cohort Studies , Genetic Testing/methods
9.
Nat Rev Neurosci ; 23(5): 275-286, 2022 05.
Article in English | MEDLINE | ID: mdl-35322263

ABSTRACT

Genetic mosaicism is the result of the accumulation of somatic mutations in the human genome starting from the first postzygotic cell generation and continuing throughout the whole life of an individual. The rapid development of next-generation and single-cell sequencing technologies is now allowing the study of genetic mosaicism in normal tissues, revealing unprecedented insights into their clonal architecture and physiology. The somatic variant repertoire of an adult human neuron is the result of somatic mutations that accumulate in the brain by different mechanisms and at different rates during development and ageing. Non-pathogenic developmental mutations function as natural barcodes that once identified in deep bulk or single-cell sequencing can be used to retrospectively reconstruct human lineages. This approach has revealed novel insights into the clonal structure of the human brain, which is a mosaic of clones traceable to the early embryo that contribute differentially to the brain and distinct areas of the cortex. Some of the mutations happening during development, however, have a pathogenic effect and can contribute to some epileptic malformations of cortical development and autism spectrum disorder. In this Review, we discuss recent findings in the context of genetic mosaicism and their implications for brain development and disease.


Subject(s)
Autism Spectrum Disorder , Mosaicism , Adult , Autism Spectrum Disorder/genetics , Brain , Humans , Mutation/genetics , Retrospective Studies
10.
Cell ; 151(3): 483-96, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23101622

ABSTRACT

A major unanswered question in neuroscience is whether there exists genomic variability between individual neurons of the brain, contributing to functional diversity or to an unexplained burden of neurological disease. To address this question, we developed a method to amplify genomes of single neurons from human brains. Because recent reports suggest frequent LINE-1 (L1) retrotransposition in human brains, we performed genome-wide L1 insertion profiling of 300 single neurons from cerebral cortex and caudate nucleus of three normal individuals, recovering >80% of germline insertions from single neurons. While we find somatic L1 insertions, we estimate <0.6 unique somatic insertions per neuron, and most neurons lack detectable somatic insertions, suggesting that L1 is not a major generator of neuronal diversity in cortex and caudate. We then genotyped single cortical cells to characterize the mosaicism of a somatic AKT3 mutation identified in a child with hemimegalencephaly. Single-neuron sequencing allows systematic assessment of genomic diversity in the human brain.


Subject(s)
Caudate Nucleus/cytology , Cerebral Cortex/cytology , Long Interspersed Nucleotide Elements , Mutation , Neurons/metabolism , Single-Cell Analysis , Caudate Nucleus/metabolism , Cerebral Cortex/metabolism , Child , Chromosomes, Human, Pair 18 , Genome-Wide Association Study , Humans , Male , Malformations of Cortical Development/genetics , Malformations of Cortical Development/pathology , Mosaicism , Proto-Oncogene Proteins c-akt/genetics , Trisomy
11.
Cell ; 151(5): 1097-112, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23178126

ABSTRACT

Microcephaly is a neurodevelopmental disorder causing significantly reduced cerebral cortex size. Many known microcephaly gene products localize to centrosomes, regulating cell fate and proliferation. Here, we identify and characterize a nuclear zinc finger protein, ZNF335/NIF-1, as a causative gene for severe microcephaly, small somatic size, and neonatal death. Znf335 null mice are embryonically lethal, and conditional knockout leads to severely reduced cortical size. RNA-interference and postmortem human studies show that ZNF335 is essential for neural progenitor self-renewal, neurogenesis, and neuronal differentiation. ZNF335 is a component of a vertebrate-specific, trithorax H3K4-methylation complex, directly regulating REST/NRSF, a master regulator of neural gene expression and cell fate, as well as other essential neural-specific genes. Our results reveal ZNF335 as an essential link between H3K4 complexes and REST/NRSF and provide the first direct genetic evidence that this pathway regulates human neurogenesis and neuronal differentiation.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Nuclear Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins , Female , Gene Knockdown Techniques , Genes, Lethal , Histone-Lysine N-Methyltransferase , Humans , Male , Mice , Mice, Knockout , Microcephaly/metabolism , Multiprotein Complexes/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Repressor Proteins/metabolism , Transcription Factors
12.
Am J Hum Genet ; 110(5): 774-789, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37054711

ABSTRACT

The Integrator complex is a multi-subunit protein complex that regulates the processing of nascent RNAs transcribed by RNA polymerase II (RNAPII), including small nuclear RNAs, enhancer RNAs, telomeric RNAs, viral RNAs, and protein-coding mRNAs. Integrator subunit 11 (INTS11) is the catalytic subunit that cleaves nascent RNAs, but, to date, mutations in this subunit have not been linked to human disease. Here, we describe 15 individuals from 10 unrelated families with bi-allelic variants in INTS11 who present with global developmental and language delay, intellectual disability, impaired motor development, and brain atrophy. Consistent with human observations, we find that the fly ortholog of INTS11, dIntS11, is essential and expressed in the central nervous systems in a subset of neurons and most glia in larval and adult stages. Using Drosophila as a model, we investigated the effect of seven variants. We found that two (p.Arg17Leu and p.His414Tyr) fail to rescue the lethality of null mutants, indicating that they are strong loss-of-function variants. Furthermore, we found that five variants (p.Gly55Ser, p.Leu138Phe, p.Lys396Glu, p.Val517Met, and p.Ile553Glu) rescue lethality but cause a shortened lifespan and bang sensitivity and affect locomotor activity, indicating that they are partial loss-of-function variants. Altogether, our results provide compelling evidence that integrity of the Integrator RNA endonuclease is critical for brain development.


Subject(s)
Drosophila Proteins , Nervous System Diseases , Adult , Animals , Humans , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Mutation/genetics , RNA, Messenger
13.
Annu Rev Neurosci ; 41: 185-206, 2018 07 08.
Article in English | MEDLINE | ID: mdl-29986162

ABSTRACT

Understanding the biological basis for human-specific cognitive traits presents both immense challenges and unique opportunities. Although the question of what makes us human has been investigated with several different methods, the rise of comparative genomics, epigenomics, and medical genetics has provided tools to help narrow down and functionally assess the regions of the genome that seem evolutionarily relevant along the human lineage. In this review, we focus on how medical genetic cases have provided compelling functional evidence for genes and loci that appear to have interesting evolutionary signatures in humans. Furthermore, we examine a special class of noncoding regions, human accelerated regions (HARs), that have been suggested to show human-lineage-specific divergence, and how the use of clinical and population data has started to provide functional information to examine these regions. Finally, we outline methods that provide new insights into functional noncoding sequences in evolution.


Subject(s)
Behavior/physiology , Biological Evolution , Brain , Genomics , Nervous System Diseases , Animals , Brain/cytology , Brain/growth & development , Brain/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Nervous System Diseases/physiopathology
14.
Nature ; 586(7828): 262-269, 2020 10.
Article in English | MEDLINE | ID: mdl-32999462

ABSTRACT

Primates and rodents, which descended from a common ancestor around 90 million years ago1, exhibit profound differences in behaviour and cognitive capacity; the cellular basis for these differences is unknown. Here we use single-nucleus RNA sequencing to profile RNA expression in 188,776 individual interneurons across homologous brain regions from three primates (human, macaque and marmoset), a rodent (mouse) and a weasel (ferret). Homologous interneuron types-which were readily identified by their RNA-expression patterns-varied in abundance and RNA expression among ferrets, mice and primates, but varied less among primates. Only a modest fraction of the genes identified as 'markers' of specific interneuron subtypes in any one species had this property in another species. In the primate neocortex, dozens of genes showed spatial expression gradients among interneurons of the same type, which suggests that regional variation in cortical contexts shapes the RNA expression patterns of adult neocortical interneurons. We found that an interneuron type that was previously associated with the mouse hippocampus-the 'ivy cell', which has neurogliaform characteristics-has become abundant across the neocortex of humans, macaques and marmosets but not mice or ferrets. We also found a notable subcortical innovation: an abundant striatal interneuron type in primates that had no molecularly homologous counterpart in mice or ferrets. These interneurons expressed a unique combination of genes that encode transcription factors, receptors and neuropeptides and constituted around 30% of striatal interneurons in marmosets and humans.


Subject(s)
Interneurons/cytology , Primates , Animals , Callithrix , Cerebral Cortex/cytology , Female , Ferrets , Hippocampus/cytology , Humans , Interneurons/metabolism , LIM-Homeodomain Proteins/metabolism , Lysosomal Membrane Proteins/metabolism , Macaca , Male , Mice , Neostriatum/cytology , Nerve Tissue Proteins/metabolism , RNA/genetics , Species Specificity , Transcription Factors/metabolism
16.
Proc Natl Acad Sci U S A ; 120(23): e2300052120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252957

ABSTRACT

Short trinucleotide expansions at the FMR1 locus are associated with the late-onset condition fragile X-associated tremor/ataxia syndrome (FXTAS), which shows very different clinical and pathological features from fragile X syndrome (associated with longer expansions), with no clear molecular explanation for these marked differences. One prevailing theory posits that the shorter, premutation expansion uniquely causes extreme neurotoxic increases in FMR1 mRNA (i.e., four to eightfold increases), but evidence to support this hypothesis is largely derived from analysis of peripheral blood. We applied single-nucleus RNA sequencing to postmortem frontal cortex and cerebellum from 7 individuals with premutation and matched controls (n = 6) to assess cell type-specific molecular neuropathology. We found only modest upregulation (~1.3-fold) of FMR1 in some glial populations associated with premutation expansions. In premutation cases, we also identified decreased astrocyte proportions in the cortex. Differential expression and gene ontology analysis demonstrated altered neuroregulatory roles of glia. Using network analyses, we identified cell type-specific and region-specific patterns of FMR1 protein target gene dysregulation unique to premutation cases, with notable network dysregulation in the cortical oligodendrocyte lineage. We used pseudotime trajectory analysis to determine how oligodendrocyte development was altered and identified differences in early gene expression in oligodendrocyte trajectories in premutation cases specifically, implicating early cortical glial developmental perturbations. These findings challenge dogma regarding extremely elevated FMR1 increases in FXTAS and implicate glial dysregulation as a critical facet of premutation pathophysiology, representing potential unique therapeutic targets directly derived from the human condition.


Subject(s)
Fragile X Syndrome , Humans , Fragile X Syndrome/pathology , Tremor/genetics , Trinucleotide Repeat Expansion , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Ataxia/genetics , Ataxia/pathology , Brain/metabolism , Astrocytes/metabolism
17.
Proc Natl Acad Sci U S A ; 120(4): e2209964120, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36669111

ABSTRACT

Sonic hedgehog signaling regulates processes of embryonic development across multiple tissues, yet factors regulating context-specific Shh signaling remain poorly understood. Exome sequencing of families with polymicrogyria (disordered cortical folding) revealed multiple individuals with biallelic deleterious variants in TMEM161B, which encodes a multi-pass transmembrane protein of unknown function. Tmem161b null mice demonstrated holoprosencephaly, craniofacial midline defects, eye defects, and spinal cord patterning changes consistent with impaired Shh signaling, but were without limb defects, suggesting a CNS-specific role of Tmem161b. Tmem161b depletion impaired the response to Smoothened activation in vitro and disrupted cortical histogenesis in vivo in both mouse and ferret models, including leading to abnormal gyration in the ferret model. Tmem161b localizes non-exclusively to the primary cilium, and scanning electron microscopy revealed shortened, dysmorphic, and ballooned ventricular zone cilia in the Tmem161b null mouse, suggesting that the Shh-related phenotypes may reflect ciliary dysfunction. Our data identify TMEM161B as a regulator of cerebral cortical gyration, as involved in primary ciliary structure, as a regulator of Shh signaling, and further implicate Shh signaling in human gyral development.


Subject(s)
Ferrets , Hedgehog Proteins , Animals , Female , Humans , Mice , Pregnancy , Central Nervous System/metabolism , Cilia/genetics , Cilia/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice, Knockout , Signal Transduction
18.
Am J Hum Genet ; 109(2): 345-360, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35045343

ABSTRACT

Free oligosaccharides (fOSs) are soluble oligosaccharide species generated during N-glycosylation of proteins. Although little is known about fOS metabolism, the recent identification of NGLY1 deficiency, a congenital disorder of deglycosylation (CDDG) caused by loss of function of an enzyme involved in fOS metabolism, has elicited increased interest in fOS processing. The catabolism of fOSs has been linked to the activity of a specific cytosolic mannosidase, MAN2C1, which cleaves α1,2-, α1,3-, and α1,6-mannose residues. In this study, we report the clinical, biochemical, and molecular features of six individuals, including two fetuses, with bi-allelic pathogenic variants in MAN2C1; the individuals are from four different families. These individuals exhibit dysmorphic facial features, congenital anomalies such as tongue hamartoma, variable degrees of intellectual disability, and brain anomalies including polymicrogyria, interhemispheric cysts, hypothalamic hamartoma, callosal anomalies, and hypoplasia of brainstem and cerebellar vermis. Complementation experiments with isogenic MAN2C1-KO HAP1 cells confirm the pathogenicity of three of the identified MAN2C1 variants. We further demonstrate that MAN2C1 variants lead to accumulation and delay in the processing of fOSs in proband-derived cells. These results emphasize the involvement of MAN2C1 in human neurodevelopmental disease and the importance of fOS catabolism.


Subject(s)
Central Nervous System Cysts/genetics , Congenital Disorders of Glycosylation/genetics , Hamartoma/genetics , Intellectual Disability/genetics , Oligosaccharides/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Polymicrogyria/genetics , alpha-Mannosidase/genetics , Adolescent , Alleles , Brain Stem/metabolism , Brain Stem/pathology , Cell Line, Tumor , Central Nervous System Cysts/metabolism , Central Nervous System Cysts/pathology , Cerebellar Vermis/metabolism , Cerebellar Vermis/pathology , Child , Child, Preschool , Congenital Disorders of Glycosylation/metabolism , Congenital Disorders of Glycosylation/pathology , Female , Fetus , Glycosylation , Hamartoma/metabolism , Hamartoma/pathology , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Intellectual Disability/metabolism , Intellectual Disability/pathology , Leukocytes/metabolism , Leukocytes/pathology , Male , Mannose/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Polymicrogyria/metabolism , Polymicrogyria/pathology , Tongue/metabolism , Tongue/pathology , alpha-Mannosidase/deficiency
19.
Annu Rev Cell Dev Biol ; 27: 653-79, 2011.
Article in English | MEDLINE | ID: mdl-21801012

ABSTRACT

Cerebral cortical progenitor cells can be classified into several different types, and each progenitor type integrates cell-intrinsic and cell-extrinsic cues to regulate neurogenesis. On one hand, cell-intrinsic mechanisms that depend upon appropriate apical-basal polarity are established by adherens junctions and apical complex proteins and are particularly important in progenitors with apical processes contacting the lateral ventricle. The apical protein complexes themselves are concentrated at the ventricular surface, and apical complex proteins regulate mitotic spindle orientation and cell fate. On the other hand, remarkably little is known about how cell-extrinsic cues signal to progenitors and couple with cell-intrinsic mechanisms to instruct neurogenesis. Recent research shows that the cerebrospinal fluid, which contacts apical progenitors at the ventricular surface and bathes the apical complex of these cells, provides growth- and survival-promoting cues for neural progenitor cells in developing and adult brain. This review addresses how the apical-basal polarity of progenitor cells regulates cell fate and allows progenitors to sample diffusible signals distributed by the cerebrospinal fluid. We also review several classes of signaling factors that the cerebrospinal fluid distributes to the developing brain to instruct neurogenesis.


Subject(s)
Cerebral Cortex/anatomy & histology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cerebrospinal Fluid/metabolism , Neurogenesis/physiology , Adherens Junctions/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage , Cell Polarity , Cell Proliferation , Cerebrospinal Fluid/chemistry , Hedgehog Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Neurons/cytology , Neurons/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology , Tretinoin/metabolism
20.
Annu Rev Genomics Hum Genet ; 22: 239-256, 2021 08 31.
Article in English | MEDLINE | ID: mdl-33979534

ABSTRACT

Somatic mutations arise postzygotically, producing genetic differences between cells in an organism. Well established as a driver of cancer, somatic mutations also exist in nonneoplastic cells, including in the brain. Technological advances in nucleic acid sequencing have enabled recent breakthroughs that illuminate the roles of somatic mutations in aging and degenerative diseases of the brain. Somatic mutations accumulate during aging in human neurons, a process termed genosenium. A number of recent studies have examined somatic mutations in Alzheimer's disease (AD), primarily from the perspective of genes causing familial AD. We have also gained new information on genome-wide mutations, providing insights into the cellular events driving somatic mutation and cellular dysfunction. This review highlights recent concepts, methods, and findings in the progress to understand the role of brain somatic mutation in aging and AD.


Subject(s)
Alzheimer Disease , Aging/genetics , Alzheimer Disease/genetics , Brain , Humans , Mutation , Neurons
SELECTION OF CITATIONS
SEARCH DETAIL