Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 218
Filtrar
1.
Nat Commun ; 15(1): 6790, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39117654

RESUMEN

Immunochemotherapy has been the mainstay of treatment for newly diagnosed diffuse large B-cell lymphoma (ndDLBCL) yet is inadequate for many patients. In this work, we perform unsupervised clustering on transcriptomic features from a large cohort of ndDLBCL patients and identify seven clusters, one called A7 with poor prognosis, and develop a classifier to identify these clusters in independent ndDLBCL cohorts. This high-risk cluster is enriched for activated B-cell cell-of-origin, low immune infiltration, high MYC expression, and copy number aberrations. We compare and contrast our methodology with recent DLBCL classifiers to contextualize our clusters and show improved prognostic utility. Finally, using pre-clinical models, we demonstrate a mechanistic rationale for IKZF1/3 degraders such as lenalidomide to overcome the low immune infiltration phenotype of A7 by inducing T-cell trafficking into tumors and upregulating MHC I and II on tumor cells, and demonstrate that TCF4 is an important regulator of MYC-related biology in A7.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Factor de Transcripción Ikaros , Lenalidomida , Linfoma de Células B Grandes Difuso , Proteínas Proto-Oncogénicas c-myc , Factor de Transcripción 4 , Transcriptoma , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Humanos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Lenalidomida/uso terapéutico , Lenalidomida/farmacología , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Linfocitos B/metabolismo , Linfocitos B/inmunología , Pronóstico , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica/métodos , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Variaciones en el Número de Copia de ADN
2.
Skelet Muscle ; 14(1): 15, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39026379

RESUMEN

BACKGROUND: TCF4 acts as a transcription factor that binds to the immunoglobulin enhancer Mu-E5/KE5 motif. Dominant variants in TCF4 are associated with the manifestation of Pitt-Hopkins syndrome, a rare disease characterized by severe mental retardation, certain features of facial dysmorphism and, in many cases, with abnormalities in respiratory rhythm (episodes of paroxysmal tachypnea and hyperventilation, followed by apnea and cyanosis). Frequently, patients also develop epilepsy, microcephaly, and postnatal short stature. Although TCF4 is expressed in skeletal muscle and TCF4 seems to play a role in myogenesis as demonstrated in mice, potential myopathological findings taking place upon the presence of dominant TCF4 variants are thus far not described in human skeletal muscle. METHOD: To address the pathological effect of a novel deletion affecting exons 15 and 16 of TCF4 on skeletal muscle, histological and immunofluorescence studies were carried out on a quadriceps biopsy in addition to targeted transcript studies and global proteomic profiling. RESULTS: We report on muscle biopsy findings from a Pitt-Hopkins patient with a novel heterozygous deletion spanning exon 15 and 16 presenting with neuromuscular symptoms. Microscopic characterization of the muscle biopsy revealed moderate fiber type I predominance, imbalance in the proportion of fibroblasts co-expressing Vimentin and CD90, and indicate activation of the complement cascade in TCF4-mutant muscle. Protein dysregulations were unraveled by proteomic profiling. Transcript studies confirmed a mitochondrial vulnerability in muscle and confirmed reduced TCF4 expression. CONCLUSION: Our combined findings, for the first time, unveil myopathological changes as phenotypical association of Pitt-Hopkins syndrome and thus expand the current clinical knowledge of the disease as well as support data obtained on skeletal muscle of a mouse model.


Asunto(s)
Hiperventilación , Discapacidad Intelectual , Factor de Transcripción 4 , Hiperventilación/genética , Hiperventilación/metabolismo , Hiperventilación/fisiopatología , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Facies , Niño , Exones , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología
3.
Cell Transplant ; 33: 9636897241259552, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38847385

RESUMEN

Thin endometrium (TE) is a significant factor contributing to fertility challenges, and addressing this condition remains a central challenge in reproductive medicine. Menstrual blood-derived mesenchymal stem cells (MenSCs) play a crucial role in tissue repair and regeneration, including that of TE. The Wnt signaling pathway, which is highly conserved and prevalent in eukaryotes, is essential for cell proliferation, tissue development, and reproductive functions. MALAT1 is implicated in various transcriptional and molecular functions, including cell proliferation and metastasis. However, the combined effects of the Wnt signaling pathway and the long non-coding RNA (lncRNA) MALAT1 on the regulation of MenSCs' regenerative capabilities in tissue engineering have not yet been explored. To elucidate the regulatory mechanism of MALAT1 in TE, we analyzed its expression levels in normal endometrium and TE tissues, finding that low expression of MALAT1 was associated with poor clinical prognosis. In addition, we conducted both in vitro and in vivo functional assays to examine the role of the MALAT1/miR-7-5p/TCF4 axis in cell proliferation and migration. Techniques such as dual-luciferase reporter assay, fluorescent in situ hybridization, and immunoblot experiments were utilized to clarify the molecular mechanism. To corroborate these findings, we established a TE model and conducted pregnancy experiments, demonstrating a strong association between MALAT1 expression and endometrial fertility. In conclusion, our comprehensive study provides strong evidence supporting that lncRNA MALAT1 modulates TCF4 expression in the Wnt signaling pathway through interaction with miR-7-5p, thus enhancing MenSCs-mediated improvement of TE and improving fertility.


Asunto(s)
Endometrio , Células Madre Mesenquimatosas , MicroARNs , ARN Largo no Codificante , Vía de Señalización Wnt , Femenino , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Endometrio/metabolismo , Endometrio/citología , MicroARNs/metabolismo , MicroARNs/genética , Animales , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Proliferación Celular/genética , Adulto , Ratones , Fertilidad/genética
4.
Invest Ophthalmol Vis Sci ; 65(6): 27, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38884552

RESUMEN

Purpose: This study evaluated the dysregulation of TCF4 isoforms and differential exon usage (DEU) in corneal endothelial cells (CECs) of Fuchs endothelial corneal dystrophy (FECD) with or without trinucleotide repeat (TNR) expansion in the intron region of the TCF4 gene. Methods: Three RNA-Seq datasets of CECs (our own and two other previously published datasets) derived from non-FECD control and FECD subjects were analyzed to identify TCF4 isoforms and DEU events dysregulated in FECD by comparing control subjects to those with FECD with TNR expansion and FECD without TNR expansion. Results: Our RNA-Seq data demonstrated upregulation of three TCF4 isoforms and downregulation of two isoforms in FECD without TNR expansion compared to the controls. In FECD with TNR expansion, one isoform was upregulated and one isoform was downregulated compared to the control. Additional analysis using two other datasets identified that the TCF4-277 isoform was upregulated in common in all three datasets in FECD with TNR expansion, whereas no isoform was dysregulated in FECD without TNR expansion. DEU analysis showed that one exon (E174) upstream of the TNR, which only encompassed TCF4-277, was upregulated in common in all three datasets, whereas eight exons downstream of the TNR were downregulated in common in all three datasets in FECD with TNR expansion. Conclusions: This study identified TCF4-277 as a dysregulated isoform in FECD with TNR expansion, suggesting a potential contribution of TCF4-277 to FECD pathophysiology.


Asunto(s)
Endotelio Corneal , Distrofia Endotelial de Fuchs , Factor de Transcripción 4 , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Endotelio Corneal/metabolismo , Endotelio Corneal/patología , Exones/genética , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/metabolismo , Regulación de la Expresión Génica , Isoformas de Proteínas/genética , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Expansión de Repetición de Trinucleótido/genética
5.
BMC Oral Health ; 24(1): 597, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778377

RESUMEN

BACKGROUND: Pitt-Hopkins syndrome (PTHS) is a rare neurodevelopmental disorder with physical, cognitive, and behavioral characteristics that is caused by heterozygous mutations in the TCF4 gene. Patients with PTHS might present a unique challenge for oral healthcare professionals because of the associated comorbidities. CASE REPORT: Here we describe a new case of PTHS in a 13-year-old girl with particular emphasis on oro-dental findings and oral healthcare management. Observed oro-dental findings in our case included shallow palate, absence of lingual frenum, gingival enlargement, thick lips and relative microdontia. The patient was unable to tolerate dental care under local anesthesia. Therefore, comprehensive dental treatment was performed under general anesthesia after a careful pre-anesthetic cardio-respiratory, neurological, and hematological evaluation. The patient was closely monitored intra-operatively for breathing rhythm, O2 saturation, and signs of respiratory distress. The patient was observed for 24 h post-op for respiratory distress and was discharged then uneventfully. CONCLUSION: Dental treatment under general anesthesia in these patients might be complicated by the abnormal breathing rhythm, and close monitoring and follow up for signs of respiratory distress after general anesthesia is necessary. Recognition of oral and dental findings might help to expand the phenotype and better characterize rare syndromes.


Asunto(s)
Discapacidad Intelectual , Fenotipo , Humanos , Femenino , Adolescente , Discapacidad Intelectual/genética , Facies , Factor de Transcripción 4/genética , Anestesia General , Anomalías de la Boca/genética , Hiperventilación , Atención Dental para Enfermos Crónicos , Frenillo Lingual/anomalías , Frenillo Lingual/cirugía
6.
Sci Rep ; 14(1): 10276, 2024 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704483

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is a complex corneal disease characterized by the progressive decline and morphological changes of corneal endothelial cells (CECs) that leads to corneal edema and vision loss. The most common mutation in FECD is an intronic CTG repeat expansion in transcription factor 4 (TCF4) that leads to its altered expression. Corneal endothelial wound healing occurs primarily through cell enlargement and migration, and FECD CECs have been shown to display increased migration speeds. In this study, we aim to determine whether TCF4 can promote cellular migration in FECD CECs. We generated stable CEC lines derived from FECD patients that overexpressed different TCF4 isoforms and investigated epithelial-to-mesenchymal (EMT) expression, morphological analysis and cellular migration speeds. We found that full length TCF4-B isoform overexpression promotes cellular migration in FECD CECs in an EMT-independent manner. RNA-sequencing identified several pathways including the negative regulation of microtubules, with TUBB4A (tubulin beta 4A class IVa) as the top upregulated gene. TUBB4A expression was increased in FECD ex vivo specimens, and there was altered expression of cytoskeleton proteins, tubulin and actin, compared to normal healthy donor ex vivo specimens. Additionally, there was increased acetylation and detyrosination of microtubules in FECD supporting that microtubule stability is altered in FECD and could promote cellular migration. Future studies could be aimed at investigating if targeting the cytoskeleton and microtubules would have therapeutic potential for FECD by promoting cellular migration and regeneration.


Asunto(s)
Movimiento Celular , Endotelio Corneal , Distrofia Endotelial de Fuchs , Microtúbulos , Factor de Transcripción 4 , Humanos , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/metabolismo , Distrofia Endotelial de Fuchs/patología , Movimiento Celular/genética , Microtúbulos/metabolismo , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Endotelio Corneal/metabolismo , Endotelio Corneal/patología , Masculino , Femenino , Transición Epitelial-Mesenquimal/genética , Anciano , Células Endoteliales/metabolismo , Células Endoteliales/patología , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/genética , Persona de Mediana Edad , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética
7.
Commun Biol ; 7(1): 545, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714724

RESUMEN

CircRNAs are covalently closed, single-stranded RNA that form continuous loops and play a crucial role in the initiation and progression of tumors. Cancer stem cells (CSCs) are indispensable for cancer development; however, the regulation of cancer stem cell-like properties in gastric cancer (GC) and its specific mechanism remain poorly understood. We elucidate the specific role of Circ-0075305 in GC stem cell properties. Circ-0075305 associated with chemotherapy resistance was identified by sequencing GC cells. Subsequent confirmation in both GC tissues and cell lines revealed that patients with high expression of Circ-0075305 had significantly better overall survival (OS) rates than those with low expression, particularly when treated with postoperative adjuvant chemotherapy for GC. In vitro and in vivo experiments confirmed that overexpression of Circ-0075305 can effectively reduce stem cell-like properties and enhance the sensitivity of GC cells to Oxaliplatin compared with the control group. Circ-0075305 promotes RPRD1A expression by acting as a sponge for corresponding miRNAs. The addition of LF3 (a ß-catenin/TCF4 interaction antagonist) confirmed that RPRD1A inhibited the formation of the TCF4-ß-catenin transcription complex through competitive to ß-catenin and suppressed the transcriptional activity of stem cell markers such as SOX9 via the Wnt/ß-catenin signaling pathway. This leads to the downregulation of stem cell-like property-related markers in GC. This study revealed the underlying mechanisms that regulate Circ-0075305 in GCSCs and suggests that its role in reducing ß-catenin signaling may serve as a potential therapeutic candidate.


Asunto(s)
Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas , ARN Circular , Factor de Transcripción SOX9 , Neoplasias Gástricas , Factor de Transcripción 4 , beta Catenina , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Humanos , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , beta Catenina/metabolismo , beta Catenina/genética , ARN Circular/genética , ARN Circular/metabolismo , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Animales , Ratones , Línea Celular Tumoral , Ratones Desnudos , Masculino , Femenino , Resistencia a Antineoplásicos/genética , Ratones Endogámicos BALB C , Persona de Mediana Edad
8.
PLoS Genet ; 20(5): e1011230, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38713708

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is an age-related cause of vision loss, and the most common repeat expansion-mediated disease in humans characterised to date. Up to 80% of European FECD cases have been attributed to expansion of a non-coding CTG repeat element (termed CTG18.1) located within the ubiquitously expressed transcription factor encoding gene, TCF4. The non-coding nature of the repeat and the transcriptomic complexity of TCF4 have made it extremely challenging to experimentally decipher the molecular mechanisms underlying this disease. Here we comprehensively describe CTG18.1 expansion-driven molecular components of disease within primary patient-derived corneal endothelial cells (CECs), generated from a large cohort of individuals with CTG18.1-expanded (Exp+) and CTG 18.1-independent (Exp-) FECD. We employ long-read, short-read, and spatial transcriptomic techniques to interrogate expansion-specific transcriptomic biomarkers. Interrogation of long-read sequencing and alternative splicing analysis of short-read transcriptomic data together reveals the global extent of altered splicing occurring within Exp+ FECD, and unique transcripts associated with CTG18.1-expansions. Similarly, differential gene expression analysis highlights the total transcriptomic consequences of Exp+ FECD within CECs. Furthermore, differential exon usage, pathway enrichment and spatial transcriptomics reveal TCF4 isoform ratio skewing solely in Exp+ FECD with potential downstream functional consequences. Lastly, exome data from 134 Exp- FECD cases identified rare (minor allele frequency <0.005) and potentially deleterious (CADD>15) TCF4 variants in 7/134 FECD Exp- cases, suggesting that TCF4 variants independent of CTG18.1 may increase FECD risk. In summary, our study supports the hypothesis that at least two distinct pathogenic mechanisms, RNA toxicity and TCF4 isoform-specific dysregulation, both underpin the pathophysiology of FECD. We anticipate these data will inform and guide the development of translational interventions for this common triplet-repeat mediated disease.


Asunto(s)
Distrofia Endotelial de Fuchs , Factor de Transcripción 4 , Expansión de Repetición de Trinucleótido , Humanos , Masculino , Empalme Alternativo/genética , Células Endoteliales/metabolismo , Endotelio Corneal/metabolismo , Endotelio Corneal/patología , Distrofia Endotelial de Fuchs/genética , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Transcriptoma/genética , Expansión de Repetición de Trinucleótido/genética , Femenino
9.
HGG Adv ; 5(3): 100289, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-38571311

RESUMEN

Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by pathogenic variants in TCF4, leading to intellectual disability, specific morphological features, and autonomic nervous system dysfunction. Epigenetic dysregulation has been implicated in PTHS, prompting the investigation of a DNA methylation (DNAm) "episignature" specific to PTHS for diagnostic purposes and variant reclassification and functional insights into the molecular pathophysiology of this disorder. A cohort of 67 individuals with genetically confirmed PTHS and three individuals with intellectual disability and a variant of uncertain significance (VUS) in TCF4 were studied. The DNAm episignature was developed with an Infinium Methylation EPIC BeadChip array analysis using peripheral blood cells. Support vector machine (SVM) modeling and clustering methods were employed to generate a DNAm classifier for PTHS. Validation was extended to an additional cohort of 11 individuals with PTHS. The episignature was assessed in relation to other neurodevelopmental disorders and its specificity was examined. A specific DNAm episignature for PTHS was established. The classifier exhibited high sensitivity for TCF4 haploinsufficiency and missense variants in the basic-helix-loop-helix domain. Notably, seven individuals with TCF4 variants exhibited negative episignatures, suggesting complexities related to mosaicism, genetic factors, and environmental influences. The episignature displayed degrees of overlap with other related disorders and biological pathways. This study defines a DNAm episignature for TCF4-related PTHS, enabling improved diagnostic accuracy and VUS reclassification. The finding that some cases scored negatively underscores the potential for multiple or nested episignatures and emphasizes the need for continued investigation to enhance specificity and coverage across PTHS-related variants.


Asunto(s)
Metilación de ADN , Hiperventilación , Discapacidad Intelectual , Factor de Transcripción 4 , Humanos , Factor de Transcripción 4/genética , Hiperventilación/genética , Hiperventilación/diagnóstico , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico , Femenino , Masculino , Niño , Facies , Adolescente , Epigenómica/métodos , Epigénesis Genética , Hipercinesia/genética , Preescolar , Adulto , Adulto Joven
10.
Breast Cancer Res ; 26(1): 70, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38654332

RESUMEN

BACKGROUND: Basal-like breast cancer (BLBC) is the most aggressive subtype of breast cancer due to its aggressive characteristics and lack of effective therapeutics. However, the mechanism underlying its aggressiveness remains largely unclear. S-adenosylmethionine decarboxylase proenzyme (AMD1) overexpression occurs specifically in BLBC. Here, we explored the potential molecular mechanisms and functions of AMD1 promoting the aggressiveness of BLBC. METHODS: The potential effects of AMD1 on breast cancer cells were tested by western blotting, colony formation, cell proliferation assay, migration and invasion assay. The spermidine level was determined by high performance liquid chromatography. The methylation status of CpG sites within the AMD1 promoter was evaluated by bisulfite sequencing PCR. We elucidated the relationship between AMD1 and Sox10 by ChIP assays and quantitative real-time PCR. The effect of AMD1 expression on breast cancer cells was evaluated by in vitro and in vivo tumorigenesis model. RESULTS: In this study, we showed that AMD1 expression was remarkably elevated in BLBC. AMD1 copy number amplification, hypomethylation of AMD1 promoter and transcription activity of Sox10 contributed to the overexpression of AMD1 in BLBC. AMD1 overexpression enhanced spermidine production, which enhanced eIF5A hypusination, activating translation of TCF4 with multiple conserved Pro-Pro motifs. Our studies showed that AMD1-mediated metabolic system of polyamine in BLBC cells promoted tumor cell proliferation and tumor growth. Clinically, elevated expression of AMD1 was correlated with high grade, metastasis and poor survival, indicating poor prognosis of breast cancer patients. CONCLUSION: Our work reveals the critical association of AMD1-mediated spermidine-eIF5A hypusination-TCF4 axis with BLBC aggressiveness, indicating potential prognostic indicators and therapeutic targets for BLBC.


Asunto(s)
Neoplasias de la Mama , Proliferación Celular , Factor 5A Eucariótico de Iniciación de Traducción , Regulación Neoplásica de la Expresión Génica , Lisina/análogos & derivados , Factores de Iniciación de Péptidos , Proteínas de Unión al ARN , Espermidina , Factor de Transcripción 4 , Humanos , Femenino , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Factores de Iniciación de Péptidos/metabolismo , Factores de Iniciación de Péptidos/genética , Ratones , Animales , Espermidina/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Línea Celular Tumoral , Regiones Promotoras Genéticas , Adenosilmetionina Descarboxilasa/metabolismo , Adenosilmetionina Descarboxilasa/genética , Movimiento Celular/genética , Metilación de ADN , Pronóstico , Factores de Transcripción SOXE/metabolismo , Factores de Transcripción SOXE/genética
11.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167178, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636614

RESUMEN

Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by haploinsufficiency of transcription factor 4 (TCF4). In this work, we focused on the cerebral cortex and investigated in detail the progenitor cell dynamics and the outcome of neurogenesis in a PTHS mouse model. Labeling and quantification of progenitors and newly generated neurons at various time points during embryonic development revealed alterations affecting the dynamic of cortical progenitors since the earliest stages of cortex formation in PTHS mice. Consequently, establishment of neuronal populations and layering of the cortex were found to be altered in heterozygotes subjects at birth. Interestingly, defective layering process of pyramidal neurons was partially rescued by reintroducing TCF4 expression using focal in utero electroporation in the cerebral cortex. Coincidentally with a defective dorsal neurogenesis, we found that ventral generation of interneurons was also defective in this model, which may lead to an excitation/inhibition imbalance in PTHS. Overall, sex-dependent differences were detected with more marked effects evidenced in males compared with females. All of this contributes to expand our understanding of PTHS, paralleling the advances of research in autism spectrum disorder and further validating the PTHS mouse model as an important tool to advance preclinical studies.


Asunto(s)
Corteza Cerebral , Modelos Animales de Enfermedad , Hiperventilación , Discapacidad Intelectual , Neurogénesis , Factor de Transcripción 4 , Animales , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Femenino , Masculino , Ratones , Hiperventilación/metabolismo , Hiperventilación/genética , Hiperventilación/patología , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Discapacidad Intelectual/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Facies , Caracteres Sexuales , Interneuronas/metabolismo , Interneuronas/patología , Células Piramidales/metabolismo , Células Piramidales/patología , Haploinsuficiencia
12.
Free Radic Biol Med ; 220: 125-138, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38657754

RESUMEN

Fusobacterium (F.) nucleatum is a carcinogenesis microbiota in colorectal cancer (CRC). Growing evidence shows that F. nucleatum contributes to chemoresistance. Ferroptosis is reported to restore the susceptibility of resistant cells to chemotherapy. However, the role of gut microbiota affecting ferroptosis in chemoresistance remains unclear. Here, we examined the CRC tissues of patients using 16S rRNA sequencing to investigate the possible connection between gut microbiota dysbiosis and the relapse of CRC. We found that a high abundance of F. nucleatum in CRC tissue is associated with relapse. We further demonstrated that F. nucleatum induced oxaliplatin resistance in vitro and in vivo. The transcriptome of an F. nucleatum-infected cell revealed ferroptosis was associated with F. nucleatum infection. We perform malondialdehyde, ferrous iron, and glutathione assays to verify the effect of F. nucleatum on ferroptosis under oxaliplatin treatment in vivo and in vitro. Mechanistically, F. nucleatum promoted oxaliplatin resistance by overexpressing GPX4 and then inhibiting ferroptosis. E-cadherin/ß-catenin/TCF4 pathway conducted the GPX4 overexpression effect of F. nucleatum. The chromatin immuno-precipitation quantitative PCR (CHIP-qPCR) and dual-luciferase reporter assay showed that F. nucleatum promoted TCF4 binding with GPX4. We also determined the E-cadherin/ß-catenin/TCF4/GPX4 axis related to tumor tissue F. nucleatum status and CRC relapse clinically. Here, we revealed the contribution of F. nucleatum to oxaliplatin resistance by inhibiting ferroptosis in CRC. Targeting F. nucleatum and ferroptosis will provide valuable insight into chemoresistance management and may improve outcomes for patients with CRC.


Asunto(s)
Cadherinas , Neoplasias Colorrectales , Resistencia a Antineoplásicos , Ferroptosis , Fusobacterium nucleatum , Microbioma Gastrointestinal , Oxaliplatino , Fosfolípido Hidroperóxido Glutatión Peroxidasa , beta Catenina , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/genética , Cadherinas/metabolismo , Cadherinas/genética , Oxaliplatino/farmacología , beta Catenina/metabolismo , beta Catenina/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Animales , Fusobacterium nucleatum/patogenicidad , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Masculino , Antígenos CD/metabolismo , Antígenos CD/genética , Femenino , Línea Celular Tumoral , Infecciones por Fusobacterium/microbiología , Infecciones por Fusobacterium/tratamiento farmacológico , Infecciones por Fusobacterium/metabolismo , Infecciones por Fusobacterium/genética , Infecciones por Fusobacterium/patología , Disbiosis/microbiología , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Ratones Desnudos
13.
Commun Biol ; 7(1): 418, 2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38582945

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is a leading indication for corneal transplantation, but its molecular etiology remains poorly understood. We performed genome-wide association studies (GWAS) of FECD in the Million Veteran Program followed by multi-ancestry meta-analysis with the previous largest FECD GWAS, for a total of 3970 cases and 333,794 controls. We confirm the previous four loci, and identify eight novel loci: SSBP3, THSD7A, LAMB1, PIDD1, RORA, HS3ST3B1, LAMA5, and COL18A1. We further confirm the TCF4 locus in GWAS for admixed African and Hispanic/Latino ancestries and show an enrichment of European-ancestry haplotypes at TCF4 in FECD cases. Among the novel associations are low frequency missense variants in laminin genes LAMA5 and LAMB1 which, together with previously reported LAMC1, form laminin-511 (LM511). AlphaFold 2 protein modeling, validated through homology, suggests that mutations at LAMA5 and LAMB1 may destabilize LM511 by altering inter-domain interactions or extracellular matrix binding. Finally, phenome-wide association scans and colocalization analyses suggest that the TCF4 CTG18.1 trinucleotide repeat expansion leads to dysregulation of ion transport in the corneal endothelium and has pleiotropic effects on renal function.


Asunto(s)
Distrofia Endotelial de Fuchs , Humanos , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/metabolismo , Estudio de Asociación del Genoma Completo , Factor de Transcripción 4/genética , Colágeno , Laminina/genética
14.
JCI Insight ; 9(8)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470486

RESUMEN

IL-17C is an epithelial cell-derived proinflammatory cytokine whose transcriptional regulation remains unclear. Analysis of the IL17C promoter region identified TCF4 as putative regulator, and siRNA knockdown of TCF4 in human keratinocytes (KCs) increased IL17C. IL-17C stimulation of KCs (along with IL-17A and TNF-α stimulation) decreased TCF4 and increased NFKBIZ and ZC3H12A expression in an IL-17RA/RE-dependent manner, thus creating a feedback loop. ZC3H12A (MCPIP1/Regnase-1), a transcriptional immune-response regulator, also increased following TCF4 siRNA knockdown, and siRNA knockdown of ZC3H12A decreased NFKBIZ, IL1B, IL36G, CCL20, and CXCL1, revealing a proinflammatory role for ZC3H12A. Examination of lesional skin from the KC-Tie2 inflammatory dermatitis mouse model identified decreases in TCF4 protein concomitant with increases in IL-17C and Zc3h12a that reversed following the genetic elimination of Il17c, Il17ra, and Il17re and improvement in the skin phenotype. Conversely, interference with Tcf4 in KC-Tie2 mouse skin increased Il17c and exacerbated the inflammatory skin phenotype. Together, these findings identify a role for TCF4 in the negative regulation of IL-17C, which, alone and with TNF-α and IL-17A, feed back to decrease TCF4 in an IL-17RA/RE-dependent manner. This loop is further amplified by IL-17C-TCF4 autocrine regulation of ZC3H12A and IL-17C regulation of NFKBIZ to promote self-sustaining skin inflammation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Interleucina-17 , Queratinocitos , Receptores de Interleucina-17 , Ribonucleasas , Transducción de Señal , Factor de Transcripción 4 , Animales , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Humanos , Interleucina-17/metabolismo , Interleucina-17/genética , Ratones , Queratinocitos/metabolismo , Ribonucleasas/metabolismo , Ribonucleasas/genética , Receptores de Interleucina-17/metabolismo , Receptores de Interleucina-17/genética , Inflamación/metabolismo , Inflamación/genética , Modelos Animales de Enfermedad , Epidermis/metabolismo , Dermatitis/metabolismo , Dermatitis/genética , Dermatitis/inmunología , Dermatitis/patología , Retroalimentación Fisiológica , Regulación de la Expresión Génica
15.
Adv Biol (Weinh) ; 8(5): e2300117, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38379270

RESUMEN

The incidence of Hepatocellular carcinoma (HCC) and HCC-related deaths have remarkably increased over the recent decades. It has been reported that ß-catenin activation can be frequently observed in HCC cases. This study identified the integrin-linked kinase-associated phosphatase (ILKAP) as a novel ß-catenin-interacting protein. ILKAP is localized both in the nucleus and cytoplasm and regulates the WNT pathway in different ways. First, it is demonstrated that ILKAP activates the WNT pathway in HCC cells by increasing the protein level of ß-catenin and other proteins associated with the WNT signaling, such as c-Myc and CyclinD1. Next, it is shown that ILKAP promotes the metastasis of HCC both in vitro and in vivo in a zebrafish xenograft model. It is also found that ILKAP dephosphorylates the GSK3ß and CK1, contributing to the reduced ubiquitination of ß-catenin. Furthermore, it is identified that ILKAP functions by mediating binding between TCF4 and ß-catenin to enhance expression of WNT target genes. Taken together, the study demonstrates a critical function of ILKAP in metastasis of HCC, since ILKAP is crucial for the activation of the WNT pathway via stabilization of ß-catenin and increased binding between TCF4 and ß-catenin.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Fosfoproteínas Fosfatasas , Vía de Señalización Wnt , beta Catenina , Animales , Humanos , beta Catenina/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Metástasis de la Neoplasia , Factor de Transcripción 4/metabolismo , Factor de Transcripción 4/genética , Vía de Señalización Wnt/fisiología , Pez Cebra , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo
16.
Poult Sci ; 103(3): 103377, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38301496

RESUMEN

Ovarian follicle development depends on the proliferation and differentiation of granulosa cells and is a complex biological process. The Wnt/ß-catenin signaling pathway can regulate ovarian follicle development, and ß-catenin, encoded by catenin beta 1 (CTNNB1), is the core component of this pathway. Although several studies of the mechanisms by which the Wnt/ß-catenin pathway regulates cell proliferation in humans and mammals have reported, it remains unclear how ß-catenin functions in poultry. To investigate the function of ß-catenin in laying hens' follicle development, we evaluated the effect of CTNNB1 on cell cycle, proliferation, and apoptosis in ovarian granulosa cells (GCs) isolated from laying hens. We demonstrated that CTNNB1 significantly affected the expression of cyclin D1 (CCND1) and v-myc avian myelocytomatosis viral oncogene homolog (c-Myc) (P < 0.01 and P < 0.05), key genes related to cell cycle and proliferation, to promote cell cycle progression from G1 to S phase, and thus accelerate granulosa cell proliferation. CTNNB1 did not however affect apoptosis or the expression of related genes baculoviral IAP repeat containing 5 (BIRC5) and BCL2 apoptosis regulator (Bcl-2). Overexpression of transcription factor 7-like 2 (TCF4) resulted in increased expression of CCND1, accelerated cell cycle progression, and granulosa cell proliferation. Direct physical interaction between ß-catenin and TCF4 was demonstrated by immunofluorescence and coimmunoprecipitation. The proliferation of granulosa cells was inhibited by silencing CCND1; overexpression of TCF4 in CCND1-silenced cells restored their proliferation rate to normal levels. These results indicate that the interaction of TCF4 and ß-catenin promotes CCND1 expression which in turn accelerates the cell cycle process of laying hen hierarchical follicular granulosa cells.


Asunto(s)
Pollos , beta Catenina , Humanos , Animales , Femenino , beta Catenina/genética , Pollos/genética , Apoptosis , Proliferación Celular , Células de la Granulosa , Mamíferos , Factor de Transcripción 4/genética
17.
Orphanet J Rare Dis ; 19(1): 51, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38331897

RESUMEN

BACKGROUND: Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder that remains underdiagnosed and its clinical presentations and mutation profiles in a diverse population are yet to be evaluated. This retrospective study aims to investigate the clinical and genetic characteristics of Chinese patients with PTHS. METHODS: The clinical, biochemical, genetic, therapeutic, and follow-up data of 47 pediatric patients diagnosed with PTHS between 2018 and 2021 were retrospectively analyzed. RESULTS: The Chinese PTHS patients presented with specific facial features and exhibited global developmental delay of wide severity range. The locus heterogeneity of the TCF4 gene in the patients was highlighted, emphasizing the significance of genetic studies for accurate diagnosis, albeit no significant correlations between genotype and phenotype were observed in this cohort. The study also reports the outcomes of patients who underwent therapeutic interventions, such as ketogenic diets and biomedical interventions. CONCLUSIONS: The findings of this retrospective analysis expand the phenotypic and molecular spectra of PTHS patients. The study underscores the need for a long-term prospective follow-up study to assess potential therapeutic interventions.


Asunto(s)
Discapacidad Intelectual , Niño , Humanos , Estudios Retrospectivos , Estudios de Seguimiento , Estudios Prospectivos , Factor de Transcripción 4/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico , Hiperventilación/genética , Hiperventilación/diagnóstico , Facies , China
18.
Pediatr Neurosurg ; 59(2-3): 109-114, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38246161

RESUMEN

INTRODUCTION: Pitt-Hopkins syndrome (PTHS) is a rare genetic syndrome associated with neurodevelopmental disorders and craniofacial dysmorphisms caused by variations in the TCF4 transition factor. The aim of this article was to report the case of two twin infants diagnosed with PTHS, confirmed by the identification of a heterozygous pathogenic variant in the TCF4 gene through DNA extracted from a buccal swab. CASE PRESENTATION: Both infants presented with craniofacial asymmetry with a metopic crest and cranial deformity. During the diagnostic investigation, computed tomography with three-dimensional reconstruction of the skull showed premature fusion of the left coronal and metopic sutures in both twins. They underwent craniofacial reconstruction at the 9th month of age using a combination of techniques. The postoperative outcomes were satisfactory in both cases. CONCLUSION: To the best of our knowledge, this is the first case report to describe the occurrence of complex craniosynostosis (CCS) in children with PTHS. Further studies are needed to determine whether the co-occurrence of PTHS and CCS described here indicates an association or is explained by chance.


Asunto(s)
Craneosinostosis , Hiperventilación , Discapacidad Intelectual , Humanos , Craneosinostosis/cirugía , Craneosinostosis/diagnóstico por imagen , Craneosinostosis/genética , Craneosinostosis/complicaciones , Discapacidad Intelectual/genética , Hiperventilación/genética , Lactante , Femenino , Masculino , Factor de Transcripción 4/genética , Facies , Enfermedades en Gemelos/cirugía , Enfermedades en Gemelos/diagnóstico por imagen , Tomografía Computarizada por Rayos X
19.
Cell Tissue Bank ; 25(2): 613-618, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38206443

RESUMEN

Fuchs endothelial corneal dystrophy (FECD) is caused by a corneal endothelial cell loss, leading to corneal edema and visual impairment. The most significant genetic risk factor for FECD is an expansion of the CTG18.1 locus in transcription factor 4 (TCF4). The current treatment for severe FECD is corneal transplantation, with Descemet stripping automated keratoplasty (DSAEK) as a common surgical method. Although successful in most cases, the risk for transplant failure due to diverse causes must be considered. In this study, we investigated if presence of TCF4 CTG18.1 expansion with more than 31 (n ≥ 31) repeats in donated corneal grafts could be a reason for corneal transplant failure after DSAEK. For this, nine consecutively failed DSAEK corneal grafts were genotyped for CTG18.1 repeat length. One-sided Mann-Whitney U test was performed to evaluate if failed DSAEK corneal grafts had longer CTG18.1 repeats than healthy controls from the same population. All failed corneal grafts had CTG18.1 n ≤ 27 with a median of 18 (IQR 8.0) repeats for the longest allele. There was no statistical difference in CTG18.1 repeat lengths between failed corneal grafts and the geographically matched healthy control group. In conclusion, none of the nine failed corneal grafts in our material had CTG18.1 repeat lengths ≥ 31, a cut-off known to have a biological relevance in FECD. Thus, our results suggest that the assessment of donors and inspection of the corneal tissue before the decision for procurement is sufficient, in terms of recognizing FECD in the donor.


Asunto(s)
Distrofia Endotelial de Fuchs , Factor de Transcripción 4 , Humanos , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Masculino , Femenino , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/cirugía , Anciano , Persona de Mediana Edad , Trasplante de Córnea , Anciano de 80 o más Años , Queratoplastia Endotelial de la Lámina Limitante Posterior , Expansión de Repetición de Trinucleótido/genética , Rechazo de Injerto/genética , Alelos , Córnea/cirugía , Genotipo
20.
Mol Biotechnol ; 66(5): 1174-1187, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38206529

RESUMEN

Circular RNA (circRNA) influences on the pathological process of osteoarthritis (OA) and may be a potential marker for disease diagnosis. The study was to scrutinize the association of circ_0045474 with OA. Clinical samples of OA patients were collected, and 12 circRNAs derived from KPNA2 gene were examined. CHON-001 cells were stimulated with IL-1ß to construct an OA chondrocyte model. miR-485-3p, transcription factor 4 (TCF4) and circ_0045474, type II procollagen (COL2A1), and human collagenase-3 (MMP13) were tested. Furthermore, cell activities were analyzed. The relationship between miR-485-3p, TCF4, and circ_0045474 was determined. The role of circ_0045474 in vivo was further confirmed by constructing an OA mouse model by anterior cruciate ligament transection. circ_0045474 expression was elevated in OA patients. Suppressing circ_0045474 restrained IL-1ß-stimulated extracellular matrix degradation, inflammatory cytokine secretion, and chondrocyte apoptosis. Circ_0045474 competitively combined with miR-485-3p, while TCF4 was the target of miR-485-3p. Circ_0045474 modulated IL-1ß-stimulated extracellular matrix degradation, inflammatory cytokine secretion, and chondrocyte apoptosis via miR-485-3p/TCF4 axis. Suppressing circ 0045474 was effective to alleviate OA in mice. Silenced circ_0045474 suppresses OA progression in vitro and vivo via miR-485-3p/TCF4 axis. In short, circ_0045474 can be considered a novel therapeutic target for OA.


Asunto(s)
Condrocitos , MicroARNs , Osteoartritis , ARN Circular , Factor de Transcripción 4 , Osteoartritis/genética , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/tratamiento farmacológico , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , Animales , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Ratones , Masculino , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Modelos Animales de Enfermedad , Persona de Mediana Edad , Femenino , Apoptosis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Línea Celular , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA