Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
N Engl J Med ; 388(26): 2422-2433, 2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37140166

RESUMEN

BACKGROUND: Telomere shortening is a well-characterized cellular aging mechanism, and short telomere syndromes cause age-related disease. However, whether long telomere length is advantageous is poorly understood. METHODS: We examined the clinical and molecular features of aging and cancer in persons carrying heterozygous loss-of-function mutations in the telomere-related gene POT1 and noncarrier relatives. RESULTS: A total of 17 POT1 mutation carriers and 21 noncarrier relatives were initially included in the study, and a validation cohort of 6 additional mutation carriers was subsequently recruited. A majority of the POT1 mutation carriers with telomere length evaluated (9 of 13) had long telomeres (>99th percentile). POT1 mutation carriers had a range of benign and malignant neoplasms involving epithelial, mesenchymal, and neuronal tissues in addition to B- and T-cell lymphoma and myeloid cancers. Five of 18 POT1 mutation carriers (28%) had T-cell clonality, and 8 of 12 (67%) had clonal hematopoiesis of indeterminate potential. A predisposition to clonal hematopoiesis had an autosomal dominant pattern of inheritance, as well as penetrance that increased with age; somatic DNMT3A and JAK2 hotspot mutations were common. These and other somatic driver mutations probably arose in the first decades of life, and their lineages secondarily accumulated a higher mutation burden characterized by a clocklike signature. Successive generations showed genetic anticipation (i.e., an increasingly early onset of disease). In contrast to noncarrier relatives, who had the typical telomere shortening with age, POT1 mutation carriers maintained telomere length over the course of 2 years. CONCLUSIONS: POT1 mutations associated with long telomere length conferred a predisposition to a familial clonal hematopoiesis syndrome that was associated with a range of benign and malignant solid neoplasms. The risk of these phenotypes was mediated by extended cellular longevity and by the capacity to maintain telomeres over time. (Funded by the National Institutes of Health and others.).


Asunto(s)
Envejecimiento , Hematopoyesis Clonal , Neoplasias , Telómero , Humanos , Envejecimiento/genética , Hematopoyesis Clonal/genética , Heterocigoto , Mutación con Pérdida de Función/genética , Mutación , Neoplasias/genética , Complejo Shelterina/genética , Síndrome , Telómero/genética , Telómero/fisiología , Homeostasis del Telómero/genética , Proteínas de Unión a Telómeros/genética
2.
BMC Cancer ; 22(1): 180, 2022 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177031

RESUMEN

BACKGROUND: Breast cancer is the most common malignancy in women, and is both pathologically and genetically heterogeneous, making early detection and treatment difficult. A subset of breast cancers express normal levels of REST (repressor element 1 silencing transcription factor) mRNA but lack functional REST protein. Loss of REST function is seen in ~ 20% of breast cancers and is associated with a more aggressive phenotype and poor prognosis. Despite the frequent loss of REST, little is known about the role of REST in the molecular pathogenesis of breast cancer. METHODS: TCGA data was analyzed for the expression of REST target genes in breast cancer patient samples. We then utilized gene knockdown in MCF-7 cells in the presence or absence of steroid hormones estrogen and/ progesterone followed by RNA sequencing, as well as chromatin immunoprecipitation and PCR in an attempt to understand the tumor suppressor role of REST in breast cancer. RESULTS: We show that REST directly regulates CEMIP (cell migration-inducing and hyaluronan-binding protein, KIAA1199) and MMP24 (matrix metallopeptidase 24), genes known to have roles in invasion and metastasis. REST knockdown in breast cancer cells leads to significant upregulation of CEMIP and MMP24. In addition, we found REST binds to RE-1 sites (repressor element-1) within the genes and influences their transcription. Furthermore, we found that the estrogen receptor (ESR1) signaling pathway is activated in the absence of REST, regardless of hormone treatment. CONCLUSIONS: We demonstrate a critical role for the loss of REST in aggressive breast cancer pathogenesis and provide evidence for REST as an important diagnostic marker for personalized treatment plans.


Asunto(s)
Neoplasias de la Mama/genética , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Hialuronoglucosaminidasa/genética , Metaloproteinasas de la Matriz Asociadas a la Membrana/genética , Biomarcadores de Tumor/genética , Femenino , Humanos , Mutación con Pérdida de Función/genética , Células MCF-7 , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Procesos Neoplásicos , Fenotipo , Pronóstico , ARN Mensajero/genética , Proteínas Represoras , Transducción de Señal/genética , Regulación hacia Arriba/genética
3.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35216443

RESUMEN

Nuclear-encoded Atp23 was previously shown to have dual functions, including processing the yeast Atp6 precursor and assisting the assembly of yeast mitochondrial ATP synthase. However, it remains unknown whether there are genes functionally complementary to ATP23 to rescue atp23 null mutant. In the present paper, we screen and characterize three revertants of atp23 null mutant and reveal a T1121G point mutation in the mitochondrial gene COX1 coding sequence, which leads to Val374Gly mutation in Cox1, the suppressor in the revertants. This was verified further by the partial restoration of mitochondrial ATP synthase assembly in atp23 null mutant transformed with exogenous hybrid COX1 T1121G mutant plasmid. The predicted tertiary structure of the Cox1 p.Val374Gly mutation showed no obvious difference from wild-type Cox1. By further chase labeling with isotope [35S]-methionine, we found that the stability of Atp6 of ATP synthase increased in the revertants compared with the atp23 null mutant. Taking all the data together, we revealed that the T1121G point mutation of mitochondrial gene COX1 could partially restore the unassembly of mitochondrial ATP synthase in atp23 null mutant by increasing the stability of Atp6. Therefore, this study uncovers a gene that is partially functionally complementary to ATP23 to rescue ATP23 deficiency, broadening our understanding of the relationship between yeast the cytochrome c oxidase complex and mitochondrial ATP synthase complex.


Asunto(s)
Complejo IV de Transporte de Electrones/genética , Genes Mitocondriales/genética , Metaloproteasas/genética , Mitocondrias/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Mutación Puntual/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Adenosina Trifosfato/genética , Secuencia de Aminoácidos , ADN Mitocondrial/genética , Mutación con Pérdida de Función/genética
4.
Nat Struct Mol Biol ; 28(12): 982-988, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34887559

RESUMEN

Neurofibromin (NF1) mutations cause neurofibromatosis type 1 and drive numerous cancers, including breast and brain tumors. NF1 inhibits cellular proliferation through its guanosine triphosphatase-activating protein (GAP) activity against rat sarcoma (RAS). In the present study, cryo-electron microscope studies reveal that the human ~640-kDa NF1 homodimer features a gigantic 30 × 10 nm array of α-helices that form a core lemniscate-shaped scaffold. Three-dimensional variability analysis captured the catalytic GAP-related domain and lipid-binding SEC-PH domains positioned against the core scaffold in a closed, autoinhibited conformation. We postulate that interaction with the plasma membrane may release the closed conformation to promote RAS inactivation. Our structural data further allow us to map the location of disease-associated NF1 variants and provide a long-sought-after structural explanation for the extreme susceptibility of the molecule to loss-of-function mutations. Collectively these findings present potential new routes for therapeutic modulation of the RAS pathway.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Neurofibromatosis 1/genética , Neurofibromina 1/metabolismo , Proteínas ras/metabolismo , Membrana Celular/metabolismo , Proliferación Celular/genética , Microscopía por Crioelectrón , Humanos , Mutación con Pérdida de Función/genética , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Conformación Proteica
5.
PLoS One ; 16(12): e0261724, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34932608

RESUMEN

Papillon-Lefèvre Syndrome (PLS) is an autosomal recessive monogenic disease caused by loss-of-function mutations in the CTSC gene, thus preventing the synthesis of the protease Cathepsin C (CTSC) in a proteolytically active form. CTSC is responsible for the activation of the pro-forms of the neutrophil serine proteases (NSPs; Elastase, Proteinase 3 and Cathepsin G), suggesting its involvement in a variety of neutrophil functions. In PLS neutrophils, the lack of CTSC protease activity leads to inactivity of the NSPs. Clinically, PLS is characterized by an early, typically pre-pubertal, onset of severe periodontal pathology and palmoplantar hyperkeratosis. However, PLS is not considered an immune deficiency as patients do not typically suffer from recurrent and severe (bacterial and fungal) infections. In this study we investigated an unusual CTSC mutation in two siblings with PLS, a 503A>G substitution in exon 4 of the CTSC gene, expected to result in an amino acid replacement from tyrosine to cysteine at position 168 of the CTSC protein. Both patients bearing this mutation presented with pronounced periodontal pathology. The characteristics and functions of neutrophils from patients homozygous for the 503A>G CTSC mutation were compared to another previously described PLS mutation (755A>T), and a small cohort of healthy volunteers. Neutrophil lysates from patients with the 503A>G substitution lacked CTSC protein and did not display any CTSC or NSP activity, yet neutrophil counts, morphology, priming, chemotaxis, radical production, and regulation of apoptosis were without any overt signs of alteration. However, NET formation upon PMA-stimulation was found to be severely depressed, but not abolished, in PLS neutrophils.


Asunto(s)
Catepsina C/genética , Trampas Extracelulares/metabolismo , Neutrófilos/patología , Enfermedad de Papillon-Lefevre/genética , Serina Proteasas/metabolismo , Adulto , Apoptosis , Catepsina C/metabolismo , Citometría de Flujo , Humanos , Mutación con Pérdida de Función/genética , Persona de Mediana Edad , Enfermedad de Papillon-Lefevre/enzimología , Enfermedad de Papillon-Lefevre/patología , Especies Reactivas de Oxígeno/metabolismo , Análisis de Secuencia de ADN
6.
Cells ; 10(11)2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34831043

RESUMEN

Studies have shown that the BH3-only domain Bad regulates brain development via the control of programmed cell death (PCD), but very few studies have addressed its effect on the molecular signaling of brain development in the system. In this work, we examined the novel role of zebrafish Bad in initial programmed cell death for brain morphogenesis through the priming of p53-mediated stress signaling. In a biological function study on the knockdown of Bad by morpholino oligonucleotides, at 24 h post-fertilization (hpf) Bad defects induced abnormal hindbrain development, as determined in a tissue section by means of HE staining which traced the damaged hindbrain. Then, genome-wide approaches for monitoring either the upregulation of apoptotic-related genes (11.8%) or the downregulation of brain development-related genes (29%) at the 24 hpf stage were implemented. The p53/caspase-8-mediated apoptotic death pathway was strongly involved, with the pathway being strongly reversed in a p53 mutant (p53M214K) line during Bad knockdown. Furthermore, we propose the involvement of a p53-mediated stress signal which is correlated with regulating Bad loss-mediated brain defects. We found that some major genes in brain development, such as crybb1, pva1b5, irx4a, pax7a, and fabp7a, were dramatically restored in the p53M214K line, and brain development recovered to return movement behavior to normal. Our findings suggest that Bad is required for (PCD) control, exerting a p53 stress signal on caspase-8/tBid-mediated death signaling and brain development-related gene regulation.


Asunto(s)
Apoptosis/genética , Encéfalo/embriología , Encéfalo/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteína Letal Asociada a bcl/genética , Animales , Animales Modificados Genéticamente , Caspasa 8/metabolismo , Regulación hacia Abajo/genética , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genoma , Mutación con Pérdida de Función/genética , Morfogénesis/genética , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Natación , Proteínas de Pez Cebra/metabolismo , Proteína Letal Asociada a bcl/metabolismo
7.
Cells ; 10(11)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34831305

RESUMEN

The WW domain-containing oxidoreductase (WWOX) gene was originally discovered as a putative tumor suppressor spanning the common fragile site FRA16D, but as time has progressed the extent of its pleiotropic function has become apparent. At present, WWOX is a major source of interest in the context of neurological disorders, and more specifically developmental and epileptic encephalopathies (DEEs). This review article aims to introduce the many model systems used through the years to study its function and roles in neuropathies. Similarities and fundamental differences between rodent and human models are discussed. Finally, future perspectives and promising research avenues are suggested.


Asunto(s)
Trastornos del Neurodesarrollo/metabolismo , Oxidorreductasa que Contiene Dominios WW/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Humanos , Mutación con Pérdida de Función/genética , Investigación Biomédica Traslacional
8.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(12): 159030, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34419589

RESUMEN

In the liver, contact sites between the endoplasmic reticulum (ER) and mitochondria (named MAMs) may be crucial hubs for the regulation of lipid metabolism, thus contributing to the exacerbation or prevention of fatty liver. We hypothesized that tether proteins located at MAMs could play a key role in preventing triglyceride accumulation in hepatocytes and nonalcoholic fatty liver disease (NAFLD) occurrence. To test this, we explored the role of two key partners in building MAM integrity and functionality, the glucose-regulated protein 75 (Grp75) and mitofusin 2 (Mfn2), which liver contents are altered in obesity and NAFLD. Grp75 or Mfn2 expression was either silenced using siRNA or overexpressed with adenoviruses in Huh7 cells. Silencing of Grp75 and Mfn2 resulted in decreased ER-mitochondria interactions, mitochondrial network fusion state and mitochondrial oxidative capacity, while overexpression of the two proteins induced mirror impacts on these parameters. Furthermore, Grp75 or Mfn2 silencing decreased cellular cholesterol content and enhanced triglyceride secretion in ApoB100 lipoproteins, while their overexpression led to reverse effects. Cellular phosphatidylcholine/phosphatidylethanolamine ratio was decreased only upon overexpression of the proteins, potentially contributing to altered ApoB100 assembly and secretion. Despite the opposite differences, both silencing and overexpression of Grp75 or Mfn2 induced triglyceride storage, although a fatty acid challenge was required to express the alteration upon protein silencing. Among the mechanisms potentially involved in this phenotype, ER stress was closely associated with altered triglyceride metabolism after Grp75 or Mfn2 overexpression, while blunted mitochondrial FA oxidation capacity may be the main defect causing triglyceride accumulation upon Grp75 or Mfn2 silencing. Further studies are required to decipher the link between modulation of Grp75 or Mfn2 expression, change in MAM integrity and alteration of cholesterol content of the cell. In conclusion, Grp75 or Mfn2 silencing and overexpression in Huh7 cells contribute to altering MAM integrity and cholesterol storage in opposite directions, but all promote triglyceride accumulation through distinct cellular pathways. This study also highlights that besides Mfn2, Grp75 could play a central role in hepatic lipid and cholesterol metabolism in obesity and NAFLD.


Asunto(s)
Apolipoproteína B-100/genética , Colesterol/metabolismo , GTP Fosfohidrolasas/genética , Proteínas HSP70 de Choque Térmico/genética , Proteínas Mitocondriales/genética , Enfermedad del Hígado Graso no Alcohólico/genética , Línea Celular , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , GTP Fosfohidrolasas/antagonistas & inhibidores , Mutación con Ganancia de Función/genética , Regulación de la Expresión Génica/genética , Silenciador del Gen , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Mutación con Pérdida de Función/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Triglicéridos/metabolismo
9.
Cell ; 184(17): 4447-4463.e20, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34363755

RESUMEN

TANK binding kinase 1 (TBK1) regulates IFN-I, NF-κB, and TNF-induced RIPK1-dependent cell death (RCD). In mice, biallelic loss of TBK1 is embryonically lethal. We discovered four humans, ages 32, 26, 7, and 8 from three unrelated consanguineous families with homozygous loss-of-function mutations in TBK1. All four patients suffer from chronic and systemic autoinflammation, but not severe viral infections. We demonstrate that TBK1 loss results in hypomorphic but sufficient IFN-I induction via RIG-I/MDA5, while the system retains near intact IL-6 induction through NF-κB. Autoinflammation is driven by TNF-induced RCD as patient-derived fibroblasts experienced higher rates of necroptosis in vitro, and CC3 was elevated in peripheral blood ex vivo. Treatment with anti-TNF dampened the baseline circulating inflammatory profile and ameliorated the clinical condition in vivo. These findings highlight the plasticity of the IFN-I response and underscore a cardinal role for TBK1 in the regulation of RCD.


Asunto(s)
Inflamación/enzimología , Proteínas Serina-Treonina Quinasas/deficiencia , Factor de Necrosis Tumoral alfa/farmacología , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Autoinmunidad/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Muerte Celular/efectos de los fármacos , Citocinas/metabolismo , Enzima Desubiquitinante CYLD/metabolismo , Femenino , Células HEK293 , Homocigoto , Humanos , Quinasa I-kappa B/metabolismo , Inmunofenotipificación , Inflamación/patología , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Mutación con Pérdida de Función/genética , Masculino , Linaje , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Receptor Toll-Like 3/metabolismo , Transcriptoma/genética , Vesiculovirus/efectos de los fármacos , Vesiculovirus/fisiología
10.
Am J Med Genet A ; 185(12): 3675-3682, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34272929

RESUMEN

Pathogenic variation in the X-linked gene FLNA causes a wide range of human developmental phenotypes. Loss-of-function is usually male embryonic-lethal, and most commonly results in a neuronal migration disorder in affected females. Gain-of-function variants cause a spectrum of skeletal dysplasias that present with variable additional, often distinctive, soft-tissue anomalies in males and females. Here we present two, unrelated, male individuals with novel, intronic variants in FLNA that are predicted to be pathogenic. Their phenotypes are reminiscent of the gain-of-function spectrum without the skeletal manifestations. Most strikingly, they manifest urethral anomalies, cardiac malformations, and keloid scarring, all commonly encountered features of frontometaphyseal dysplasia. Both variants prevent inclusion of exon 40 into the FLNA transcript, predicting the in-frame deletion of 42 amino acids, however the abundance of FLNA protein was equivalent to that observed in healthy individuals. Loss of these 42 amino acids removes sites that mediate key FLNA functions, including binding of some ligands and phosphorylation. This phenotype further expands the spectrum of the FLNA filaminopathies.


Asunto(s)
Filaminas/genética , Frente/anomalías , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Predisposición Genética a la Enfermedad , Osteocondrodisplasias/genética , Niño , Cicatriz/complicaciones , Cicatriz/genética , Cicatriz/fisiopatología , Exones/genética , Frente/fisiopatología , Genes Ligados a X , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Variación Genética/genética , Humanos , Lactante , Queloide/complicaciones , Queloide/genética , Queloide/fisiopatología , Mutación con Pérdida de Función/genética , Masculino , Mutación/genética , Osteocondrodisplasias/fisiopatología , Linaje , Fenotipo , Fosforilación/genética , Uretra/anomalías , Uretra/fisiopatología
11.
Taiwan J Obstet Gynecol ; 60(3): 530-533, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33966742

RESUMEN

OBJECTIVE: We present a novel homozygous splice site mutation in the PIGN gene identified by whole exome sequencing and explored the genotype-phenotype correlation. CASE REPORT: A healthy 32-year-old woman underwent an ultrasound at 13 + 5 weeks of gestation. The ultrasound revealed multiple anomalies again including cystic hygroma, omphalocele and a ventricular septal defect. The pregnancy was subsequently terminated, and whole exome sequencing revealed a novel homozygous splice site mutation in the PIGN gene c.963 G > A (p.Gln321Gln). The same variant was also detected by pedigree-based Sanger sequencing in both parents as heterozygous, while they had normal karyotypes. CONCLUSION: Our case report enhances the phenotype-genotype correlation associated with homozygous loss of function mutations in the PIGN gene.


Asunto(s)
Anomalías Múltiples/diagnóstico , ADN Recombinante/genética , Mutación con Pérdida de Función/genética , Fosfotransferasas/genética , Ultrasonografía Prenatal , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Aborto Eugénico , Adulto , Femenino , Estudios de Asociación Genética , Homocigoto , Humanos , Linaje , Embarazo , Secuenciación del Exoma
12.
Hum Mol Genet ; 30(10): 908-922, 2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-33822942

RESUMEN

Mucolipidosis IV (MLIV) is an orphan disease leading to debilitating psychomotor deficits and vision loss. It is caused by loss-of-function mutations in the MCOLN1 gene that encodes the lysosomal transient receptor potential channel mucolipin1, or TRPML1. With no existing therapy, the unmet need in this disease is very high. Here, we showed that AAV-mediated CNS-targeted gene transfer of the human MCOLN1 gene rescued motor function and alleviated brain pathology in the MLIV mouse model. Using the AAV-PHP.b vector in symptomatic mice, we showed long-term reversal of declined motor function and significant delay of paralysis. Next, using self-complementary AAV9 clinical candidate vector, we showed that its intracerebroventricular administration in post-natal day 1 mice significantly improved motor function, myelination and reduced lysosomal storage load in the MLIV mouse brain. Based on our data and general advancements in the gene therapy field, we propose scAAV9-mediated CSF-targeted MCOLN1 gene transfer as a therapeutic strategy in MLIV.


Asunto(s)
Terapia Genética , Mucolipidosis/terapia , Enfermedades del Sistema Nervioso/terapia , Canales de Potencial de Receptor Transitorio/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Humanos , Mutación con Pérdida de Función/genética , Lisosomas/genética , Lisosomas/patología , Ratones , Mucolipidosis/líquido cefalorraquídeo , Mucolipidosis/genética , Mucolipidosis/patología , Enfermedades del Sistema Nervioso/líquido cefalorraquídeo , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/patología
13.
Am J Med Genet A ; 185(6): 1649-1665, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33783954

RESUMEN

Wiedemann-Steiner syndrome (WSS) is an autosomal dominant disorder caused by monoallelic variants in KMT2A and characterized by intellectual disability and hypertrichosis. We performed a retrospective, multicenter, observational study of 104 individuals with WSS from five continents to characterize the clinical and molecular spectrum of WSS in diverse populations, to identify physical features that may be more prevalent in White versus Black Indigenous People of Color individuals, to delineate genotype-phenotype correlations, to define developmental milestones, to describe the syndrome through adulthood, and to examine clinicians' differential diagnoses. Sixty-nine of the 82 variants (84%) observed in the study were not previously reported in the literature. Common clinical features identified in the cohort included: developmental delay or intellectual disability (97%), constipation (63.8%), failure to thrive (67.7%), feeding difficulties (66.3%), hypertrichosis cubiti (57%), short stature (57.8%), and vertebral anomalies (46.9%). The median ages at walking and first words were 20 months and 18 months, respectively. Hypotonia was associated with loss of function (LoF) variants, and seizures were associated with non-LoF variants. This study identifies genotype-phenotype correlations as well as race-facial feature associations in an ethnically diverse cohort, and accurately defines developmental trajectories, medical comorbidities, and long-term outcomes in individuals with WSS.


Asunto(s)
Predisposición Genética a la Enfermedad , Trastornos del Crecimiento/genética , N-Metiltransferasa de Histona-Lisina/genética , Hipertricosis/congénito , Discapacidad Intelectual/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Población Negra/genética , Estreñimiento/epidemiología , Estreñimiento/genética , Estreñimiento/patología , Insuficiencia de Crecimiento/epidemiología , Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/patología , Estudios de Asociación Genética , Trastornos del Crecimiento/epidemiología , Trastornos del Crecimiento/patología , Humanos , Hipertricosis/epidemiología , Hipertricosis/genética , Hipertricosis/patología , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/patología , Mutación con Pérdida de Función/genética , Estudios Retrospectivos , Población Blanca/genética
14.
Sci Rep ; 11(1): 5838, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712646

RESUMEN

Chemotherapy resistance is the main impediment in the treatment of acute myeloid leukaemia (AML). Despite rapid advances, the various mechanisms inducing resistance development remain to be defined in detail. Here we report that loss-of-function mutations (LOF) in the histone methyltransferase EZH2 have the potential to confer resistance against the chemotherapeutic agent cytarabine. We identify seven distinct EZH2 mutations leading to loss of H3K27 trimethylation via multiple mechanisms. Analysis of matched diagnosis and relapse samples reveal a heterogenous regulation of EZH2 and a loss of EZH2 in 50% of patients. We confirm that loss of EZH2 induces resistance against cytarabine in the cell lines HEK293T and K562 as well as in a patient-derived xenograft model. Proteomics and transcriptomics analysis reveal that resistance is conferred by upregulation of multiple direct and indirect EZH2 target genes that are involved in apoptosis evasion, augmentation of proliferation and alteration of transmembrane transporter function. Our data indicate that loss of EZH2 results in upregulation of its target genes, providing the cell with a selective growth advantage, which mediates chemotherapy resistance.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación con Pérdida de Función/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/diagnóstico , Recurrencia Local de Neoplasia/patología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Cutan Pathol ; 48(7): 877-883, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33543489

RESUMEN

BACKGROUND: Filaggrin is a protein integral to the structure and function of the epidermis. Filaggrin (FLG) loss-of-function (LOF) mutations are common and increase the risk of developing atopic dermatitis (AD) and ichthyosis vulgaris (IV). Epidemiologic data suggest a link between skin cancer and AD. We examined if FLG staining pattern can be used to characterize cutaneous squamous cell carcinomas (SCC), basal cell carcinomas (BCC), and reactive squamous epithelium. METHODS: Tissue microarrays (TMAs) were created from 196 cases of formalin-fixed paraffin-embedded (FFPE) SCC and 144 BCC cases. TMAs and sections of reactive squamous epithelium were stained with optimized anti-FLG antibody and evaluated for FLG expression (normal, abnormal, or negative). RESULTS: FLG was absent in poorly differentiated (PD) compared to well-differentiated (WD) SCC (P < .0001) and moderately-differentiated (MD) (P = .0231) SCC, and in MD compared to WD SCC (P = .0099). Abnormal staining was significantly increased in PD compared to WD cases (P = .0039) and in MD compared to WD cases (P = .0006). Most BCC did not exhibit FLG expression (P < .05). Reactive squamous epithelium demonstrated normal, but exaggerated FLG expression. CONCLUSIONS: Our findings demonstrate the differences in FLG expression patterns in types of keratinocyte carcinomas and their mimickers.


Asunto(s)
Carcinoma Basocelular/diagnóstico , Carcinoma de Células Escamosas/diagnóstico , Inmunohistoquímica/métodos , Proteínas de Filamentos Intermediarios/genética , Neoplasias Cutáneas/patología , Anciano , Carcinoma Basocelular/genética , Carcinoma de Células Escamosas/genética , Diferenciación Celular/genética , Dermatitis Atópica/epidemiología , Dermatitis Atópica/genética , Dermatitis Atópica/metabolismo , Dermatitis Atópica/patología , Epidermis/metabolismo , Epidermis/patología , Femenino , Proteínas Filagrina , Predisposición Genética a la Enfermedad , Humanos , Ictiosis Vulgar/epidemiología , Ictiosis Vulgar/genética , Ictiosis Vulgar/metabolismo , Ictiosis Vulgar/patología , Proteínas de Filamentos Intermediarios/inmunología , Mutación con Pérdida de Función/genética , Masculino , Coloración y Etiquetado/métodos , Análisis de Matrices Tisulares/métodos
16.
Am J Med Genet A ; 185(4): 1275-1281, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33527719

RESUMEN

Individuals carrying biallelic loss-of-function mutations in PCDH12 have been reported with three different conditions: the diencephalic-mesencephalic junction dysplasia syndrome 1 (DMJDS1), a disorder characterized by global developmental delay, microcephaly, dystonia, and a midbrain malformation at the diencephalic-mesencephalic junction; cerebral palsy combined with a neurodevelopmental disorder; and cerebellar ataxia with retinopathy. We report an additional patient carrying a homozygous PCDH12 frameshift, whose anamnesis combines the most recurrent DMJDS1 clinical features, that is, global developmental delay, microcephaly, and ataxia, with exudative vitreoretinopathy. This case and previously published DMJDS1 patients presenting with nonspecific visual impairments and ophthalmic disorders suggest that ophthalmic alterations are an integral part of clinical features associated with PCDH12 loss-of-function.


Asunto(s)
Ataxia/genética , Cadherinas/genética , Discapacidades del Desarrollo/genética , Microcefalia/genética , Adolescente , Adulto , Ataxia/diagnóstico , Ataxia/patología , Niño , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/patología , Diencéfalo/diagnóstico por imagen , Diencéfalo/patología , Femenino , Homocigoto , Humanos , Mutación con Pérdida de Función/genética , Masculino , Microcefalia/diagnóstico , Microcefalia/patología , Malformaciones del Sistema Nervioso/diagnóstico , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Linaje , Protocadherinas , Enfermedades de la Retina/diagnóstico por imagen , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología
17.
J Biol Chem ; 296: 100445, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33617877

RESUMEN

Within the AGC kinase superfamily, gene fusions resulting from chromosomal rearrangements have been most frequently described for protein kinase C (PKC), with gene fragments encoding either the C-terminal catalytic domain or the N-terminal regulatory moiety fused to other genes. Kinase fusions that eliminate regulatory domains are typically gain of function and often oncogenic. However, several quality control pathways prevent accumulation of aberrant PKC, suggesting that PKC fusions may paradoxically be loss of function. To explore this topic, we used biochemical, cellular, and genome editing approaches to investigate the function of fusions that retain the portion of the gene encoding either the catalytic domain or regulatory domain of PKC. Overexpression studies revealed that PKC catalytic domain fusions were constitutively active but vulnerable to degradation. Genome editing of endogenous genes to generate a cancer-associated PKC fusion resulted in cells with detectable levels of fusion transcript but no detectable protein. Hence, PKC catalytic domain fusions are paradoxically loss of function as a result of their instability, preventing appreciable accumulation of protein in cells. Overexpression of a PKC regulatory domain fusion suppressed both basal and agonist-induced endogenous PKC activity, acting in a dominant-negative manner by competing for diacylglycerol. For both catalytic and regulatory domain fusions, the PKC component of the fusion proteins mediated the effects of the full-length fusions on the parameters examined, suggesting that the partner protein is dispensable in these contexts. Taken together, our findings reveal that PKC gene fusions are distinct from oncogenic fusions and present a mechanism by which loss of PKC function occurs in cancer.


Asunto(s)
Neoplasias/metabolismo , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Animales , Sitios de Unión , Células COS , Dominio Catalítico , Línea Celular Tumoral , Chlorocebus aethiops , Diglicéridos/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Humanos , Mutación con Pérdida de Función/genética , Fosforilación , Dominios Proteicos , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
18.
J Exp Med ; 218(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33606008

RESUMEN

Juvenile idiopathic arthritis is the most common chronic rheumatic disease in children, and its etiology remains poorly understood. Here, we explored four families with early-onset arthritis carrying homozygous loss-of-expression mutations in LACC1. To understand the link between LACC1 and inflammation, we performed a functional study of LACC1 in human immune cells. We showed that LACC1 was primarily expressed in macrophages upon mTOR signaling. We found that LACC1 deficiency had no obvious impact on inflammasome activation, type I interferon response, or NF-κB regulation. Using bimolecular fluorescence complementation and biochemical assays, we showed that autophagy-inducing proteins, RACK1 and AMPK, interacted with LACC1. Autophagy blockade in macrophages was associated with LACC1 cleavage and degradation. Moreover, LACC1 deficiency reduced autophagy flux in primary macrophages. This was associated with a defect in the accumulation of lipid droplets and mitochondrial respiration, suggesting that LACC1-dependent autophagy fuels macrophage bioenergetics metabolism. Altogether, LACC1 deficiency defines a novel form of genetically inherited juvenile arthritis associated with impaired autophagy in macrophages.


Asunto(s)
Artritis Juvenil/metabolismo , Artritis Juvenil/patología , Autofagia , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Macrófagos/metabolismo , Adenilato Quinasa/metabolismo , Adolescente , Secuencia de Aminoácidos , Apoptosis/efectos de los fármacos , Artritis Juvenil/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Bacterias/metabolismo , Diferenciación Celular/efectos de los fármacos , Niño , Exoma/genética , Femenino , Homocigoto , Humanos , Inflamasomas/metabolismo , Inflamación/complicaciones , Inflamación/patología , Interferones/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Mutación con Pérdida de Función/genética , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Monocitos/efectos de los fármacos , Monocitos/patología , FN-kappa B/metabolismo , Linaje , Proteómica , Receptores de Cinasa C Activada/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Adulto Joven
19.
Arthritis Rheumatol ; 73(8): 1543-1549, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33559312

RESUMEN

OBJECTIVE: The gene TNFRSF11B encodes for osteoprotegerin (OPG) and was recently identified as the CCAL1 locus associated with familial calcium pyrophosphate deposition disease (CPDD). While the CCAL1 OPG mutation (OPG-XL) was originally believed to be a gain-of-function mutation, loss of OPG activity causes arthritis-associated osteolysis in mice, which is likely related to excess subchondral osteoclast formation and/or activity. The purpose of the present study was to further explore the effect of OPG-XL in osteoclastogenesis. METHODS: The effects of recombinant OPG-XL and wild-type (WT) OPG were determined in monoculture and coculture models of RANKL-induced osteoclastogenesis. The effects of OPG-XL on osteoclast survival as well as on TRAIL-induced apoptosis were determined using standard in vitro assays and compared to WT OPG. The ability of OPG-XL and WT OPG to bind to osteoblasts was measured with enzyme-linked immunosorbent assay and flow cytometry using the osteoblastic MC3T3-E1 cell line. RESULTS: OPG-XL was less effective than WT OPG at blocking RANKL-induced osteoclastogenesis in monoculture and coculture models. Osteoclast survival and inhibition of TRAIL-induced apoptosis were similar in the presence of OPG-XL and WT OPG. Compared to WT OPG, considerably less OPG-XL bound to cells. CONCLUSION: These findings indicate that OPG-XL is a loss-of-function mutation as it relates to RANKL-mediated osteoclastogenesis, and thus may permit increased osteoclast numbers and heightened bone turnover. Further studies are necessary to demonstrate how this mutation contributes to arthritis in individuals carrying this mutation.


Asunto(s)
Condrocalcinosis/genética , Mutación con Pérdida de Función/genética , Osteogénesis/genética , Osteoprotegerina/genética , Animales , Remodelación Ósea/genética , Línea Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ratones
20.
Rheumatology (Oxford) ; 60(10): 4888-4898, 2021 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33493343

RESUMEN

OBJECTIVE: To investigate phenotypic and molecular characteristics of a consanguineous family with autosomal-recessive, polyarticular, juvenile isiopathic arthriris (JIA) with extra-articular manifestations, including renal amyloidosis and Crohn's disease, associated with a novel homozygous truncating variant in LACC1. METHODS: Whole exome sequencing (WES) or targeted Sanger verification were performed in 15 participants. LACC1 expression and cytokine array were analysed in patient-derived and CRISPR/Cas9-generated LACC1-knockout macrophages (Mϕ). RESULTS: A homozygous truncating variant (p.Glu348Ter) in LACC1 was identified in three affected and one asymptomatic family member, and predicted harmful by causing premature stop of the LACC1 protein sequences, and by absence from ethnically-matched controls and public variation databases. Expression studies in patient-derived macrophages (Mϕ) showed no endogenous p.Glu348Ter-LACC1 RNA transcription or protein expression, compatible with nonsense-mediated mRNA decay. WES analysis in the asymptomatic homozygous subject for p. Glu348Ter-LACC1 detected an exclusive heterozygous variant (p.Arg928Gln) in complement component C5. Further complement activity analysis suggested a protective role for the p.Arg928Gln-C5 variant as a phenotypic modifier of LACC1-associated disease. Finally, cytokine profile analysis indicated increased levels of pro-inflammatory cytokines in LACC1-disrupted as compared with wild-type Mϕ. CONCLUSIONS: Our findings reinforce the role of LACC1 disruption in autosomal-recessive JIA, extend the clinical spectrum and intra-familial heterogeneity of the disease-associated phenotype, indicate a modulatory effect of complement factor C5 on phenotypic severity, and suggest an inhibitory role for wild-type LACC1 on pro-inflammatory pathways.


Asunto(s)
Artritis Juvenil/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación con Pérdida de Función/genética , Adolescente , Adulto , Artritis Juvenil/patología , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Citocinas/sangre , Femenino , Citometría de Flujo , Edición Génica , Humanos , Immunoblotting , Masculino , Linaje , Secuenciación del Exoma , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA