Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 603
Filtrar
1.
Nat Med ; 30(9): 2679-2691, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39095595

RESUMEN

While single-cell technologies have greatly advanced our comprehension of human brain cell types and functions, studies including large numbers of donors and multiple brain regions are needed to extend our understanding of brain cell heterogeneity. Integrating atlas-level single-cell data presents a chance to reveal rare cell types and cellular heterogeneity across brain regions. Here we present the Brain Cell Atlas, a comprehensive reference atlas of brain cells, by assembling single-cell data from 70 human and 103 mouse studies of the brain throughout major developmental stages across brain regions, covering over 26.3 million cells or nuclei from both healthy and diseased tissues. Using machine-learning based algorithms, the Brain Cell Atlas provides a consensus cell type annotation, and it showcases the identification of putative neural progenitor cells and a cell subpopulation of PCDH9high microglia in the human brain. We demonstrate the gene regulatory difference of PCDH9high microglia between hippocampus and prefrontal cortex and elucidate the cell-cell communication network. The Brain Cell Atlas presents an atlas-level integrative resource for comparing brain cells in different environments and conditions within the Human Cell Atlas.


Asunto(s)
Encéfalo , Cadherinas , Análisis de la Célula Individual , Transcriptoma , Humanos , Encéfalo/citología , Encéfalo/metabolismo , Ratones , Animales , Cadherinas/genética , Cadherinas/metabolismo , Microglía/metabolismo , Microglía/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Protocadherinas , Atlas como Asunto , Hipocampo/citología , Hipocampo/metabolismo , Aprendizaje Automático , Comunicación Celular/genética
2.
Artículo en Ruso | MEDLINE | ID: mdl-39113443

RESUMEN

Mutations in the human PCDH19 gene lead to epileptic encephalopathy of early childhood. It is characterized by the early onset of serial seizures, cognitive impairment and behavioral disorders (including autistic personality traits). In most cases, difficulties arise in selecting therapy due to pharmacoresistance. The pathogenesis of the disease is complex. The data available to us at the moment from numerous studies present the pathogenesis of «PCDH19 syndrome¼ as multi-level, affecting both the epigenetic support of cell life, and development of stem cells and progenitor cells in the process of neuroontogenesis, and the influence on the neurotransmitter mechanisms of the brain, and disruption of the formation of neural networks with an inevitable increase in the excitability of the cerebral cortex as a whole, and local changes in the highly labile regulatory structures of the hippocampal region. And it is not surprising that all these changes entail not only (and perhaps not so much) epileptization, but a profound disruption of the regulation of brain activity, accompanied by autism spectrum disorders, more profound disorders in the form of schizophrenia or cyclothymia, and the formation of delayed psychomotor development. A «side branch¼ of these pathogenetic processes can also be considered the participation of PCDH19 dysfunctions in certain variants of oncogenesis. The need for polypharmacy (in most cases) confirms the diversity of mechanisms involved in the pathogenesis of the disease and makes the prospects for the development of effective and rational treatment regimens very vague. Cautious optimism is caused only by attempts at relatively specific treatment with ganaxolone.


Asunto(s)
Epilepsia , Polifarmacia , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/tratamiento farmacológico , Encéfalo , Cadherinas/genética , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Mutación , Protocadherinas
3.
Mol Brain ; 17(1): 58, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39175067

RESUMEN

Protocadherin 19 (Pcdh19) is a homophilic cell adhesion molecule and is involved in a variety of neuronal functions. Here, we tested whether Pcdh19 has a regulatory role in axon guidance using the developing Xenopus retinotectal system. We performed targeted microinjections of a translation blocking antisense morpholino oligonucleotide to knock down the expression of Pcdh19 selectively in the central nervous system. Knocking down Pcdh19 expression resulted in navigational errors of retinal ganglion cell (RGC) axons specifically at the optic chiasm. Instead of projecting to the contralateral optic tectum, RGC axons in the Pcdh19-depleted embryo misprojected ipsilaterally. Although incorrectly delivered into the ipsilateral brain hemisphere, these axons correctly reached the optic tectum. These data suggest that Pcdh19 has a critical role in preventing mixing of RGC axons originating from the opposite eyes at the optic chiasm, highlighting the importance of cell adhesion in bundling of RGC axons.


Asunto(s)
Orientación del Axón , Axones , Cadherinas , Protocadherinas , Células Ganglionares de la Retina , Proteínas de Xenopus , Xenopus laevis , Animales , Cadherinas/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas de Xenopus/genética , Células Ganglionares de la Retina/metabolismo , Xenopus laevis/embriología , Axones/metabolismo , Retina/metabolismo , Retina/embriología , Vías Visuales , Técnicas de Silenciamiento del Gen , Quiasma Óptico/embriología , Quiasma Óptico/metabolismo , Colículos Superiores/embriología , Colículos Superiores/metabolismo , Regulación del Desarrollo de la Expresión Génica
4.
PLoS One ; 19(8): e0309536, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39208202

RESUMEN

Biomarkers for classifying and grading gliomas have been extensively explored, whereas populations in public databases were mostly Western/European. Based on public databases cannot accurately represent Chinese population. To identify molecular characteristics associated with clinical outcomes of lower-grade glioma (LGG) and glioblastoma (GBM) in the Chinese population, we performed whole-exome sequencing (WES) in 16 LGG and 35 GBM tumor tissues. TP53 (36/51), TERT (31/51), ATRX (16/51), EFGLAM (14/51), and IDH1 (13/51) were the most common genes harboring mutations. IDH1 mutation (c.G395A; p.R132H) was significantly enriched in LGG, whereas PCDHGA10 mutation (c.A265G; p.I89V) in GBM. IDH1-wildtype and PCDHGA10 mutation were significantly related to poor prognosis. IDH1 is an important biomarker in gliomas, whereas PCDHGA10 mutation has not been reported to correlate with gliomas. Different copy number variations (CNVs) and oncogenic signaling pathways were identified between LGG and GBM. Differential genomic landscapes between LGG and GBM were revealed in the Chinese population, and PCDHGA10, for the first time, was identified as the prognostic factor of gliomas. Our results might provide a basis for molecular classification and identification of diagnostic biomarkers and even potential therapeutic targets for gliomas.


Asunto(s)
Neoplasias Encefálicas , Variaciones en el Número de Copia de ADN , Glioblastoma , Glioma , Isocitrato Deshidrogenasa , Mutación , Humanos , Glioblastoma/genética , Glioblastoma/patología , Masculino , Femenino , Isocitrato Deshidrogenasa/genética , Glioma/genética , Glioma/patología , Persona de Mediana Edad , Adulto , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Secuenciación del Exoma , Telomerasa/genética , Biomarcadores de Tumor/genética , Clasificación del Tumor , Cadherinas/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína p53 Supresora de Tumor/genética , Anciano , Pronóstico , Genómica/métodos , Protocadherinas , Adulto Joven
5.
Nature ; 632(8025): 614-621, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39048821

RESUMEN

Western equine encephalitis virus (WEEV) is an arthropod-borne virus (arbovirus) that frequently caused major outbreaks of encephalitis in humans and horses in the early twentieth century, but the frequency of outbreaks has since decreased markedly, and strains of this alphavirus isolated in the past two decades are less virulent in mammals than strains isolated in the 1930s and 1940s1-3. The basis for this phenotypic change in WEEV strains and coincident decrease in epizootic activity (known as viral submergence3) is unclear, as is the possibility of re-emergence of highly virulent strains. Here we identify protocadherin 10 (PCDH10) as a cellular receptor for WEEV. We show that multiple highly virulent ancestral WEEV strains isolated in the 1930s and 1940s, in addition to binding human PCDH10, could also bind very low-density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2), which are recognized by another encephalitic alphavirus as receptors4. However, whereas most of the WEEV strains that we examined bind to PCDH10, a contemporary strain has lost the ability to recognize mammalian PCDH10 while retaining the ability to bind avian receptors, suggesting WEEV adaptation to a main reservoir host during enzootic circulation. PCDH10 supports WEEV E2-E1 glycoprotein-mediated infection of primary mouse cortical neurons, and administration of a soluble form of PCDH10 protects mice from lethal WEEV challenge. Our results have implications for the development of medical countermeasures and for risk assessment for re-emerging WEEV strains.


Asunto(s)
Virus de la Encefalitis Equina del Oeste , Especificidad del Huésped , Protocadherinas , Receptores Virales , Animales , Femenino , Humanos , Masculino , Ratones , Aves/metabolismo , Aves/virología , Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/virología , Virus de la Encefalitis Equina del Oeste/clasificación , Virus de la Encefalitis Equina del Oeste/metabolismo , Virus de la Encefalitis Equina del Oeste/patogenicidad , Encefalomielitis Equina/epidemiología , Encefalomielitis Equina/virología , Proteínas Relacionadas con Receptor de LDL/metabolismo , Neuronas/metabolismo , Neuronas/virología , Fenotipo , Protocadherinas/metabolismo , Receptores de LDL/metabolismo , Receptores de LDL/genética , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Zoonosis Virales/epidemiología , Zoonosis Virales/virología
6.
Biochem Biophys Res Commun ; 732: 150431, 2024 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-39047401

RESUMEN

Brain metastasis (BM) is one of the main causes of death in patients with non-small cell lung carcinoma. The specific pathological processes of BM, which are inextricably linked to the brain tumor microenvironment, such as the abundance of astrocytes, lead to limited treatment options and poor prognosis. Reactive astrocytes are acquired in the BM; however, the underlying mechanisms remain unclear. This study aimed to explore the mechanisms by which astrocytes promote BM development. We determined the crucial role of reactive astrocytes in promoting the proliferation and migration of brain metastatic lung tumor cells by upregulating protocadherin 1 (PCDH1) expression in an in vitro co-culture model. The overexpression of PCDH1 was confirmed in clinical BM samples using immunohistochemical staining. Survival analysis indicated that high-PCDH1 expression was associated with poor survival in patients with lung adenocarcinoma. In vivo assays further showed that silence of PCDH1 effectively inhibited the tumor progression of brain metastases and prolonged the survival of animals. RNA sequencing has revealed that PCDH1 plays an important role in cell proliferation and adhesion. In conclusion, the present study revealed the promoting role of astrocytes in enhancing the aggressive phenotype of brain metastatic tumor cells by regulating the expression of PCDH1, which might be a biomarker for BM diagnosis and prognosis, suggesting the potential efficacy of targeting important astrocyte-tumor interactions in the treatment of patients with non-small cell lung carcinoma with BM.


Asunto(s)
Astrocitos , Neoplasias Encefálicas , Cadherinas , Proliferación Celular , Neoplasias Pulmonares , Protocadherinas , Regulación hacia Arriba , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Humanos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Astrocitos/metabolismo , Astrocitos/patología , Cadherinas/metabolismo , Cadherinas/genética , Animales , Proliferación Celular/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Ratones , Masculino , Ratones Desnudos , Movimiento Celular/genética , Ratones Endogámicos BALB C , Femenino
7.
F S Sci ; 5(3): 283-292, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38942387

RESUMEN

OBJECTIVE: To study the identification of rare genetic variants in the PCDH genetic family in a cohort of transgender women (TGW) and their potential role in gender identity. DESIGN: Exome sequencing and functional ontology analysis. SETTING: Outpatient gender health and reproductive endocrinology clinics. PATIENT(S): A total of 24 TGW and 22 cisgender men (CM). INTERVENTION(S): Exome sequencing followed by variant confirmation through Sanger sequencing and functional classification analysis using the Database for Annotation, Visualization, and Integrated Discovery tool. MAIN OUTCOME MEASURE(S): Identification of rare, functionally significant genetic variants in the PCDH gene family and their prevalence in TGW compared with CM. RESULT(S): Exome sequencing revealed 38,524 genetic variants, of which 2,441 were rare and predicted to be functionally significant. The Database for Annotation, Visualization, and Integrated Discovery analysis demonstrated a statistically enriched functional group, "homophilic cell adhesion via plasma membrane adhesion molecules," containing 55 genes, including 18 PCDH gene family members. A total of 37 rare variants in 21 PCDH genes were identified, with 36 confirmed using Sanger sequencing. A statistically significant increase in these variants was observed in TGW compared with CM (Z = 2.08905). CONCLUSION(S): Transgender women exhibited a greater than threefold increase in functionally significant PCDH gene variants compared with CM. These findings suggest that the PCDH family may play a role in the genetic pathways associated with gender identity in TGW.


Asunto(s)
Cadherinas , Variación Genética , Personas Transgénero , Humanos , Femenino , Cadherinas/genética , Masculino , Variación Genética/genética , Adulto , Estudios de Cohortes , Secuenciación del Exoma , Identidad de Género , Protocadherinas
8.
Open Biol ; 14(6): 240113, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38889770

RESUMEN

Autism spectrum disorder (ASD) is a group of neurodevelopmental conditions associated with deficits in social interaction and communication, together with repetitive behaviours. The cell adhesion molecule protocadherin10 (PCDH10) is linked to ASD in humans. Pcdh10 is expressed in the nervous system during embryonic and early postnatal development and is important for neural circuit formation. In mice, strong expression of Pcdh10 in the ganglionic eminences and in the basolateral complex (BLC) of the amygdala was observed at mid and late embryonic stages, respectively. Both inhibitory and excitatory neurons expressed Pcdh10 in the BLC at perinatal stages and vocalization-related genes were enriched in Pcdh10-expressing neurons in adult mice. An epitope-tagged Pcdh10-HAV5 mouse line revealed endogenous interactions of PCDH10 with synaptic proteins in the young postnatal telencephalon. Nuanced socio-affective communication changes in call emission rates, acoustic features and call subtype clustering were primarily observed in heterozygous pups of a conditional knockout (cKO) with selective deletion of Pcdh10 in Gsh2-lineage interneurons. These changes were less prominent in heterozygous ubiquitous Pcdh10 KO pups, suggesting that altered anxiety levels associated with Gsh2-lineage interneuron functioning might drive the behavioural effects. Together, loss of Pcdh10 specifically in interneurons contributes to behavioural alterations in socio-affective communication with relevance to ASD.


Asunto(s)
Amígdala del Cerebelo , Cadherinas , Interneuronas , Ratones Noqueados , Protocadherinas , Animales , Cadherinas/metabolismo , Cadherinas/genética , Interneuronas/metabolismo , Ratones , Protocadherinas/metabolismo , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/crecimiento & desarrollo , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/genética , Vocalización Animal/fisiología , Masculino , Conducta Social
9.
Int J Mol Sci ; 25(11)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38891919

RESUMEN

Developmental and epileptic encephalopathy-9 (DEE9) is characterized by seizure onset in infancy, mild to severe intellectual impairment, and psychiatric features and is caused by a mutation in the PCDH19 gene on chromosome Xq22. The rare, unusual X-linked type of disorder affects heterozygous females and mosaic males; transmitting males are unaffected. In our study, 165 patients with epilepsy were tested by Next Generation Sequencing (NGS)-based panel and exome sequencing using Illumina technology. PCDH19 screening identified three point mutations, one indel, and one 29 bp-long deletion in five unrelated female probands. Two novel mutations, c.1152_1180del (p.Gln385Serfs*6) and c.830_831delinsAA (p.Phe277*), were identified and found to be de novo pathogenic. Moreover, among the three inherited mutations, two originated from asymptomatic mothers and one from an affected father. The PCDH19 c.1682C>T and c.1711G>T mutations were present in the DNA samples of asymptomatic mothers. After targeted parental testing, X chromosome inactivation tests and Sanger sequencing were carried out for mosaicism examination on maternal saliva samples in the two asymptomatic PCDH19 mutation carrier subjects. Tissue mosaicism and X-inactivation tests were negative. Our results support the opportunity for reduced penetrance in DEE9 and contribute to expanding the genotype-phenotype spectrum of PCDH19-related epilepsy.


Asunto(s)
Cadherinas , Epilepsia , Secuenciación de Nucleótidos de Alto Rendimiento , Mutación , Protocadherinas , Humanos , Femenino , Cadherinas/genética , Epilepsia/genética , Linaje , Masculino , Preescolar , Niño , Lactante , Edad de Inicio
10.
Genes (Basel) ; 15(6)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38927638

RESUMEN

Approximately half of the cases of chronic kidney disease (CKD) in childhood are caused by congenital anomalies of the kidney and urinary tract (CAKUT). Specific genes were identified as having significant importance in regard to the underlying genetic factors responsible for the CAKUT phenotype, and in our research, we focused on analyzing and comparing the expression levels of ectodysplasin A2 receptor (EDA2R), protocadherin9 (PCDH9), and TNF receptor-associated factor 7 (TRAF7) proteins in the cortex and medulla of healthy control kidneys during developmental phases 2, 3, and 4. We also performed an analysis of the area percentages of the mentioned proteins in the cortical and medullary sections of healthy embryonic and fetal kidneys compared to those affected by CAKUT, including duplex kidneys (DK), horseshoe kidneys (HK), hypoplastic kidneys (HYP), and dysplastic kidneys (DYS). We found that the CAKUT candidate gene proteins EDA2R, PCDH9, and TRAF7 are all expressed during normal human kidney development stages. In DYS, the expression of EDA2R was higher than in normal kidneys, likely due to EDA2R's role in apoptosis, which was upregulated in specific cases and could possibly contribute to the formation of DYS. The expression of PCDH9 was lower in HK, which can be attributed to the possible role of PCDH9 in cell migration suppression. Decreased PCDH9 expression is linked to increased cell migration, potentially contributing to the development of HK. The level of TRAF7 expression was reduced in all examined kidney disorders compared to normal kidneys, suggesting that this reduction might be attributed to the crucial role of TRAF7 in the formation of endothelium and ciliogenesis, both of which are essential for normal kidney development. Further research is required to ascertain the function of these proteins in both the typical development of the kidney and in CAKUT.


Asunto(s)
Cadherinas , Riñón , Anomalías Urogenitales , Reflujo Vesicoureteral , Humanos , Cadherinas/genética , Cadherinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Riñón/metabolismo , Riñón/anomalías , Riñón/crecimiento & desarrollo , Riñón/embriología , Protocadherinas , Sistema Urinario/anomalías , Sistema Urinario/metabolismo , Anomalías Urogenitales/genética , Anomalías Urogenitales/patología , Reflujo Vesicoureteral/genética , Reflujo Vesicoureteral/patología
11.
Proc Natl Acad Sci U S A ; 121(21): e2321388121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38748583

RESUMEN

Protocadherin19 (PCDH19)-related epilepsy syndrome is a rare disorder characterized by early-onset epilepsy, intellectual disability, and autistic behaviors. PCDH19 is located on the X chromosome and encodes a calcium-dependent single-pass transmembrane protein, which regulates cell-to-cell adhesion through homophilic binding. In human, 90% of heterozygous females, containing PCDH19 wild-type and mutant cells due to random X inactivation, are affected, whereas mutant males, containing only mutant cells, are typically not. The current view, the cellular interference, is that the altered interactions between wild-type and mutant cells during development, rather than loss of function itself, are responsible. However, studies using Pcdh19 knockout mice showed that the complete loss of function also causes autism-like behaviors both in males and females, suggesting that other functions of PCDH19 may also contribute to pathogenesis. To address whether mosaicism is required for PCDH19-related epilepsy, we generated Xenopus tropicalis tadpoles with complete or mosaic loss of function by injecting antisense morpholino oligonucleotides into the blastomeres of neural lineage at different stages of development. We found that either mosaic or complete knockdown results in seizure-like behaviors, which could be rescued by antiseizure medication, and repetitive behaviors. Our results suggest that the loss of PCDH19 function itself, in addition to cellular interference, may also contribute to PCDH19-related epilepsy.


Asunto(s)
Cadherinas , Epilepsia , Protocadherinas , Animales , Femenino , Masculino , Conducta Animal , Cadherinas/genética , Cadherinas/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Mosaicismo , Xenopus
12.
J Mol Med (Berl) ; 102(7): 899-912, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38739269

RESUMEN

Immune checkpoint inhibitors (ICIs) have achieved impressive success in lung adenocarcinoma (LUAD). However, the response to ICIs varies among patients, and predictive biomarkers are urgently needed. PCDH11X is frequently mutated in LUAD, while its role in ICI treatment is unclear. In this study, we curated genomic and clinical data of 151 LUAD patients receiving ICIs from three independent cohorts. Relations between PCDH11X and treatment outcomes of ICIs were examined. A melanoma cohort collected from five published studies, a pan-cancer cohort, and non-ICI-treated TCGA-LUAD cohort were also examined to investigate whether PCDH11X mutation is a specific predictive biomarker for LUAD ICI treatment. Among the three ICI-treated LUAD cohorts, PCDH11X mutation (PCDH11X-MUT) was associated with better clinical response compared to wild-type PCDH11X (PCDH11X-WT). While in ICI-treated melanoma cohort, the pan-cancer cohort excluding LUAD, and the non-ICI-treated TCGA-LUAD cohort, no significant differences in overall survival (OS) were observed between the PCDH11X-MUT and PCDH11X-WT groups. PCDH11X mutation was associated with increased PD-L1 expression, tumor mutation burden (TMB), neoantigen load, DNA damage repair (DDR) mutations, and hot tumor microenvironment in TCGA-LUAD cohort. Our findings suggested that the PCDH11X mutation might serve as a specific biomarker to predict the efficacy of ICIs for LUAD patients. Considering the relatively small sample size of ICI-treated cohorts, future research with larger cohorts and prospective clinical trials will be essential for validating and further exploring the role of PCDH11X mutation in the context of immunotherapy outcomes in LUAD. KEY MESSAGES: PCDH11X mutation is associated with better clinical response compared to wild type PCDH11X in three ICIs-treated LUAD cohorts. In ICIs-treated melanoma cohort, the pan-cancer cohort excluding LUAD, and non-ICIs-treated TCGA-LUAD cohorts PCDH11X mutation is not associated with better clinical response, suggesting PCDH11X mutation might be a specific biomarker to predict the efficacy of ICIs treatment for LUAD patients. PCDH11X mutation is associated with increased PD-L1 expression, tumor mutation burden, and neoantigen load in TCGA-LUAD cohort. PCDH11X mutation is associated with hot tumor microenvironment in TCGA-LUAD cohort.


Asunto(s)
Adenocarcinoma del Pulmón , Biomarcadores de Tumor , Cadherinas , Inhibidores de Puntos de Control Inmunológico , Neoplasias Pulmonares , Mutación , Protocadherinas , Humanos , Biomarcadores de Tumor/genética , Cadherinas/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Femenino , Masculino , Persona de Mediana Edad , Anciano , Adulto , Pronóstico , Estudios de Cohortes
13.
Transl Psychiatry ; 14(1): 216, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38806495

RESUMEN

Genetic factors significantly affect the pathogenesis of psychiatric disorders. However, the specific pathogenic mechanisms underlying these effects are not fully understood. Recent extensive genomic studies have implicated the protocadherin-related 15 (PCDH15) gene in the onset of psychiatric disorders, such as bipolar disorder (BD). To further investigate the pathogenesis of these psychiatric disorders, we developed a mouse model lacking Pcdh15. Notably, although PCDH15 is primarily identified as the causative gene of Usher syndrome, which presents with visual and auditory impairments, our mice with Pcdh15 homozygous deletion (Pcdh15-null) did not exhibit observable structural abnormalities in either the retina or the inner ear. The Pcdh15-null mice showed very high levels of spontaneous motor activity which was too disturbed to perform standard behavioral testing. However, the Pcdh15 heterozygous deletion mice (Pcdh15-het) exhibited enhanced spontaneous locomotor activity, reduced prepulse inhibition, and diminished cliff avoidance behavior. These observations agreed with the symptoms observed in patients with various psychiatric disorders and several mouse models of psychiatric diseases. Specifically, the hyperactivity may mirror the manic episodes in BD. To obtain a more physiological, long-term quantification of the hyperactive phenotype, we implanted nano tag® sensor chips in the animals, to enable the continuous monitoring of both activity and body temperature. During the light-off period, Pcdh15-null exhibited elevated activity and body temperature compared with wild-type (WT) mice. However, we observed a decreased body temperature during the light-on period. Comprehensive brain activity was visualized using c-Fos mapping, which was assessed during the activity and temperature peak and trough. There was a stark contrast between the distribution of c-Fos expression in Pcdh15-null and WT brains during both the light-on and light-off periods. These results provide valuable insights into the neural basis of the behavioral and thermal characteristics of Pcdh15-deletion mice. Therefore, Pcdh15-deletion mice can be a novel model for BD with mania and other psychiatric disorders, with a strong genetic component that satisfies both construct and surface validity.


Asunto(s)
Trastorno Bipolar , Temperatura Corporal , Cadherinas , Modelos Animales de Enfermedad , Locomoción , Ratones Noqueados , Animales , Masculino , Ratones , Conducta Animal , Trastorno Bipolar/genética , Trastorno Bipolar/fisiopatología , Cadherinas/genética , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Locomoción/genética , Ratones Endogámicos C57BL , Inhibición Prepulso/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Protocadherinas
14.
Zool Res ; 45(3): 535-550, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38747058

RESUMEN

Proper regulation of synapse formation and elimination is critical for establishing mature neuronal circuits and maintaining brain function. Synaptic abnormalities, such as defects in the density and morphology of postsynaptic dendritic spines, underlie the pathology of various neuropsychiatric disorders. Protocadherin 17 (PCDH17) is associated with major mood disorders, including bipolar disorder and depression. However, the molecular mechanisms by which PCDH17 regulates spine number, morphology, and behavior remain elusive. In this study, we found that PCDH17 functions at postsynaptic sites, restricting the number and size of dendritic spines in excitatory neurons. Selective overexpression of PCDH17 in the ventral hippocampal CA1 results in spine loss and anxiety- and depression-like behaviors in mice. Mechanistically, PCDH17 interacts with actin-relevant proteins and regulates actin filament (F-actin) organization. Specifically, PCDH17 binds to ROCK2, increasing its expression and subsequently enhancing the activity of downstream targets such as LIMK1 and the phosphorylation of cofilin serine-3 (Ser3). Inhibition of ROCK2 activity with belumosudil (KD025) ameliorates the defective F-actin organization and spine structure induced by PCDH17 overexpression, suggesting that ROCK2 mediates the effects of PCDH17 on F-actin content and spine development. Hence, these findings reveal a novel mechanism by which PCDH17 regulates synapse development and behavior, providing pathological insights into the neurobiological basis of mood disorders.


Asunto(s)
Citoesqueleto de Actina , Cadherinas , Espinas Dendríticas , Protocadherinas , Quinasas Asociadas a rho , Animales , Ratones , Citoesqueleto de Actina/metabolismo , Cadherinas/metabolismo , Cadherinas/genética , Espinas Dendríticas/metabolismo , Espinas Dendríticas/fisiología , Regulación de la Expresión Génica , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/genética , Protocadherinas/genética , Protocadherinas/metabolismo
15.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673968

RESUMEN

The pathogenesis of IgAV, the most common systemic vasculitis in childhood, appears to be complex and requires further elucidation. We aimed to investigate the potential role of galactose-deficient immunoglobulin A1 (Gd-IgA1), high-mobility group box 1 (HMGB1), receptor for advanced glycation end products (RAGE) and protocadherin 1 (PCDH1) in the pathogenesis of IgAV. Our prospective study enrolled 86 patients with IgAV and 70 controls. HMGB1, RAGE, Gd-IgA1 and PCDH1 in serum and urine were determined by the enzyme-linked immunosorbent assay (ELISA) method at the onset of the disease and after a six-month interval in patients and once in the control group. Serum concentrations of HMGB1, RAGE and PCDH1 and urinary concentrations of HMGB1, RAGE, Gd-IgA1 and PCDH1 were significantly higher in patients with IgAV than in the control group (p < 0.001). Concentrations of HMGB1 (5573 pg/mL vs. 3477 pg/mL vs. 1088 pg/mL, p < 0.001) and RAGE (309 pg/mL vs. 302.4 pg/mL vs. 201.3 pg/mL, p = 0.012) in the serum of patients remained significantly elevated when the disease onset was compared with the six-month follow-up interval, and thus could be a potential marker of disease activity. Urinary concentration of HMGB1 measured in the follow-up period was higher in patients with nephritis compared to IgAV without nephritis (270.9 (146.7-542.7) ng/mmol vs. 133.2 (85.9-318.6) ng/mmol, p = 0.049) and significantly positively correlated with the urine albumine to creatinine ratio (τ = 0.184, p < 0.05), the number of erythrocytes in urine samples (τ = 0.193, p < 0.05) and with the outcome of nephritis (τ = 0.287, p < 0.05); therefore, HMGB1 could be a potential tool for monitoring patients with IgAV who develop nephritis. Taken together, our results imply a possible interplay of Gd-IgA1, HMGB1, RAGE and PCDH1 in the development of IgAV. The identification of sensitive biomarkers in IgAV may provide disease prevention and future therapeutics.


Asunto(s)
Cadherinas , Proteína HMGB1 , Receptor para Productos Finales de Glicación Avanzada , Niño , Preescolar , Femenino , Humanos , Masculino , Biomarcadores/orina , Biomarcadores/sangre , Cadherinas/sangre , Cadherinas/genética , Cadherinas/orina , Estudios de Casos y Controles , Proteína HMGB1/sangre , Proteína HMGB1/orina , Vasculitis por IgA/sangre , Vasculitis por IgA/orina , Inmunoglobulina A/sangre , Estudios Prospectivos , Protocadherinas , Receptor para Productos Finales de Glicación Avanzada/sangre
16.
Open Biol ; 14(4): 230383, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38629124

RESUMEN

Non-clustered protocadherins (ncPcdhs) are adhesive molecules with spatio-temporally regulated overlapping expression in the developing nervous system. Although their unique role in neurogenesis has been widely studied, their combinatorial role in brain physiology and pathology is poorly understood. Using probabilistic cell typing by in situ sequencing, we demonstrate combinatorial inter- and intra-familial expression of ncPcdhs in the developing mouse cortex and hippocampus, at single-cell resolution. We discovered the combinatorial expression of Protocadherin-19 (Pcdh19), a protein involved in PCDH19-clustering epilepsy, with Pcdh1, Pcdh9 or Cadherin 13 (Cdh13) in excitatory neurons. Using aggregation assays, we demonstrate a code-specific adhesion function of PCDH19; mosaic PCDH19 absence in PCDH19+9 and PCDH19 + CDH13, but not in PCDH19+1 codes, alters cell-cell interaction. Interestingly, we found that PCDH19 as a dominant protein in two heterophilic adhesion codes could promote trans-interaction between them. In addition, we discovered increased CDH13-mediated cell adhesion in the presence of PCDH19, suggesting a potential role of PCDH19 as an adhesion mediator of CDH13. Finally, we demonstrated novel cis-interactions between PCDH19 and PCDH1, PCDH9 and CDH13. These observations suggest that there is a unique combinatorial code with a cell- and region-specific characteristic where a single molecule defines the heterophilic cell-cell adhesion properties of each code.


Asunto(s)
Encéfalo , Adhesión Celular , Protocadherinas , Animales , Ratones , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Epilepsia/metabolismo , Neuronas/metabolismo
17.
Int J Mol Sci ; 25(7)2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38612920

RESUMEN

X-linked epilepsies are a heterogeneous group of epileptic conditions, which often overlap with X-linked intellectual disability. To date, various X-linked genes responsible for epilepsy syndromes and/or developmental and epileptic encephalopathies have been recognized. The electro-clinical phenotype is well described for some genes in which epilepsy represents the core symptom, while less phenotypic details have been reported for other recently identified genes. In this review, we comprehensively describe the main features of both X-linked epileptic syndromes thoroughly characterized to date (PCDH19-related DEE, CDKL5-related DEE, MECP2-related disorders), forms of epilepsy related to X-linked neuronal migration disorders (e.g., ARX, DCX, FLNA) and DEEs associated with recently recognized genes (e.g., SLC9A6, SLC35A2, SYN1, ARHGEF9, ATP6AP2, IQSEC2, NEXMIF, PIGA, ALG13, FGF13, GRIA3, SMC1A). It is often difficult to suspect an X-linked mode of transmission in an epilepsy syndrome. Indeed, different models of X-linked inheritance and modifying factors, including epigenetic regulation and X-chromosome inactivation in females, may further complicate genotype-phenotype correlations. The purpose of this work is to provide an extensive and updated narrative review of X-linked epilepsies. This review could support clinicians in the genetic diagnosis and treatment of patients with epilepsy featuring X-linked inheritance.


Asunto(s)
Epilepsia , Espasmos Infantiles , Femenino , Humanos , Genes Ligados a X , Epigénesis Genética , Genes cdc , Epilepsia/genética , Receptor de Prorenina , Protocadherinas , Factores de Intercambio de Guanina Nucleótido , Factores de Intercambio de Guanina Nucleótido Rho , N-Acetilglucosaminiltransferasas
18.
Mol Psychiatry ; 29(7): 2199-2210, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38454084

RESUMEN

Clustering Epilepsy (CE) is a neurological disorder caused by pathogenic variants of the Protocadherin 19 (PCDH19) gene. PCDH19 encodes a protein involved in cell adhesion and Estrogen Receptor α mediated-gene regulation. To gain further insights into the molecular role of PCDH19 in the brain, we investigated the PCDH19 interactome in the developing mouse hippocampus and cortex. Combined with a meta-analysis of all reported PCDH19 interacting proteins, our results show that PCDH19 interacts with proteins involved in actin, microtubule, and gene regulation. We report CAPZA1, αN-catenin and, importantly, ß-catenin as novel PCDH19 interacting proteins. Furthermore, we show that PCDH19 is a regulator of ß-catenin transcriptional activity, and that this pathway is disrupted in CE individuals. Overall, our results support the involvement of PCDH19 in the cytoskeletal network and point to signalling pathways where PCDH19 plays critical roles.


Asunto(s)
Encéfalo , Cadherinas , Hipocampo , Proteómica , Protocadherinas , Vía de Señalización Wnt , beta Catenina , Animales , Cadherinas/metabolismo , Cadherinas/genética , Ratones , Vía de Señalización Wnt/fisiología , Vía de Señalización Wnt/genética , beta Catenina/metabolismo , beta Catenina/genética , Proteómica/métodos , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo , Humanos , Hipocampo/metabolismo , Epilepsia/metabolismo , Epilepsia/genética , Corteza Cerebral/metabolismo , Ratones Endogámicos C57BL
19.
Epilepsy Behav ; 154: 109730, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38521028

RESUMEN

PCDH19 clustering epilepsy (PCDH19-CE) is an X-linked epilepsy disorder associated with intellectual disability (ID) and behavioral disturbances, which is caused by PCDH19 gene variants. PCDH19 pathogenic variant leads to epilepsy in heterozygous females, not in hemizygous males and the inheritance pattern is unusual. The hypothesis of cellular interference was described as a key pathogenic mechanism. According to that, males do not develop the disease because of the uniform expression of PCDH19 (variant or wild type) unless they have a somatic variation. We conducted a literature review on PCDH19-CE pathophysiology and concluded that other significant mechanisms could contribute to pathogenesis including: asymmetric cell division and heterochrony, female-related allopregnanolone deficiency, altered steroid gene expression, decreased Gamma-aminobutyric acid receptor A (GABAA) function, and blood-brain barrier (BBB) dysfunction. Being aware of these mechanisms helps us when we should decide which therapeutic option is more suitable for which patient.


Asunto(s)
Cadherinas , Epilepsia , Protocadherinas , Humanos , Epilepsia/genética , Epilepsia/fisiopatología , Cadherinas/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Femenino , Masculino , Relevancia Clínica
20.
Hepatology ; 80(3): 566-577, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38373106

RESUMEN

BACKGROUND AND AIMS: Sustained inflammation and hepatocyte injury in chronic liver disease activate HSCs to transdifferentiate into fibrogenic, contractile myofibroblasts. We investigated the role of protocadherin 7 (PCDH7), a cadherin family member not previously characterized in the liver, whose expression is restricted to HSCs. APPROACH AND RESULTS: We created a PCDH7 fl/fl mouse line, which was crossed to lecithin retinol acyltransferase-Cre mice to generate HSC-specific PCDH7 knockout animals. HSC contraction in vivo was tested in response to the HSC-selective vasoconstrictor endothelin-1 using intravital multiphoton microscopy. To establish a PCDH7 null HSC line, cells were isolated from PCDH7 fl/fl mice and infected with adenovirus-expressing Cre. Hepatic expression of PCDH7 was strictly restricted to HSCs. Knockout of PCDH7 in vivo abrogated HSC-mediated sinusoidal contraction in response to endothelin-1. In cultured HSCs, loss of PCDH7 markedly attenuated contractility within collagen gels and led to altered gene expression in pathways governing adhesion and vasoregulation. Loss of contractility in PCDH7 knockout cells was impaired Rho-GTPase signaling, as demonstrated by altered gene expression, reduced assembly of F-actin fibers, and loss of focal adhesions. CONCLUSIONS: The stellate cell-specific cadherin, PCDH7, is a novel regulator of HSC contractility whose loss leads to cytoskeletal remodeling and sinusoidal relaxation.


Asunto(s)
Cadherinas , Células Estrelladas Hepáticas , Ratones Noqueados , Animales , Ratones , Cadherinas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/fisiología , Protocadherinas , Endotelina-1/metabolismo , Células Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA