Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47.427
Filtrar
1.
Mol Autism ; 15(1): 42, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39350244

RESUMEN

BACKGROUND: SETBP1 Haploinsufficiency Disorder (SETBP1-HD) is characterised by mild to moderate intellectual disability, speech and language impairment, mild motor developmental delay, behavioural issues, hypotonia, mild facial dysmorphisms, and vision impairment. Despite a clear link between SETBP1 mutations and neurodevelopmental disorders the precise role of SETBP1 in neural development remains elusive. We investigate the functional effects of three SETBP1 genetic variants including two pathogenic mutations p.Glu545Ter and SETBP1 p.Tyr1066Ter, resulting in removal of SKI and/or SET domains, and a point mutation p.Thr1387Met in the SET domain. METHODS: Genetic variants were introduced into induced pluripotent stem cells (iPSCs) and subsequently differentiated into neurons to model the disease. We measured changes in cellular differentiation, SETBP1 protein localisation, and gene expression changes. RESULTS: The data indicated a change in the WNT pathway, RNA polymerase II pathway and identified GATA2 as a central transcription factor in disease perturbation. In addition, the genetic variants altered the expression of gene sets related to neural forebrain development matching characteristics typical of the SETBP1-HD phenotype. LIMITATIONS: The study investigates changes in cellular function in differentiation of iPSC to neural progenitor cells as a human model of SETBP1 HD disorder. Future studies may provide additional information relevant to disease on further neural cell specification, to derive mature neurons, neural forebrain cells, or brain organoids. CONCLUSIONS: We developed a human SETBP1-HD model and identified perturbations to the WNT and POL2RA pathway, genes regulated by GATA2. Strikingly neural cells for both the SETBP1 truncation mutations and the single nucleotide variant displayed a SETBP1-HD-like phenotype.


Asunto(s)
Proteínas Portadoras , Diferenciación Celular , Haploinsuficiencia , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Humanos , Proteínas Portadoras/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mutación , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Neuronas/metabolismo , Células-Madre Neurales/metabolismo , Vía de Señalización Wnt/genética , Discapacidad Intelectual/genética , Fenotipo
3.
J Transl Med ; 22(1): 914, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39380010

RESUMEN

The heterogeneous nuclear ribonucleoprotein C (HNRNPC) plays a crucial role in tumorigenesis, yet its role in papillary thyroid carcinoma (PTC) remains elusive. Herein, we elucidated the function and molecular mechanism of HNRNPC in PTC tumorigenesis and progression. Our study unveiled a significant upregulation of HNRNPC in PTC, and knockdown of HNRNPC markedly inhibited the proliferation, invasion, and metastasis of BCPAP cells. Furthermore, HNRNPC modulated PKM alternative splicing in BCPAP cells primarily through m6A modification. Additionally, by upregulating PKM2 expression, HNRNPC promoted aerobic glycolysis in BCPAP cells, thereby facilitating malignant progression in PTC. In summary, our findings demonstrate that HNRNPC regulates PKM alternative splicing through m6A methylation modification and promotes the proliferation, invasion and metastasis of PTC through glucose metabolism pathways mediated by PKM2. These discoveries provide new biomarkers for screening and diagnosing PTC patients and offer novel therapeutic targets for personalized treatment strategies.


Asunto(s)
Empalme Alternativo , Proteínas Portadoras , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Glucólisis , Ribonucleoproteína Heterogénea-Nuclear Grupo C , Proteínas de la Membrana , Cáncer Papilar Tiroideo , Proteínas de Unión a Hormona Tiroide , Hormonas Tiroideas , Neoplasias de la Tiroides , Regulación hacia Arriba , Humanos , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/patología , Cáncer Papilar Tiroideo/metabolismo , Regulación hacia Arriba/genética , Línea Celular Tumoral , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Neoplasias de la Tiroides/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Empalme Alternativo/genética , Hormonas Tiroideas/metabolismo , Glucólisis/genética , Metilación , Ribonucleoproteína Heterogénea-Nuclear Grupo C/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo C/genética , Animales , Invasividad Neoplásica , Metástasis de la Neoplasia , Piruvato Quinasa/metabolismo , Piruvato Quinasa/genética , Ratones Desnudos , Adenosina/análogos & derivados , Adenosina/metabolismo
4.
J Gen Physiol ; 156(12)2024 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-39373654

RESUMEN

Myosin-binding protein H (MyBP-H) is a component of the vertebrate skeletal muscle sarcomere with sequence and domain homology to myosin-binding protein C (MyBP-C). Whereas skeletal muscle isoforms of MyBP-C (fMyBP-C, sMyBP-C) modulate muscle contractility via interactions with actin thin filaments and myosin motors within the muscle sarcomere "C-zone," MyBP-H has no known function. This is in part due to MyBP-H having limited expression in adult fast-twitch muscle and no known involvement in muscle disease. Quantitative proteomics reported here reveal that MyBP-H is highly expressed in prenatal rat fast-twitch muscles and larval zebrafish, suggesting a conserved role in muscle development and prompting studies to define its function. We take advantage of the genetic control of the zebrafish model and a combination of structural, functional, and biophysical techniques to interrogate the role of MyBP-H. Transgenic, FLAG-tagged MyBP-H or fMyBP-C both localize to the C-zones in larval myofibers, whereas genetic depletion of endogenous MyBP-H or fMyBP-C leads to increased accumulation of the other, suggesting competition for C-zone binding sites. Does MyBP-H modulate contractility in the C-zone? Globular domains critical to MyBP-C's modulatory functions are absent from MyBP-H, suggesting that MyBP-H may be functionally silent. However, our results suggest an active role. In vitro motility experiments indicate MyBP-H shares MyBP-C's capacity as a molecular "brake." These results provide new insights and raise questions about the role of the C-zone during muscle development.


Asunto(s)
Proteínas Portadoras , Fibras Musculares de Contracción Rápida , Pez Cebra , Animales , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Rápida/fisiología , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Sarcómeros/metabolismo , Contracción Muscular/fisiología , Desarrollo de Músculos/fisiología , Ratas , Citoesqueleto de Actina/metabolismo
5.
PLoS One ; 19(10): e0311761, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39388496

RESUMEN

Hypertrophic cardiomyopathy (HCM) is the most common heart disease in domestic cats, often leading to congestive heart failure and death, with current treatment strategies unable to reverse or prevent progression of the disease. The underlying pathological processes driving HCM remain unclear, which hinders novel drug discovery. The aim of this study was to generate a cellular model of the feline HCM-causing MYBPC3 mutation R820W. Using CRISPR/Cas9 gene editing we introduced the R820W mutation into a human induced pluripotent stem cell (iPSC) line. We differentiated both homozygous mutant clones and isogenic control clones to cardiomyocytes (iPSC-CMs). Protein quantification indicated that haploinsufficiency is not the disease mechanism of the mutation. Homozygous mutant iPSC-CMs had a larger cell area than isogenic controls, with the sarcomere structure and incorporation of cMyBP-C appearing similar between mutant and control iPSC-CMs. Contraction kinetic analysis indicated that homozygous iPSC-CMs have impaired relaxation and are hypocontractile compared to isogenic control iPSC-CMs. In summary, we demonstrate successful generation of an iPSC model of a feline MYBPC3 mutation, with the cellular model recapitulating aspects of HCM including cellular hypertrophy and impaired relaxation kinetics. We anticipate that further study of this model will lead to improved understanding of the disease-causing molecular mechanism, ultimately leading to novel drug discovery.


Asunto(s)
Sistemas CRISPR-Cas , Cardiomiopatía Hipertrófica , Proteínas Portadoras , Edición Génica , Células Madre Pluripotentes Inducidas , Mutación , Miocitos Cardíacos , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Gatos , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/veterinaria , Proteínas Portadoras/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Humanos , Diferenciación Celular , Enfermedades de los Gatos/genética , Enfermedades de los Gatos/patología
6.
BMC Nephrol ; 25(1): 340, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39390411

RESUMEN

BACKGROUND: Mutation of MMACHC gene causes cobalamin C disease (cblC), an inherited metabolic disorder, which presents as combined methylmalonic aciduria (MMA-uria) and hyperhomocysteinaemia in clinical. Renal complications may also be present in patients with this inborn deficiency. The most common histological change is thrombotic microangiopathy (TMA). However, to our acknowledge, renal tubular injury in the late-onset presentation of cblC is rarely been reported. This study provides a detailed description of the characteristics of kidney disease in cblC deficiency, aiming to improve the early recognition of this treatable disease for clinical nephrologists. CASE PRESENTATION: Here we described three teenage patients who presented with hematuria, proteinuria, and hypertension in clinical presentation. They were diagnosed with renal involvement due to cblC deficiency after laboratory tests revealing elevated serum and urine homocysteine, renal biopsy showing TMA and tubular injury, along with genetic testing showing heterogeneous compound mutations in MMACHC. Hydroxocobalamin, betaine, and L-carnitine were administered to these patients. All of them got improved, with decreased homocysteine, controlled blood pressure, and kidney outcomes recovered. CONCLUSIONS: The clinical diagnosis of cblC disease associated with kidney injury should be considered in patients with unclear TMA accompanied by a high concentration of serum homocysteine, even in teenagers or adults. Early diagnosis and timely intervention are vital to improving the prognosis of cobalamin C disease. CLINICAL TRIAL NUMBER: Not applicable.


Asunto(s)
Homocistinuria , Microangiopatías Trombóticas , Humanos , Masculino , Femenino , Homocistinuria/complicaciones , Homocistinuria/diagnóstico , Adolescente , Microangiopatías Trombóticas/etiología , Microangiopatías Trombóticas/complicaciones , Hidroxocobalamina/uso terapéutico , Proteínas Portadoras/genética , Deficiencia de Vitamina B 12/complicaciones , Deficiencia de Vitamina B 12/congénito , Túbulos Renales/patología , Oxidorreductasas , Betaína/uso terapéutico , Carnitina/uso terapéutico , Carnitina/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/complicaciones , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico
7.
Cell Death Dis ; 15(10): 728, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39368995

RESUMEN

Patients with lung adenocarcinoma (LUAD) generally have poor prognosis. Abnormal cellular energy metabolism is a hallmark of LUAD. Glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) is a member of the γ-glutamylcyclotransferase family and an unfolded protein response pathway regulatory gene. Its biological function and molecular regulatory mechanism, especially regarding energy metabolism underlying LUAD, remain unclear. By utilizing tissue microarray and data from The Cancer Genome Atlas and Gene Expression Omnibus, we found that CHAC1 expression was markedly higher in LUAD tissues than in non-tumor tissues, and was positively correlated with poor prognosis. Phenotypically, CHAC1 overexpression enhanced the proliferation, migration, invasion, tumor sphere formation, and glycolysis ability of LUAD cells, resulting in tumor growth both in vitro and in vivo. Mechanistically, through a shotgun mass spectrometry-based proteomic approach and high-throughput RNA sequencing, we found that CHAC1 acted as a bridge connecting UBA2 and PKM2, enhancing the SUMOylation of PKM2. The SUMOylated PKM2 then transferred from the cytoplasm to the nucleus, activating the expression of glycolysis-related genes and enhancing the Warburg effect. Lastly, E2F Transcription Factor 1 potently activated CHAC1 transcription by directly binding to the CHAC1 promoter in LUAD cells. The results of this study implied that CHAC1 regulates energy metabolism and promotes glycolysis in LUAD progression.


Asunto(s)
Adenocarcinoma del Pulmón , Proteínas Portadoras , Glucosa , Neoplasias Pulmonares , Proteínas de la Membrana , Proteínas de Unión a Hormona Tiroide , Hormonas Tiroideas , Humanos , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Hormonas Tiroideas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Glucosa/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Animales , Progresión de la Enfermedad , gamma-Glutamilciclotransferasa/metabolismo , gamma-Glutamilciclotransferasa/genética , Ratones , Línea Celular Tumoral , Proliferación Celular , Ratones Desnudos , Núcleo Celular/metabolismo , Masculino , Regulación Neoplásica de la Expresión Génica , Glucólisis , Femenino , Movimiento Celular , Ratones Endogámicos BALB C
8.
Int J Mol Med ; 54(5)2024 11.
Artículo en Inglés | MEDLINE | ID: mdl-39219281

RESUMEN

Psoriasis is a chronic inflammatory skin condition with numerous causes, including genetic, immunological and infectious factors. The course of psoriasis is long and recurrence is common; pathogenesis is not completely understood. However, there is an association between advancement of psoriasis and aberrant microRNA (miR or miRNA)­155 expression. Through bioinformatics, the present study aimed to analyze the differentially expressed genes and miRNAs in psoriasis and its biological mechanism and function psoriatic inflammation. First of all, differentially expressed genes (DEGs) and miRNAs (DEMs) in patients with psoriasis were identified using GEO2R interactive web application. A psoriasis inflammatory model was established using lipopolysaccharide (LPS)­treated HaCaT keratinocytes, which were transfected with miR­155 mimic or inhibitor. Cell Counting Kit­8 was used for the assessment of cell viability and proliferation, and changes in the cell cycle were examined using flow cytometry. ELISA and reverse transcription­quantitative PCR (RT­qPCR) were used to detect the expression levels of the inflammatory factors IL­1ß and IL­6. The dual­luciferase reporter assay was used to verify the targeting association between miR­155­5p and IFN regulatory factor 2 binding protein 2 (IRF2BP2). To verify the targeting association of miR­155 and the IRF2BP2/kruppel­like factor 2 (KLF2)/NF­κB signaling pathway, expression levels of IRF2BP2, KLF2 and p65 were identified by RT­qPCR and western blotting. IRF2BP2 levels were also confirmed by immunofluorescence, in conjunction with bioinformatics database analysis. Overexpression of miR­155 inhibited proliferation of HaCaT cells and increased the number of cells in S phase and decreasing number of cells in G1 and G2 phase. In the LPS­induced inflammatory state, miR­155 overexpression heightened the inflammatory response of HaCaT cells while inhibition of miR­155 lessened it. Suppression of inflammatory cytokine expression by miR­155­5p inhibitor was reversed by knockdown of IRF2BP2. miR­155 was shown to interact with IRF2BP2 to negatively regulate its expression, leading to decreased KLF2 expression and increased p65 expression and secretion of inflammatory factors, intensifying the inflammatory response of HaCaT cells. Therefore, miR­155 may contribute to development of psoriasis by inducing tissue and cell damage by increasing the inflammatory response of HaCaT cells via the IRF2BP2/KLF2/NF­κB pathway. In conclusion, the results of the present study offer novel perspectives on the role of miR­155 in the onset and progression of psoriasis.


Asunto(s)
Inflamación , Factores de Transcripción de Tipo Kruppel , MicroARNs , FN-kappa B , Psoriasis , Transducción de Señal , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Psoriasis/genética , Psoriasis/metabolismo , Psoriasis/patología , FN-kappa B/metabolismo , Transducción de Señal/genética , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Células HaCaT , Proliferación Celular/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Queratinocitos/metabolismo , Lipopolisacáridos/farmacología , Proteínas de Unión al ADN , Factores de Transcripción
9.
Bone Res ; 12(1): 50, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39231936

RESUMEN

Osteoarthritis (OA) is the most common form of arthritic disease, and phenotypic modification of chondrocytes is an important mechanism that contributes to the loss of cartilage homeostasis. This study identified that Fascin actin-bundling protein 1 (FSCN1) plays a pivotal role in regulating chondrocytes phenotype and maintaining cartilage homeostasis. Proteome-wide screening revealed markedly upregulated FSCN1 protein expression in human OA cartilage. FSCN1 accumulation was confirmed in the superficial layer of OA cartilage from humans and mice, primarily in dedifferentiated-like chondrocytes, associated with enhanced actin stress fiber formation and upregulated type I and III collagens. FSCN1-inducible knockout mice exhibited delayed cartilage degeneration following experimental OA surgery. Mechanistically, FSCN1 promoted actin polymerization and disrupted the inhibition of Decorin on TGF-ß1, leading to excessive TGF-ß1 production and ALK1/Smad1/5 signaling activation, thus, accelerated chondrocyte dedifferentiation. Intra-articular injection of FSCN1-overexpressing adeno-associated virus exacerbated OA progression in mice, which was mitigated by an ALK1 inhibitor. Moreover, FSCN1 inhibitor NP-G2-044 effectively reduced extracellular matrix degradation in OA mice, cultured human OA chondrocytes, and cartilage explants by suppressing ALK1/Smad1/5 signaling. These findings suggest that targeting FSCN1 represents a promising therapeutic approach for OA.


Asunto(s)
Proteínas Portadoras , Condrocitos , Proteínas de Microfilamentos , Osteoartritis , Animales , Humanos , Masculino , Ratones , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Cartílago Articular/metabolismo , Cartílago Articular/patología , Condrocitos/metabolismo , Condrocitos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Osteoartritis/patología , Osteoartritis/metabolismo , Osteoartritis/genética , Fenotipo , Receptores Odorantes , Transducción de Señal
10.
Biol Res ; 57(1): 66, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285301

RESUMEN

BACKGROUND: Spermatogonial stem cells (SSCs) are essential for the maintenance and initiation of male spermatogenesis. Despite the advances in understanding SSC biology in mouse models, the mechanisms underlying human SSC development remain elusive. RESULTS: Here, we analyzed the signaling pathways involved in SSC regulation by testicular somatic cells using single-cell sequencing data (GEO datasets: GSE149512 and GSE112013) and identified that Leydig cells communicate with SSCs through pleiotrophin (PTN) and its receptor syndecan-2 (SDC2). Immunofluorescence, STRING prediction, and protein immunoprecipitation assays confirmed the interaction between PTN and SDC2 in spermatogonia, but their co-localization was observed only in approximately 50% of the cells. The knockdown of SDC2 in human SSC lines impaired cell proliferation, DNA synthesis, and the expression of PLZF, a key marker for SSC self-renewal. Transcriptome analysis revealed that SDC2 knockdown downregulated the expression of GFRA1, a crucial factor for SSC proliferation and self-renewal, and inhibited the HIF-1 signaling pathway. Exogenous PTN rescued the proliferation and GFRA1 expression in SDC2 knockdown SSC lines. In addition, we found downregulation of PTN and SDC2 as well as altered localization in non-obstructive azoospermia (NOA) patients, suggesting that downregulation of PTN and SDC2 may be associated with impaired spermatogenesis. CONCLUSIONS: Our results uncover a novel mechanism of human SSC regulation by the testicular microenvironment and suggest a potential therapeutic target for male infertility.


Asunto(s)
Proteínas Portadoras , Proliferación Celular , Citocinas , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial , Células Intersticiales del Testículo , Sindecano-2 , Masculino , Humanos , Proliferación Celular/fisiología , Células Intersticiales del Testículo/metabolismo , Citocinas/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Sindecano-2/metabolismo , Sindecano-2/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Supervivencia Celular/fisiología , Espermatogonias/metabolismo , Transducción de Señal/fisiología , Células Madre Germinales Adultas/metabolismo , Células Madre Germinales Adultas/fisiología
11.
Nat Commun ; 15(1): 8381, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333095

RESUMEN

The presence of noncanonical open reading frames within lncRNAs (long non-coding RNAs) suggests their potential for translation, yielding various functional peptides or proteins. However, the existence and specific roles of these products in gastric cancer remain largely unclear. Here we identify the HOXA10-HOXA9-derived small protein (HDSP) in gastric cancer through comprehensive analysis and experimental validation, including mass spectrometry and western blotting. HDSP exhibits high expression and oncogenic roles in gastric cancer. Mechanistically, HDSP blocks TRIM25-mediated ubiquitination and degradation by interacting with MECOM, leading to MECOM accumulation and enhanced SPINK1 transcription-a gene promoting cancer via the EGFR signaling pathway. Furthermore, MECOM fosters HOXA10-HOXA9 transcription, establishing a feedback loop activating SPINK1-EGFR signaling. HDSP knockdown inhibits tumor growth in a PDX (patient-derived xenograft) model, and infusion of an artificially synthesized HDSP peptide as a neoantigen enhances immune cell-mediated anti-tumor efficacy against gastric cancer in vitro and in vivo. These findings propose HDSP as a potential therapeutic target or neoantigen candidate for gastric cancer treatment.


Asunto(s)
Receptores ErbB , Transducción de Señal , Neoplasias Gástricas , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Humanos , Receptores ErbB/metabolismo , Receptores ErbB/genética , Animales , Línea Celular Tumoral , Ratones , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Femenino , Progresión de la Enfermedad , Ratones Desnudos , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Masculino , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteínas Homeobox A10 , Proteínas Adaptadoras Transductoras de Señales , Complejos de Clasificación Endosomal Requeridos para el Transporte
12.
J Exp Clin Cancer Res ; 43(1): 265, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39342278

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) features KRAS mutations in approximately 90% of human cases and excessive stromal response, termed desmoplastic reaction. Oncogenic KRAS drives pancreatic carcinogenesis by acting on both epithelial cells and tumor microenvironment (TME). We have previously shown that Homeodomain-Interacting Protein Kinase 2 (HIPK2) cooperates with KRAS in sustaining ERK1/2 phosphorylation in human colorectal cancers. Here, we investigated whether HIPK2 contributes to oncogenic KRAS-driven tumorigenesis in vivo, in the onset of pancreatic cancer. METHODS: We employed an extensively characterized model of KRASG12D-dependent preinvasive PDAC, the Pdx1-Cre;LSL-KRasG12D/+ (KC) mice. In these mice, HIPK2 was inhibited by genetic knockout in the pancreatic epithelial cells (KCH-/-) or by pharmacologic inactivation with the small molecule 5-IodoTubercidin (5-ITu). The development of preneoplastic acinar-to-ductal metaplasia (ADM), intraepithelial neoplasia (PanIN), and their associated desmoplastic reaction were analyzed. RESULTS: In Hipk2-KO mice (KCH-/-), ERK phosphorylation was lowered, the appearance of ADM was slowed down, and both the number and pathologic grade of PanIN were reduced compared to Hipk2-WT KC mice. The pancreatic lesion phenotype in KCH-/- mice was characterized by abundant collagen fibers and reduced number of αSMA+ and pSTAT3+ desmoplastic cells. These features were reminiscent of the recently described human "deserted" sub-TME, poor in cells, rich in matrix, and associated with tumor differentiation. In contrast, the desmoplastic reaction of KC mice resembled the "reactive" sub-TME, rich in stromal cells and associated with tumor progression. These observations were confirmed by the pharmacologic inhibition of HIPK2 in KC mice. CONCLUSION: This study demonstrates that HIPK2 inhibition weakens oncogenic KRAS activity and pancreatic tumorigenesis providing a rationale for testing HIPK2 inhibitors to mitigate the incidence of PDAC development in high-risk individuals.


Asunto(s)
Neoplasias Pancreáticas , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas p21(ras) , Animales , Humanos , Ratones , Carcinogénesis , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/prevención & control , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Ratones Noqueados , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/prevención & control , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Microambiente Tumoral
13.
Development ; 151(17)2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39250531

RESUMEN

miR-31 is a highly conserved microRNA that plays crucial roles in cell proliferation, migration and differentiation. We discovered that miR-31 and some of its validated targets are enriched on the mitotic spindle of the dividing sea urchin embryo and mammalian cells. Using the sea urchin embryo, we found that miR-31 inhibition led to developmental delay correlated with increased cytoskeletal and chromosomal defects. We identified miR-31 to directly suppress several actin remodeling transcripts, including ß-actin, Gelsolin, Rab35 and Fascin. De novo translation of Fascin occurs at the mitotic spindle of sea urchin embryos and mammalian cells. Importantly, miR-31 inhibition leads to a significant a increase of newly translated Fascin at the spindle of dividing sea urchin embryos. Forced ectopic localization of Fascin transcripts to the cell membrane and translation led to significant developmental and chromosomal segregation defects, highlighting the importance of the regulation of local translation by miR-31 at the mitotic spindle to ensure proper cell division. Furthermore, miR-31-mediated post-transcriptional regulation at the mitotic spindle may be an evolutionarily conserved regulatory paradigm of mitosis.


Asunto(s)
MicroARNs , Biosíntesis de Proteínas , Huso Acromático , Animales , MicroARNs/metabolismo , MicroARNs/genética , Huso Acromático/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/genética , Mitosis/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Desarrollo Embrionario/genética , Embrión no Mamífero/metabolismo , Segregación Cromosómica/genética , Actinas/metabolismo , Actinas/genética , Erizos de Mar/embriología , Erizos de Mar/genética , Erizos de Mar/metabolismo
14.
Signal Transduct Target Ther ; 9(1): 258, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39341825

RESUMEN

Epigenetic readers frequently affect gene regulation, correlate with disease prognosis, and hold significant potential as therapeutic targets for cancer. Zinc finger MYND-type containing 11 (ZMYND11) is notably recognized for reading the epigenetic marker H3.3K36me3; however, its broader functions and mechanisms of action in cancer remain underexplored. Here, we report that ZMYND11 downregulation is prevalent across various cancers and profoundly correlates with poorer outcomes in prostate cancer patients. Depletion of ZMYND11 promotes tumor cell growth, migration, and invasion in vitro, as well as tumor formation and metastasis in vivo. Mechanistically, we discover that ZMYND11 exhibits tumor suppressive roles by recognizing arginine-194-methylated HNRNPA1 dependent on its MYND domain, thereby retaining HNRNPA1 in the nucleus and preventing the formation of stress granules in the cytoplasm. Furthermore, ZMYND11 counteracts the HNRNPA1-driven increase in the PKM2/PKM1 ratio, thus mitigating the aggressive tumor phenotype promoted by PKM2. Remarkably, ZMYND11 recognition of HNRNPA1 can be disrupted by pharmaceutical inhibition of the arginine methyltransferase PRMT5. Tumors with low ZMYND11 expression show sensitivity to PRMT5 inhibitors. Taken together, our findings uncover a previously unexplored noncanonical role of ZMYND11 as a nonhistone methylation reader and underscore the critical importance of arginine methylation in the ZMYND11-HNRNPA1 interaction for restraining tumor progression, thereby proposing novel therapeutic targets and potential biomarkers for cancer treatment.


Asunto(s)
Epigénesis Genética , Ribonucleoproteína Nuclear Heterogénea A1 , Humanos , Ribonucleoproteína Nuclear Heterogénea A1/genética , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Epigénesis Genética/genética , Masculino , Gránulos de Estrés/genética , Gránulos de Estrés/metabolismo , Línea Celular Tumoral , Ratones , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Carcinogénesis/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN , Proteínas de Ciclo Celular , Proteínas Co-Represoras
15.
Nat Commun ; 15(1): 7438, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39256350

RESUMEN

Gene therapy holds promise for treatment of inherited retinal dystrophies, a group of rare genetic disorders characterized by severe loss of vision. Here, we report up to 3-year pre-specified interim safety and efficacy results of an open-label first-in-human dose-escalation phase 1/2 gene therapy clinical trial in 12 patients with retinal dystrophy caused by biallelic mutations in the retinaldehyde-binding protein 1 (RLBP1) gene of the visual cycle. The primary endpoints were systemic and ocular safety and recovery of dark adaptation. Secondary endpoints included microperimetry, visual field sensitivity, dominant eye test and patient-reported outcomes. Subretinal delivery of an adeno-associated viral vector (AAV8-RLBP1) was well tolerated with dose-dependent intraocular inflammation which responded to corticosteroid treatment, and focal atrophy of the retinal pigment epithelium as the dose limiting toxicity. Dark adaptation kinetics, the primary efficacy endpoint, improved significantly in all dose-cohorts. Treatment with AAV8-RLBP1 resulted in the resolution of disease-related retinal deposits, suggestive of successful restoration of the visual cycle. In conclusion, to date, AAV8-RLBP1 has shown preliminary safety and efficacy in patients with RLBP1-associated retinal dystrophy. Trial number: NCT03374657.


Asunto(s)
Dependovirus , Terapia Genética , Distrofias Retinianas , Humanos , Terapia Genética/métodos , Distrofias Retinianas/genética , Distrofias Retinianas/terapia , Masculino , Adulto , Femenino , Dependovirus/genética , Proteínas Portadoras/genética , Persona de Mediana Edad , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Adulto Joven , Resultado del Tratamiento , Mutación , Adolescente
16.
Commun Biol ; 7(1): 1157, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39284869

RESUMEN

Neuroligin-2 (Nlgn2) is a key synaptic adhesion protein at virtually all GABAergic synapses, which recruits GABAARs by promoting assembly of the postsynaptic gephyrin scaffold. Intriguingly, loss of Nlgn2 differentially affects subsets of GABAergic synapses, indicating that synapse-specific interactors and redundancies define its function, but the nature of these interactions remain poorly understood. Here we investigated how Nlgn2 function in hippocampal area CA1 is modulated by two proposed interaction partners, MDGA1 and MDGA2. We show that loss of MDGA1 expression, but not heterozygous deletion of MDGA2, ameliorates the abnormal cytosolic gephyrin aggregation, the reduction in inhibitory synaptic transmission and the exacerbated anxiety-related behaviour characterizing Nlgn2 knockout (KO) mice. Additionally, combined Nlgn2 and MDGA1 deletion causes an exacerbated layer-specific loss of gephyrin puncta. Given that both Nlgn2 and the MDGA1 have been correlated with many psychiatric disorders, our data support the notion that cytosolic gephyrin aggregation may represent an interesting target for novel therapeutic strategies.


Asunto(s)
Proteínas Portadoras , Moléculas de Adhesión Celular Neuronal , Proteínas de la Membrana , Ratones Noqueados , Receptores de GABA-A , Sinapsis , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Sinapsis/metabolismo , Receptores de GABA-A/metabolismo , Receptores de GABA-A/genética , Citosol/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Transmisión Sináptica , Ratones Endogámicos C57BL , Región CA1 Hipocampal/metabolismo
17.
PLoS One ; 19(9): e0310219, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39259742

RESUMEN

Nucleostemin (NS) plays a role in liver regeneration, and aging reduces its expression in the baseline and regenerating livers following 70% partial hepatectomy (PHx). Here we interrogate the mechanism controlling NS expression during liver regeneration and aging. The NS promoter was analyzed by TRANSFAC. Functional studies were performed using cell-based luciferase assay, endogenous NS expression in Hep3B cells, mouse livers with a gain-of-function mutation of C/EBPα (S193D), and mouse livers with C/EBPα knockdown. We found a CAAT box with four C/EBPα binding sites (-1216 to -735) and a GC box with consensus binding sites for c-Myc, E2F1, and p300-associated protein complex (-633 to -1). Age-related changes in NS expression correlated positively with the expression of c-Myc, E2F1, and p300, and negatively with that of C/EBPα and C/EBPß. PHx upregulated NS expression at 1d, coinciding with an increase in E2F1 and a decrease in C/EBPα. C/EBPα bound to the consensus sequences found in the NS promoter in vitro and in vivo, inhibited its transactivational activity in a binding site-dependent manner, and decreased the expression of endogenous NS in Hep3B cells. In vivo activation of C/EBPα by the S193D mutation resulted in a 4th-day post-PHx reduction of NS, a feature shared by 16-m/o livers. Finally, C/EBPα knockdown increased its expression in aged (24-m/o) livers under both baseline and regeneration conditions. This study reports the C/EBPα suppression of NS expression in aged livers, providing a new perspective on the mechanistic orchestration of tissue homeostasis in aging.


Asunto(s)
Envejecimiento , Proteínas de Unión al GTP , Regeneración Hepática , Proteínas Nucleares , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc , Animales , Regeneración Hepática/genética , Regeneración Hepática/fisiología , Ratones , Envejecimiento/metabolismo , Envejecimiento/fisiología , Envejecimiento/genética , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/genética , Hepatectomía , Sitios de Unión , Hígado/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Regulación de la Expresión Génica , Transcripción Genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Masculino , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Ratones Endogámicos C57BL , Línea Celular Tumoral , Proteínas de Unión al ARN
18.
BMC Med ; 22(1): 357, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39227839

RESUMEN

BACKGROUND: Our previous genome­wide association studies (GWAS) have suggested rs912304 in 14q12 as a suggestive risk variant for type 1 diabetes (T1D). However, the association between this risk region and T1D subgroups and related clinical risk features, the underlying causal functional variant(s), putative candidate gene(s), and related mechanisms are yet to be elucidated. METHODS: We assessed the association between variant rs912304 and T1D, as well as islet autoimmunity and islet function, stratified by the diagnosed age of 12. We used epigenome bioinformatics analyses, dual luciferase reporter assays, and expression quantitative trait loci (eQTL) analyses to prioritize the most likely functional variant and potential causal gene. We also performed functional experiments to evaluate the role of the causal gene on islet function and its related mechanisms. RESULTS: We identified rs912304 as a risk variant for T1D subgroups with diagnosed age ≥ 12 but not < 12. This variant is associated with residual islet function but not islet-specific autoantibody positivity in T1D individuals. Bioinformatics analysis indicated that rs912304 is a functional variant exhibiting spatial overlaps with enhancer active histone marks (H3K27ac and H3K4me1) and open chromatin status (ATAC-seq) in the human pancreas and islet tissues. Luciferase reporter gene assays and eQTL analyses demonstrated that the biallelic sites of rs912304 had differential allele-specific enhancer activity in beta cell lines and regulated STXBP6 expression, which was defined as the most putative causal gene based on Open Targets Genetics, GTEx v8 and Tiger database. Moreover, Stxbp6 was upregulated by T1D-related proinflammatory cytokines but not high glucose/fat. Notably, Stxbp6 over-expressed INS-1E cells exhibited decreasing insulin secretion and increasing cell apoptosis through Glut1 and Gadd45ß, respectively. CONCLUSIONS: This study expanded the genomic landscape regarding late-onset T1D risk and supported islet function mechanistically connected to T1D pathogenesis.


Asunto(s)
Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Adolescente , Animales , Niño , Femenino , Humanos , Masculino , Edad de Inicio , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/genética , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Islotes Pancreáticos/metabolismo , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo , Proteínas Portadoras/genética
19.
Pestic Biochem Physiol ; 204: 106058, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39277374

RESUMEN

The juvenile hormone binding protein (JHBP) and takeout (TO) genes, mediated by the juvenile hormone (JH), play a crucial role in regulating the reproductive physiology of insects. Our previous study revealed that spinosad-resistant Frankliniella occidentalis (NIL-R) exhibited reduced fecundity and significant changes in JHBP/TO family gene expression. We hypothesized that these genes were involved in regulating the fitness costs associated with resistance. In this study, 45 JHBP/TO genes were identified in F. occidentalis, among which FoTO2 and FoTO10 were duplicates. Additionally, eight genes exhibited significant down-regulation in the NIL-R population. Two genes (FoTO6 and FoTO24) that exhibited the most significant differential expression between the spinosad-susceptible (Ivf03) and NIL-R populations were selected to investigate their roles in resistance fitness using RNA interference (RNAi). Following interference with FoTO6, FoTO24, and their combination, the expression levels of vitellogenin (Vg) were downregulated by 3%-30%, 13%-28%, and 14%-32% from the 2nd day to the 5th day, respectively; Krüppel-homolog 1 (Kr-h1) expression was down-regulated by 3%-65%, 11%-34%, and 11%-39% from the 2nd day to the 5th day, respectively; ovariole length was shortened by approximately 18%, 21%, and 24%, respectively; and the average number of eggs decreased from 407 to 260, 148, and 106, respectively. Additionally, a JH supplementation experiment on the NIL-R population revealed that the expression levels of both FoTO6, FoTO24, Vg and Kr-h1 were significantly upregulated compared with those observed in the Ivf03 population, resulting in increased fecundity. These results suggest that FoTO6 and FoTO24 are involved in JH-mediated regulation of the reproductive fitness cost of resistance to spinosad. Further, FoTO6 and FoTO24 can be considered potential target genes for applying RNAi technology in the scientific management of F. occidentalis.


Asunto(s)
Proteínas de Insectos , Resistencia a los Insecticidas , Thysanoptera , Animales , Resistencia a los Insecticidas/genética , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Thysanoptera/genética , Thysanoptera/fisiología , Thysanoptera/efectos de los fármacos , Insecticidas/farmacología , Femenino , Reproducción/genética , Macrólidos/farmacología , Vitelogeninas/genética , Vitelogeninas/metabolismo , Combinación de Medicamentos , Hormonas Juveniles/metabolismo , Hormonas Juveniles/farmacología , Interferencia de ARN , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Aptitud Genética
20.
Gut Microbes ; 16(1): 2395907, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39262376

RESUMEN

Gut microbiome dysbiosis has been widely implicated in cognitive impairment, but the identity of the specific bacterial taxa and mechanisms are not fully elucidated. Brain glucose hypometabolism coincides with the cognitive decline. This study explored the link among cognition, gut microbiota and glucose uptake based on the fecal microbiota transplantation from mild cognitive impairment individuals (MCI-FMT) and investigated whether similar mechanisms were involved in 27-hydroxycholesterol (27-OHC)-induced cognitive decline. Our results showed that the MCI-FMT mice exhibited learning and memory decline and morphological lesions in the brain and colon tissues. There were reduced 18F-fluorodeoxyglucose uptake, downregulated expression of glucose transporters (GLUT1,3,4) and upregulated negative regulator of glucose uptake (TXNIP) in the brain. MCI-FMT altered the bacterial composition and diversity of the recipient mice, and the microbial signatures highlighted by the increased abundance of Bacteroides recapitulated the negative effects of MCI bacterial colonization. However, inhibiting Bacteroidetes or TXNIP increased the expression of GLUT1 and GLUT4, significantly improving brain glucose uptake and cognitive performance in 27-OHC-treated mice. Our study verified that cognitive decline and abnormal cerebral glucose uptake were associated with gut microbiota dysbiosis; we also revealed the involvement of Bacteroidetes and molecular mechanisms of TXNIP-related glucose uptake in cognitive deficits caused by 27-OHC.


Asunto(s)
Bacteroidetes , Encéfalo , Cognición , Disfunción Cognitiva , Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Glucosa , Transducción de Señal , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/microbiología , Ratones , Glucosa/metabolismo , Encéfalo/metabolismo , Bacteroidetes/metabolismo , Disbiosis/microbiología , Disbiosis/metabolismo , Masculino , Humanos , Ratones Endogámicos C57BL , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Tiorredoxinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA