Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 94.617
1.
Sci Rep ; 14(1): 10583, 2024 05 08.
Article En | MEDLINE | ID: mdl-38719848

Identifying marker combinations for robust prognostic validation in primary tumour compartments remains challenging. We aimed to assess the prognostic significance of CSC markers (ALDH1, CD44, p75NTR, BMI-1) and E-cadherin biomarkers in OSCC. We analysed 94 primary OSCC and 67 metastatic lymph node samples, including central and invasive tumour fronts (ITF), along with clinicopathological data. We observed an increase in ALDH1+/CD44+/BMI-1- tumour cells in metastatic lesions compared to primary tumours. Multivariate analysis highlighted that elevated p75NTR levels (at ITF) and reduced E-cadherin expression (at the tumour centre) independently predicted metastasis, whilst ALDH1high exhibited independent predictive lower survival at the ITF, surpassing the efficacy of traditional tumour staging. Then, specifically at the ITF, profiles characterized by CSChighE-cadherinlow (ALDH1highp75NTRhighE-cadherinlow) and CSCintermediateE-cadherinlow (ALDH1 or p75NTRhighE-cadherinlow) were significantly associated with worsened overall survival and increased likelihood of metastasis in OSCC patients. In summary, our study revealed diverse tumour cell profiles in OSCC tissues, with varying CSC and E-cadherin marker patterns across primary tumours and metastatic sites. Given the pivotal role of reduced survival rates as an indicator of unfavourable prognosis, the immunohistochemistry profile identified as CSChighE-cadherinlow at the ITF of primary tumours, emerges as a preferred prognostic marker closely linked to adverse outcomes in OSCC.


Aldehyde Dehydrogenase 1 Family , Biomarkers, Tumor , Cadherins , Carcinoma, Squamous Cell , Immunohistochemistry , Mouth Neoplasms , Humans , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Mouth Neoplasms/diagnosis , Cadherins/metabolism , Female , Male , Prognosis , Biomarkers, Tumor/metabolism , Middle Aged , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/mortality , Aged , Aldehyde Dehydrogenase 1 Family/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Nerve Growth Factor/metabolism , Retinal Dehydrogenase/metabolism , Hyaluronan Receptors/metabolism , Adult , Lymphatic Metastasis , Nerve Tissue Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/genetics
2.
Invest Ophthalmol Vis Sci ; 65(5): 8, 2024 May 01.
Article En | MEDLINE | ID: mdl-38700874

Purpose: In the present study, we aim to elucidate the underlying molecular mechanism of endoplasmic reticulum (ER) stress induced delayed corneal epithelial wound healing and nerve regeneration. Methods: Human limbal epithelial cells (HLECs) were treated with thapsigargin to induce excessive ER stress and then RNA sequencing was performed. Immunofluorescence, qPCR, Western blot, and ELISA were used to detect the expression changes of SLIT3 and its receptors ROBO1-4. The role of recombinant SLIT3 protein in corneal epithelial proliferation and migration were assessed by CCK8 and cell scratch assay, respectively. Thapsigargin, exogenous SLIT3 protein, SLIT3-specific siRNA, and ROBO4-specific siRNA was injected subconjunctivally to evaluate the effects of different intervention on corneal epithelial and nerve regeneration. In addition, Ki67 staining was performed to evaluate the proliferation ability of epithelial cells. Results: Thapsigargin suppressed normal corneal epithelial and nerve regeneration significantly. RNA sequencing genes related to development and regeneration revealed that thapsigargin induced ER stress significantly upregulated the expression of SLIT3 and ROBO4 in corneal epithelial cells. Exogenous SLIT3 inhibited normal corneal epithelial injury repair and nerve regeneration, and significantly suppressed the proliferation and migration ability of cultured mouse corneal epithelial cells. SLIT3 siRNA inhibited ROBO4 expression and promoted epithelial wound healing under thapsigargin treatment. ROBO4 siRNA significantly attenuated the delayed corneal epithelial injury repair and nerve regeneration induced by SLIT3 treatment or thapsigargin treatment. Conclusions: ER stress inhibits corneal epithelial injury repair and nerve regeneration may be related with the upregulation of SLIT3-ROBO4 pathway.


Cell Proliferation , Endoplasmic Reticulum Stress , Epithelium, Corneal , Nerve Regeneration , Receptors, Immunologic , Roundabout Proteins , Signal Transduction , Wound Healing , Animals , Humans , Mice , Blotting, Western , Cell Movement/physiology , Cells, Cultured , Endoplasmic Reticulum Stress/physiology , Enzyme-Linked Immunosorbent Assay , Epithelium, Corneal/metabolism , Limbus Corneae/cytology , Nerve Regeneration/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction/physiology , Wound Healing/physiology
3.
Cell Mol Life Sci ; 81(1): 209, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710967

As an integral lysosomal transmembrane protein, transmembrane protein 106B (TMEM106B) regulates several aspects of lysosomal function and is associated with neurodegenerative diseases. The TMEM106B gene mutations lead to lysosomal dysfunction and accelerate the pathological progression of Neurodegenerative diseases. Yet, the precise mechanism of TMEM106B in Neurodegenerative diseases remains unclear. Recently, different research teams discovered that TMEM106B is an amyloid protein and the C-terminal domain of TMEM106B forms amyloid fibrils in various Neurodegenerative diseases and normally elderly individuals. In this review, we discussed the physiological functions of TMEM106B. We also included TMEM106B gene mutations that cause neurodegenerative diseases. Finally, we summarized the identification and cryo-electronic microscopic structure of TMEM106B fibrils, and discussed the promising therapeutic strategies aimed at TMEM106B fibrils and the future directions for TMEM106B research in neurodegenerative diseases.


Membrane Proteins , Mutation , Nerve Tissue Proteins , Neurodegenerative Diseases , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/chemistry , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/chemistry , Animals , Lysosomes/metabolism , Lysosomes/genetics , Amyloid/metabolism , Amyloid/genetics , Amyloid/chemistry
4.
Proc Natl Acad Sci U S A ; 121(23): e2318740121, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38805275

Repressor element-1 silencing transcription factor (REST) is required for the formation of mature neurons. REST dysregulation underlies a key mechanism of neurodegeneration associated with neurological disorders. However, the mechanisms leading to alterations of REST-mediated silencing of key neurogenesis genes are not known. Here, we show that BRCA1 Associated ATM Activator 1 (BRAT1), a gene linked to neurodegenerative diseases, is required for the activation of REST-responsive genes during neuronal differentiation. We find that INTS11 and INTS9 subunits of Integrator complex interact with BRAT1 as a distinct trimeric complex to activate critical neuronal genes during differentiation. BRAT1 depletion results in persistence of REST residence on critical neuronal genes disrupting the differentiation of NT2 cells into astrocytes and neuronal cells. We identified BRAT1 and INTS11 co-occupying the promoter region of these genes and pinpoint a role for BRAT1 in recruiting INTS11 to their promoters. Disease-causing mutations in BRAT1 diminish its association with INTS11/INTS9, linking the manifestation of disease phenotypes with a defect in transcriptional activation of key neuronal genes by BRAT1/INTS11/INTS9 complex. Finally, loss of Brat1 in mouse embryonic stem cells leads to a defect in neuronal differentiation assay. Importantly, while reconstitution with wild-type BRAT1 restores neuronal differentiation, the addition of a BRAT1 mutant is unable to associate with INTS11/INTS9 and fails to rescue the neuronal phenotype. Taken together, our study highlights the importance of BRAT1 association with INTS11 and INTS9 in the development of the nervous system.


Cell Differentiation , Chromatin , Neurogenesis , Neurons , Repressor Proteins , Chromatin/metabolism , Chromatin/genetics , Neurons/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Humans , Neurogenesis/genetics , Animals , Mice , Promoter Regions, Genetic , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Nerve Tissue Proteins , Co-Repressor Proteins
5.
Cell Death Dis ; 15(5): 367, 2024 May 28.
Article En | MEDLINE | ID: mdl-38806484

Mitochondria dysfunctions and mitophagy failure have been associated with several Alzheimer's disease (AD) related molecular actors including amyloid beta (Aß) and recently the amyloid precursor protein-C terminal fragments (APP-CTFs). The efficacy of the mitophagy process in neurons relies on regulated mitochondrial transport along axons involving a complex molecular machinery. The contribution of the amyloid precursor protein (APP) and its derived fragments to the mitochondrial transport machinery alterations in AD have not been investigated before. We report herein a change of the expression of mitochondrial transport proteins (SNPH and Miro1), motor adapters (TRANK1 and TRAK2), and components of the dynein and kinesin motors (i.e., IC1,2 and Kif5 (A, B, C) isoforms) by endogenous APP and by overexpression of APP carrying the familial Swedish mutation (APPswe). We show that APP-CTFs and Aß concomitantly regulate the expression of a set of transport proteins as demonstrated in APPswe cells treated with ß- and γ-secretase inhibitors and in cells Knock-down for presenilin 1 and 2. We further report the impact of APP-CTFs on the expression of transport proteins in AAV-injected C99 mice brains. Our data also indicate that both Aß oligomers (Aßo) and APP-CTFs impair the colocalization of mitochondria and transport proteins. This has been demonstrated in differentiated SH-SY5Y naive cells treated with Aßo and in differentiated SH-SY5Y and murine primary neurons expressing APPswe and treated with the γ-secretase inhibitor. Importantly, we uncover that the expression of a set of transport proteins is modulated in a disease-dependent manner in 3xTgAD mice and in human sporadic AD brains. This study highlights molecular mechanisms underlying mitochondrial transport defects in AD that likely contribute to mitophagy failure and disease progression.


Alzheimer Disease , Amyloid beta-Protein Precursor , Mitochondria , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Mitochondria/metabolism , Humans , Mice , Mice, Transgenic , Neurons/metabolism , Amyloid beta-Peptides/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Amyloid Precursor Protein Secretases/metabolism , Kinesins/metabolism , Biological Transport , Mitophagy , Nerve Tissue Proteins , rho GTP-Binding Proteins , Intracellular Signaling Peptides and Proteins
6.
Proc Natl Acad Sci U S A ; 121(22): e2400648121, 2024 May 28.
Article En | MEDLINE | ID: mdl-38781210

After central nervous system injury, a rapid cellular and molecular response is induced. This response can be both beneficial and detrimental to neuronal survival in the first few days and increases the risk for neurodegeneration if persistent. Semaphorin4B (Sema4B), a transmembrane protein primarily expressed by cortical astrocytes, has been shown to play a role in neuronal cell death following injury. Our study shows that after cortical stab wound injury, cytokine expression is attenuated in Sema4B-/- mice, and microglia/macrophage reactivity is altered. In vitro, Sema4B enhances the reactivity of microglia following injury, suggesting astrocytic Sema4B functions as a ligand. Moreover, injury-induced microglia reactivity is attenuated in the presence of Sema4B-/- astrocytes compared to Sema4B+/- astrocytes. In vitro experiments indicate that Plexin-B2 is the Sema4B receptor on microglia. Consistent with this, in microglia/macrophage-specific Plexin-B2-/- mice, similar to Sema4B-/- mice, microglial/macrophage reactivity and neuronal cell death are attenuated after cortical injury. Finally, in Sema4B/Plexin-B2 double heterozygous mice, microglial/macrophage reactivity is also reduced after injury, supporting the idea that both Sema4B and Plexin-B2 are part of the same signaling pathway. Taken together, we propose a model in which following injury, astrocytic Sema4B enhances the response of microglia/macrophages via Plexin-B2, leading to increased reactivity.


Astrocytes , Mice, Knockout , Microglia , Nerve Tissue Proteins , Semaphorins , Animals , Microglia/metabolism , Microglia/pathology , Semaphorins/metabolism , Semaphorins/genetics , Astrocytes/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Cell Communication , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/genetics , Mice, Inbred C57BL , Macrophages/metabolism
7.
Biomolecules ; 14(5)2024 May 17.
Article En | MEDLINE | ID: mdl-38786001

During the first and second stages of postnatal development, neocortical neurons exhibit a wide range of spontaneous synchronous activity (SSA). Towards the end of the second postnatal week, the SSA is replaced by a more sparse and desynchronized firing pattern. The developmental desynchronization of neocortical spontaneous neuronal activity is thought to be intrinsically generated, since sensory deprivation from the periphery does not affect the time course of this transition. The extracellular protein reelin controls various aspects of neuronal development through multimodular signaling. However, so far it is unclear whether reelin contributes to the developmental desynchronization transition of neocortical neurons. The present study aims to investigate the role of reelin in postnatal cortical developmental desynchronization using a conditional reelin knockout (RelncKO) mouse model. Conditional reelin deficiency was induced during early postnatal development, and Ca2+ recordings were conducted from organotypic cultures (OTCs) of the somatosensory cortex. Our results show that both wild type (wt) and RelncKO exhibited an SSA pattern during the early postnatal week. However, at the end of the second postnatal week, wt OTCs underwent a transition to a desynchronized network activity pattern, while RelncKO activity remained synchronous. This changing activity pattern suggests that reelin is involved in regulating the developmental desynchronization of cortical neuronal network activity. Moreover, the developmental desynchronization impairment observed in RelncKO was rescued when RelncKO OTCs were co-cultured with wt OTCs. Finally, we show that the developmental transition to a desynchronized state at the end of the second postnatal week is not dependent on glutamatergic signaling. Instead, the transition is dependent on GABAAR and GABABR signaling. The results suggest that reelin controls developmental desynchronization through GABAAR and GABABR signaling.


Extracellular Matrix Proteins , Mice, Knockout , Neocortex , Nerve Tissue Proteins , Reelin Protein , Serine Endopeptidases , Animals , Mice , Neocortex/metabolism , Neocortex/growth & development , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Neurons/metabolism , Nerve Net/metabolism , Nerve Net/growth & development , Somatosensory Cortex/metabolism , Somatosensory Cortex/growth & development
8.
Zool Res ; 45(3): 663-678, 2024 May 18.
Article En | MEDLINE | ID: mdl-38766748

A growing number of studies have demonstrated that repeated exposure to sevoflurane during development results in persistent social abnormalities and cognitive impairment. Davunetide, an active fragment of the activity-dependent neuroprotective protein (ADNP), has been implicated in social and cognitive protection. However, the potential of davunetide to attenuate social deficits following sevoflurane exposure and the underlying developmental mechanisms remain poorly understood. In this study, ribosome and proteome profiles were analyzed to investigate the molecular basis of sevoflurane-induced social deficits in neonatal mice. The neuropathological basis was also explored using Golgi staining, morphological analysis, western blotting, electrophysiological analysis, and behavioral analysis. Results indicated that ADNP was significantly down-regulated following developmental exposure to sevoflurane. In adulthood, anterior cingulate cortex (ACC) neurons exposed to sevoflurane exhibited a decrease in dendrite number, total dendrite length, and spine density. Furthermore, the expression levels of Homer, PSD95, synaptophysin, and vglut2 were significantly reduced in the sevoflurane group. Patch-clamp recordings indicated reductions in both the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs). Notably, davunetide significantly ameliorated the synaptic defects, social behavior deficits, and cognitive impairments induced by sevoflurane. Mechanistic analysis revealed that loss of ADNP led to dysregulation of Ca 2+ activity via the Wnt/ß-catenin signaling, resulting in decreased expression of synaptic proteins. Suppression of Wnt signaling was restored in the davunetide-treated group. Thus, ADNP was identified as a promising therapeutic target for the prevention and treatment of neurodevelopmental toxicity caused by general anesthetics. This study provides important insights into the mechanisms underlying social and cognitive disturbances caused by sevoflurane exposure in neonatal mice and elucidates the regulatory pathways involved.


Animals, Newborn , Cognitive Dysfunction , Proteome , Sevoflurane , Social Behavior , Animals , Sevoflurane/adverse effects , Mice , Cognitive Dysfunction/chemically induced , Ribosomes/drug effects , Ribosomes/metabolism , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/toxicity , Anesthetics, Inhalation/pharmacology , Nerve Tissue Proteins/metabolism , Male , Behavior, Animal/drug effects
9.
Mol Genet Genomic Med ; 12(5): e2469, 2024 May.
Article En | MEDLINE | ID: mdl-38778723

BACKGROUND: Paroxysmal kinesigenic dyskinesia (PKD) is the most prevalent kind type of paroxysmal Dyskinesia, characterized by recurrent and transient episodes of involuntary movements. Most PKD cases were attributed to the proline-rich transmembrane protein 2 (PRRT2) gene, in which the c.649 region is a hotspot for known mutations. Even though some patients with PKD have been genetically diagnosed using whole-exome sequencing (WES) and Sanger sequencing, there are still cases of missed diagnoses due to the limitations of sequencing technology and analytic methods on throughput. METHODS: Patients meeting the diagnosis criteria of PKD with negative results of PRRT2-Sanger sequencing and WES were included in this study. Mutation screening and targeted high-throughput sequencing were performed to analyze and verify the sequencing results of the potential mutations. RESULTS: Six patients with PKD with high mutation ratios of c.649dupC were screened using our targeted high-throughput sequencing from 26 PKD patients with negative results of PRRT2-Sanger sequencing and WES (frequency = 23.1%), which compensated for the comparatively shallow sequencing depth and statistical flaws in this region. Compared with the local normal population and other patients with PKD, the mutation ratios of c.649dupC of these six patients with PKD were much higher and also had truncated protein structures and differentially altered mRNA expression. CONCLUSION: Based on the above studies, we emphasize the routine targeted high-throughput sequencing of the c.649 site in the PRRT2 gene in so-called genetic-testing-negative patients with PKD, and manually calculate the deletion and duplication mutations depth and ratios to lower the rate of clinical misdiagnosis.


Dystonia , Genetic Testing , Membrane Proteins , Nerve Tissue Proteins , Humans , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Female , Male , Dystonia/genetics , Dystonia/diagnosis , Child , Adolescent , Genetic Testing/methods , Genetic Testing/standards , Adult , High-Throughput Nucleotide Sequencing/methods , Mutation , Child, Preschool , Exome Sequencing/methods
10.
Elife ; 132024 May 23.
Article En | MEDLINE | ID: mdl-38780416

Protein phosphorylation is one of the major molecular mechanisms regulating protein activity and function throughout the cell. Pannexin 1 (PANX1) is a large-pore channel permeable to ATP and other cellular metabolites. Its tyrosine phosphorylation and subsequent activation have been found to play critical roles in diverse cellular conditions, including neuronal cell death, acute inflammation, and smooth muscle contraction. Specifically, the non-receptor kinase Src has been reported to phosphorylate Tyr198 and Tyr308 of mouse PANX1 (equivalent to Tyr199 and Tyr309 of human PANX1), resulting in channel opening and ATP release. Although the Src-dependent PANX1 activation mechanism has been widely discussed in the literature, independent validation of the tyrosine phosphorylation of PANX1 has been lacking. Here, we show that commercially available antibodies against the two phosphorylation sites mentioned above-which were used to identify endogenous PANX1 phosphorylation at these two sites-are nonspecific and should not be used to interpret results related to PANX1 phosphorylation. We further provide evidence that neither tyrosine residue is a major phosphorylation site for Src kinase in heterologous expression systems. We call on the field to re-examine the existing paradigm of tyrosine phosphorylation-dependent activation of the PANX1 channel.


Connexins , Nerve Tissue Proteins , src-Family Kinases , Phosphorylation , Connexins/metabolism , Connexins/genetics , Humans , src-Family Kinases/metabolism , src-Family Kinases/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Tyrosine/metabolism , Animals , HEK293 Cells , Mice
11.
Sci Adv ; 10(21): eadk2149, 2024 May 24.
Article En | MEDLINE | ID: mdl-38781326

Understanding the genetic programs that drive neuronal diversification into classes and subclasses is key to understand nervous system development. All neurons can be classified into two types: commissural and ipsilateral, based on whether their axons cross the midline or not. However, the gene regulatory program underlying this binary division is poorly understood. We identified a pair of basic helix-loop-helix transcription factors, Nhlh1 and Nhlh2, as a global transcriptional mechanism that controls the laterality of all floor plate-crossing commissural axons in mice. Mechanistically, Nhlh1/2 play an essential role in the expression of Robo3, the key guidance molecule for commissural axon projections. This genetic program appears to be evolutionarily conserved in chick. We further discovered that Isl1, primarily expressed in ipsilateral neurons within neural tubes, negatively regulates the Robo3 induction by Nhlh1/2. Our findings elucidate a gene regulatory strategy where a conserved global mechanism intersects with neuron class-specific regulators to control the partitioning of neurons based on axon laterality.


Basic Helix-Loop-Helix Transcription Factors , Gene Expression Regulation, Developmental , Neurons , Animals , Neurons/metabolism , Neurons/cytology , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Axons/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Chick Embryo , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Gene Regulatory Networks
12.
Nihon Yakurigaku Zasshi ; 159(3): 165-168, 2024.
Article Ja | MEDLINE | ID: mdl-38692881

Molecular oxygen suffices the ATP production required for the survival of us aerobic organisms. But it is also true that oxygen acts as a source of reactive oxygen species that elicit a spectrum of damages in living organisms. To cope with such intrinsic ambiguity of biological activity oxygen exerts, aerobic mechanisms are equipped with an exquisite adaptive system, which sensitively detects partial pressure of oxygen within the body and controls appropriate oxygen supply to the tissues. Physiological responses to hypoxia are comprised of the acute and chronic phases, in the former of which the oxygen-sensing remains controversial particularly from mechanistic points of view. Recently, we have revealed that the prominently redox-sensitive cation channel TRPA1 plays key roles in oxygen-sensing mechanisms identified in the peripheral tissues and the central nervous system. In this review, we summarize recent development of researches on oxygen-sensing mechanisms including that in the carotid body, which has been recognized as the oxygen receptor organ central to acute oxygen-sensing. We also discuss how ubiquitously the TRPA1 contributes to the mechanisms underlying the acute phase of adaptation to hypoxia.


Oxygen , TRPA1 Cation Channel , Transient Receptor Potential Channels , TRPA1 Cation Channel/metabolism , Humans , Oxygen/metabolism , Animals , Transient Receptor Potential Channels/metabolism , Hypoxia/metabolism , Calcium Channels/metabolism , Nerve Tissue Proteins/metabolism , Reactive Oxygen Species/metabolism , Carotid Body/metabolism
13.
Cereb Cortex ; 34(13): 161-171, 2024 May 02.
Article En | MEDLINE | ID: mdl-38696595

Autism spectrum disorder (ASD) is a developmental disorder with a rising prevalence and unknown etiology presenting with deficits in cognition and abnormal behavior. We hypothesized that the investigation of the synaptic component of prefrontal cortex may provide proteomic signatures that may identify the biological underpinnings of cognitive deficits in childhood ASD. Subcellular fractions of synaptosomes from prefrontal cortices of age-, brain area-, and postmortem-interval-matched samples from children and adults with idiopathic ASD vs. controls were subjected to HPLC-tandem mass spectrometry. Analysis of data revealed the enrichment of ASD risk genes that participate in slow maturation of the postsynaptic density (PSD) structure and function during early brain development. Proteomic analysis revealed down regulation of PSD-related proteins including AMPA and NMDA receptors, GRM3, DLG4, olfactomedins, Shank1-3, Homer1, CaMK2α, NRXN1, NLGN2, Drebrin1, ARHGAP32, and Dock9 in children with autism (FDR-adjusted P < 0.05). In contrast, PSD-related alterations were less severe or unchanged in adult individuals with ASD. Network analyses revealed glutamate receptor abnormalities. Overall, the proteomic data support the concept that idiopathic autism is a synaptopathy involving PSD-related ASD risk genes. Interruption in evolutionarily conserved slow maturation of the PSD complex in prefrontal cortex may lead to the development of ASD in a susceptible individual.


Dorsolateral Prefrontal Cortex , Proteomics , Humans , Child , Male , Female , Adult , Dorsolateral Prefrontal Cortex/metabolism , Child, Preschool , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Synapses/metabolism , Adolescent , Young Adult , Autistic Disorder/metabolism , Autistic Disorder/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Synaptosomes/metabolism , Prefrontal Cortex/metabolism , Post-Synaptic Density/metabolism
14.
Front Immunol ; 15: 1350837, 2024.
Article En | MEDLINE | ID: mdl-38745654

Introduction: Anti-NMDA receptor encephalitis is an autoimmune disorder caused by autoantibodies (abs) against the conformational epitope on GluN1 subunits. GluN1-abs have been determined with cell-based assay (CBA) co-expressing GluN1/GluN2 subunits. However, commercial fixed CBA expressing only GluN1 subunit has increasingly been used in clinical practice. The ab titers can be determined with serial dilutions, but its clinical significance remains unclear. We aimed to develop an H-intensity scale (HIS) score to estimate GluN1-ab titers in cerebrospinal fluid (CSF) with one-time immunostaining using both commercial CBA and immunohistochemistry and report its usefulness. "H" is the initial of a patient with high CSF GluN1-ab titers (1:2,048). Methods: We first determined the reliability of CBA in 370 patients with suspected autoimmune encephalitis by comparing the results between commercial CBA and established assay in Dalmau's Lab. Then, we made positive control panels using the patient H's CSF diluted in a fourfold serial dilution method (1:2, 1:8, 1:32, 1:128, 1:512, and 1:2,048). Based on the panels, we scored the intensity of ab reactivity of 79 GluN1-ab-positive patients' CSF (diluted at 1:2) on a scale from 0 to 6 (with ≥1 considered positive). To assess inter-assay reliability, we performed immunostaining twice in 21 patients' CSF. We investigated an association between the score of CSF obtained at diagnosis and the clinical/paraclinical features. Results: The sensitivity and specificity of CBA were 93.7% (95% CI: 86.0-97.3) and 98.6% (95% CI: 96.5-99.5), respectively. Linear regression analysis showed a good agreement between the scores of the first and second assays. Patients with a typical spectrum, need for mechanical ventilation support, autonomic symptoms/central hypoventilation, dyskinesias, speech dysfunction, decreased level of consciousness, preceding headache, ovarian teratoma, and CSF leukocyte count >20 cells/µL had a higher median HIS score than those without, but HIS score was not associated with sex, age at onset, or seizure. HIS score at diagnosis had a significant effect on 1-year functional status. Discussion: The severity of disease and four of the six core symptoms were associated with higher GluN1-ab titers in CSF at diagnosis, which may play a role in poor 1-year functional status. An incomplete phenotype can be attributed to low CSF GluN1-ab titers.


Anti-N-Methyl-D-Aspartate Receptor Encephalitis , Autoantibodies , Receptors, N-Methyl-D-Aspartate , Humans , Female , Autoantibodies/cerebrospinal fluid , Autoantibodies/immunology , Middle Aged , Adult , Male , Receptors, N-Methyl-D-Aspartate/immunology , Aged , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/diagnosis , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/cerebrospinal fluid , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/immunology , Young Adult , Adolescent , Child , Immunohistochemistry , Child, Preschool , Nerve Tissue Proteins/immunology , Reproducibility of Results , Biomarkers/cerebrospinal fluid , Aged, 80 and over
15.
Sci Immunol ; 9(95): eadi7418, 2024 May 17.
Article En | MEDLINE | ID: mdl-38758807

Immune checkpoint blockade is a promising approach to activate antitumor immunity and improve the survival of patients with cancer. V-domain immunoglobulin suppressor of T cell activation (VISTA) is an immune checkpoint target; however, the downstream signaling mechanisms are elusive. Here, we identify leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) as a VISTA binding partner, which acts as an inhibitory receptor by engaging VISTA and suppressing T cell receptor signaling pathways. Mice with T cell-specific LRIG1 deletion developed superior antitumor responses because of expansion of tumor-specific cytotoxic T lymphocytes (CTLs) with increased effector function and survival. Sustained tumor control was associated with a reduction of quiescent CTLs (TCF1+ CD62Lhi PD-1low) and a reciprocal increase in progenitor and memory-like CTLs (TCF1+ PD-1+). In patients with melanoma, elevated LRIG1 expression on tumor-infiltrating CD8+ CTLs correlated with resistance to immunotherapies. These results delineate the role of LRIG1 as an inhibitory immune checkpoint receptor and propose a rationale for targeting the VISTA/LRIG1 axis for cancer immunotherapy.


B7 Antigens , CD8-Positive T-Lymphocytes , Membrane Glycoproteins , Mice, Inbred C57BL , Animals , Mice , CD8-Positive T-Lymphocytes/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/genetics , Humans , B7 Antigens/immunology , B7 Antigens/genetics , Mice, Knockout , Cell Line, Tumor , Female , Membrane Proteins , Nerve Tissue Proteins
16.
ACS Chem Neurosci ; 15(10): 2018-2027, 2024 May 15.
Article En | MEDLINE | ID: mdl-38701380

In silico modeling was applied to study the efficiency of two ligands, namely, UCB-J and UCB-F, to bind to isoforms of the synaptic vesicle glycoprotein 2 (SV2) that are involved in the regulation of synaptic function in the nerve terminals, with the ultimate goal to understand the selectivity of the interaction between UCB-J and UCB-F to different isoforms of SV2. Docking and large-scale molecular dynamics simulations were carried out to unravel various binding patterns, types of interactions, and binding free energies, covering hydrogen bonding and nonspecific hydrophobic interactions, water bridge, π-π, and cation-π interactions. The overall preference for bonding types of UCB-J and UCB-F with particular residues in the protein pockets can be disclosed in detail. A unique interaction fingerprint, namely, hydrogen bonding with additional cation-π interaction with the pyridine moiety of UCB-J, could be established as an explanation for its high selectivity over the SV2 isoform A (SV2A). Other molecular details, primarily referring to the presence of π-π interactions and hydrogen bonding, could also be analyzed as sources of selectivity of the UCB-F tracer for the three isoforms. The simulations provide atomic details to support future development of new selective tracers targeting synaptic vesicle glycoproteins and their associated diseases.


Membrane Glycoproteins , Molecular Dynamics Simulation , Nerve Tissue Proteins , Protein Isoforms , Ligands , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/chemistry , Humans , Hydrogen Bonding , Protein Binding/physiology , Molecular Docking Simulation/methods , Synaptic Vesicles/metabolism
17.
PLoS Biol ; 22(5): e3002599, 2024 May.
Article En | MEDLINE | ID: mdl-38713721

Synaptic adhesion molecules (SAMs) are evolutionarily conserved proteins that play an important role in the form and function of neuronal synapses. Teneurins (Tenms) and latrophilins (Lphns) are well-known cell adhesion molecules that form a transsynaptic complex. Recent studies suggest that Tenm3 and Lphn2 (gene symbol Adgrl2) are involved in hippocampal circuit assembly via their topographical expression. However, it is not known whether other teneurins and latrophilins display similar topographically restricted expression patterns during embryonic and postnatal development. Here, we reveal the cartography of all teneurin (Tenm1-4) and latrophilin (Lphn1-3 [Adgrl1-3]) paralog expression in the mouse hippocampus across prenatal and postnatal development as monitored by large-scale single-molecule RNA in situ hybridization mapping. Our results identify a striking heterogeneity in teneurin and latrophilin expression along the spatiotemporal axis of the hippocampus. Tenm2 and Tenm4 expression levels peak at the neonatal stage when compared to Tenm1 and Tenm3, while Tenm1 expression is restricted to the postnatal pyramidal cell layer. Tenm4 expression in the dentate gyrus (DG) exhibits an opposing topographical expression pattern in the embryonic and neonatal hippocampus. Our findings were validated by analyses of multiple RNA-seq datasets at bulk, single-cell, and spatial levels. Thus, our study presents a comprehensive spatiotemporal map of Tenm and Lphn expression in the hippocampus, showcasing their diverse expression patterns across developmental stages in distinct spatial axes.


Gene Expression Regulation, Developmental , Hippocampus , Nerve Tissue Proteins , Receptors, Peptide , Animals , Hippocampus/metabolism , Hippocampus/embryology , Mice , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Receptors, Peptide/metabolism , Receptors, Peptide/genetics , Female , Mice, Inbred C57BL , Male , Tenascin , Receptors, G-Protein-Coupled
18.
Nat Commun ; 15(1): 4132, 2024 May 16.
Article En | MEDLINE | ID: mdl-38755165

The regulated release of chemical messengers is crucial for cell-to-cell communication; abnormalities in which impact coordinated human body function. During vesicular secretion, multiple SNARE complexes assemble at the release site, leading to fusion pore opening. How membrane fusion regulators act on heterogeneous SNARE populations to assemble fusion pores in a timely and synchronized manner, is unknown. Here, we demonstrate the role of SNARE chaperones Munc13-1 and Munc18-1 in rescuing individual nascent fusion pores from their diacylglycerol lipid-mediated inhibitory states. At the onset of membrane fusion, Munc13-1 clusters multiple SNARE complexes at the release site and synchronizes release events, while Munc18-1 stoichiometrically interacts with trans-SNARE complexes to enhance N- to C-terminal zippering. When both Munc proteins are present simultaneously, they differentially access dynamic trans-SNARE complexes to regulate pore properties. Overall, Munc proteins' direct action on fusion pore assembly indicates their role in controlling quantal size during vesicular secretion.


Membrane Fusion , Munc18 Proteins , Nerve Tissue Proteins , SNARE Proteins , Munc18 Proteins/metabolism , Munc18 Proteins/genetics , SNARE Proteins/metabolism , SNARE Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Animals , Humans , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Rats
19.
Sci Adv ; 10(20): eadi7024, 2024 May 17.
Article En | MEDLINE | ID: mdl-38758791

At the synapse, presynaptic neurotransmitter release is tightly controlled by release machinery, involving the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and Munc13. The Ca2+ sensor Doc2 cooperates with Munc13 to regulate neurotransmitter release, but the underlying mechanisms remain unclear. In our study, we have characterized the binding mode between Doc2 and Munc13 and found that Doc2 originally occludes Munc13 to inhibit SNARE complex assembly. Moreover, our investigation unveiled that EphB2, a presynaptic adhesion molecule (SAM) with inherent tyrosine kinase functionality, exhibits the capacity to phosphorylate Doc2. This phosphorylation attenuates Doc2 block on Munc13 to promote SNARE complex assembly, which functionally induces spontaneous release and synaptic augmentation. Consistently, application of a Doc2 peptide that interrupts Doc2-Munc13 interplay impairs excitatory synaptic transmission and leads to dysfunction in spatial learning and memory. These data provide evidence that SAMs modulate neurotransmitter release by controlling SNARE complex assembly.


Calcium-Binding Proteins , Nerve Tissue Proteins , Neurotransmitter Agents , Receptor, EphB2 , SNARE Proteins , Synaptic Transmission , SNARE Proteins/metabolism , Animals , Neurotransmitter Agents/metabolism , Nerve Tissue Proteins/metabolism , Phosphorylation , Receptor, EphB2/metabolism , Receptor, EphB2/genetics , Calcium-Binding Proteins/metabolism , Protein Binding , Humans , Mice , Rats
20.
PLoS One ; 19(5): e0299380, 2024.
Article En | MEDLINE | ID: mdl-38748694

Autism Spectrum Disorder (ASD) is a neurodevelopmental behavioral disorder characterized by social, communicative, and motor deficits. There is no single etiological cause for ASD, rather, there are various genetic and environmental factors that increase the risk for ASD. It is thought that some of these factors influence the same underlying neural mechanisms, and that an interplay of both genetic and environmental factors would better explain the pathogenesis of ASD. To better appreciate the influence of genetic-environment interaction on ASD-related behaviours, rats lacking a functional copy of the ASD-linked gene Cntnap2 were exposed to maternal immune activation (MIA) during pregnancy and assessed in adolescence and adulthood. We hypothesized that Cntnap2 deficiency interacts with poly I:C MIA to aggravate ASD-like symptoms in the offspring. In this double-hit model, we assessed attention, a core deficit in ASD due to prefrontal cortical dysfunction. We employed a well-established attentional paradigm known as the 5-choice serial reaction time task (5CSRTT). Cntnap2-/- rats exhibited greater perseverative responses which is indicative of repetitive behaviors. Additionally, rats exposed to poly I:C MIA exhibited premature responses, a marker of impulsivity. The rats exposed to both the genetic and environmental challenge displayed an increase in impulsive activity; however, this response was only elicited in the presence of an auditory distractor. This implies that exacerbated symptomatology in the double-hit model may situation-dependent and not generally expressed.


Attention , Autism Spectrum Disorder , Disease Models, Animal , Gene-Environment Interaction , Nerve Tissue Proteins , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/etiology , Rats , Female , Attention/physiology , Pregnancy , Nerve Tissue Proteins/genetics , Male , Membrane Proteins/genetics , Poly I-C , Behavior, Animal , Prenatal Exposure Delayed Effects/genetics
...