Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Cell ; 182(2): 297-316.e27, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32619424

RESUMEN

The most aggressive B cell lymphomas frequently manifest extranodal distribution and carry somatic mutations in the poorly characterized gene TBL1XR1. Here, we show that TBL1XR1 mutations skew the humoral immune response toward generating abnormal immature memory B cells (MB), while impairing plasma cell differentiation. At the molecular level, TBL1XR1 mutants co-opt SMRT/HDAC3 repressor complexes toward binding the MB cell transcription factor (TF) BACH2 at the expense of the germinal center (GC) TF BCL6, leading to pre-memory transcriptional reprogramming and cell-fate bias. Upon antigen recall, TBL1XR1 mutant MB cells fail to differentiate into plasma cells and instead preferentially reenter new GC reactions, providing evidence for a cyclic reentry lymphomagenesis mechanism. Ultimately, TBL1XR1 alterations lead to a striking extranodal immunoblastic lymphoma phenotype that mimics the human disease. Both human and murine lymphomas feature expanded MB-like cell populations, consistent with a MB-cell origin and delineating an unforeseen pathway for malignant transformation of the immune system.


Asunto(s)
Memoria Inmunológica/fisiología , Linfoma de Células B Grandes Difuso/patología , Proteínas Nucleares/genética , Células Precursoras de Linfocitos B/inmunología , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Represoras/genética , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Cromatina/química , Cromatina/metabolismo , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Co-Represor 2 de Receptor Nuclear/química , Co-Represor 2 de Receptor Nuclear/metabolismo , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-6/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Transcripción Genética
2.
Nat Immunol ; 22(2): 240-253, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33432228

RESUMEN

During the germinal center (GC) reaction, B cells undergo extensive redistribution of cohesin complex and three-dimensional reorganization of their genomes. Yet, the significance of cohesin and architectural programming in the humoral immune response is unknown. Herein we report that homozygous deletion of Smc3, encoding the cohesin ATPase subunit, abrogated GC formation, while, in marked contrast, Smc3 haploinsufficiency resulted in GC hyperplasia, skewing of GC polarity and impaired plasma cell (PC) differentiation. Genome-wide chromosomal conformation and transcriptional profiling revealed defects in GC B cell terminal differentiation programs controlled by the lymphoma epigenetic tumor suppressors Tet2 and Kmt2d and failure of Smc3-haploinsufficient GC B cells to switch from B cell- to PC-defining transcription factors. Smc3 haploinsufficiency preferentially impaired the connectivity of enhancer elements controlling various lymphoma tumor suppressor genes, and, accordingly, Smc3 haploinsufficiency accelerated lymphomagenesis in mice with constitutive Bcl6 expression. Collectively, our data indicate a dose-dependent function for cohesin in humoral immunity to facilitate the B cell to PC phenotypic switch while restricting malignant transformation.


Asunto(s)
Linfocitos B/metabolismo , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteínas Cromosómicas no Histona/deficiencia , Proteínas Cromosómicas no Histona/genética , Dosificación de Gen , Centro Germinal/metabolismo , Inmunidad Humoral , Linfoma de Células B/genética , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Proteoglicanos Tipo Condroitín Sulfato/deficiencia , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Centro Germinal/inmunología , Centro Germinal/patología , Haploinsuficiencia , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Cohesinas
3.
Nature ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862028

RESUMEN

Spaceflight induces molecular, cellular and physiological shifts in astronauts and poses myriad biomedical challenges to the human body, which are becoming increasingly relevant as more humans venture into space1-6. Yet current frameworks for aerospace medicine are nascent and lag far behind advancements in precision medicine on Earth, underscoring the need for rapid development of space medicine databases, tools and protocols. Here we present the Space Omics and Medical Atlas (SOMA), an integrated data and sample repository for clinical, cellular and multi-omic research profiles from a diverse range of missions, including the NASA Twins Study7, JAXA CFE study8,9, SpaceX Inspiration4 crew10-12, Axiom and Polaris. The SOMA resource represents a more than tenfold increase in publicly available human space omics data, with matched samples available from the Cornell Aerospace Medicine Biobank. The Atlas includes extensive molecular and physiological profiles encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics and microbiome datasets, which reveal some consistent features across missions, including cytokine shifts, telomere elongation and gene expression changes, as well as mission-specific molecular responses and links to orthologous, tissue-specific mouse datasets. Leveraging the datasets, tools and resources in SOMA can help to accelerate precision aerospace medicine, bringing needed health monitoring, risk mitigation and countermeasure data for upcoming lunar, Mars and exploration-class missions.

4.
Nature ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862027

RESUMEN

The recent acceleration of commercial, private and multi-national spaceflight has created an unprecedented level of activity in low Earth orbit, concomitant with the largest-ever number of crewed missions entering space and preparations for exploration-class (lasting longer than one year) missions. Such rapid advancement into space from many new companies, countries and space-related entities has enabled a 'second space age'. This era is also poised to leverage, for the first time, modern tools and methods of molecular biology and precision medicine, thus enabling precision aerospace medicine for the crews. The applications of these biomedical technologies and algorithms are diverse, and encompass multi-omic, single-cell and spatial biology tools to investigate human and microbial responses to spaceflight. Additionally, they extend to the development of new imaging techniques, real-time cognitive assessments, physiological monitoring and personalized risk profiles tailored for astronauts. Furthermore, these technologies enable advancements in pharmacogenomics, as well as the identification of novel spaceflight biomarkers and the development of corresponding countermeasures. In this Perspective, we highlight some of the recent biomedical research from the National Aeronautics and Space Administration, Japan Aerospace Exploration Agency, European Space Agency and other space agencies, and detail the entrance of the commercial spaceflight sector (including SpaceX, Blue Origin, Axiom and Sierra Space) into aerospace medicine and space biology, the first aerospace medicine biobank, and various upcoming missions that will utilize these tools to ensure a permanent human presence beyond low Earth orbit, venturing out to other planets and moons.

5.
Nature ; 589(7841): 299-305, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33299181

RESUMEN

Linker histone H1 proteins bind to nucleosomes and facilitate chromatin compaction1, although their biological functions are poorly understood. Mutations in the genes that encode H1 isoforms B-E (H1B, H1C, H1D and H1E; also known as H1-5, H1-2, H1-3 and H1-4, respectively) are highly recurrent in B cell lymphomas, but the pathogenic relevance of these mutations to cancer and the mechanisms that are involved are unknown. Here we show that lymphoma-associated H1 alleles are genetic driver mutations in lymphomas. Disruption of H1 function results in a profound architectural remodelling of the genome, which is characterized by large-scale yet focal shifts of chromatin from a compacted to a relaxed state. This decompaction drives distinct changes in epigenetic states, primarily owing to a gain of histone H3 dimethylation at lysine 36 (H3K36me2) and/or loss of repressive H3 trimethylation at lysine 27 (H3K27me3). These changes unlock the expression of stem cell genes that are normally silenced during early development. In mice, loss of H1c and H1e (also known as H1f2 and H1f4, respectively) conferred germinal centre B cells with enhanced fitness and self-renewal properties, ultimately leading to aggressive lymphomas with an increased repopulating potential. Collectively, our data indicate that H1 proteins are normally required to sequester early developmental genes into architecturally inaccessible genomic compartments. We also establish H1 as a bona fide tumour suppressor and show that mutations in H1 drive malignant transformation primarily through three-dimensional genome reorganization, which leads to epigenetic reprogramming and derepression of developmentally silenced genes.


Asunto(s)
Transformación Celular Neoplásica/genética , Cromatina/química , Cromatina/genética , Histonas/deficiencia , Histonas/genética , Linfoma/genética , Linfoma/patología , Alelos , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Autorrenovación de las Células , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes Supresores de Tumor , Centro Germinal/patología , Histonas/metabolismo , Humanos , Linfoma/metabolismo , Ratones , Mutación , Células Madre/metabolismo , Células Madre/patología
6.
Immunity ; 41(6): 1001-12, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526311

RESUMEN

Decreased HIV-specific CD8(+) T cell proliferation is a hallmark of chronic infection, but the mechanisms of decline are unclear. We analyzed gene expression profiles from antigen-stimulated HIV-specific CD8(+) T cells from patients with controlled and uncontrolled infection and identified caspase-8 as a correlate of dysfunctional CD8(+) T cell proliferation. Caspase-8 activity was upregulated in HIV-specific CD8(+) T cells from progressors and correlated positively with disease progression and programmed cell death-1 (PD-1) expression, but negatively with proliferation. In addition, progressor cells displayed a decreased ability to upregulate membrane-associated caspase-8 activity and increased necrotic cell death following antigenic stimulation, implicating the programmed cell death pathway necroptosis. In vitro necroptosis blockade rescued HIV-specific CD8(+) T cell proliferation in progressors, as did silencing of necroptosis mediator RIPK3. Thus, chronic stimulation leading to upregulated caspase-8 activity contributes to dysfunctional HIV-specific CD8(+) T cell proliferation through activation of necroptosis and increased cell death.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Caspasa 8/metabolismo , Infecciones por VIH/inmunología , VIH/fisiología , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T CD8-positivos/virología , Proliferación Celular/genética , Células Cultivadas , Progresión de la Enfermedad , Activación Enzimática , Regulación de la Expresión Génica , Proteína p24 del Núcleo del VIH/inmunología , Humanos , Necrosis , Fragmentos de Péptidos/inmunología , Receptor de Muerte Celular Programada 1/genética , ARN Interferente Pequeño/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transcriptoma , Carga Viral
7.
Prostaglandins Other Lipid Mediat ; 164: 106702, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36529320

RESUMEN

Soluble epoxide hydrolase (sEH) and fatty acid amide hydrolase (FAAH) are potential targets for several diseases. Previous studies have reported that concomitant selective inhibition of sEH and FAAH produced antinociception effects in an animal model of pain. However, the co-administration of a selective sEH inhibitor and a selective FAAH inhibitor might produce serious side effects due to drug-drug interactions that could complicate drug development in the long term. Thus, discovering dual sEH/FAAH inhibitors, single small molecules that can simultaneously inhibit both sEH and FAAH, would be a significant accomplishment in the medicinal chemistry field. Herein, we report the synthesis and biological evaluation of benzothiazole-phenyl-based analogs as potential dual sEH/FAAH inhibitors. This work represents a follow-up structure-activity relationship (SAR) and metabolic-stability studies of our best dual sEH/FAAH inhibitor identified previously, as well as in vivo evaluation of its effects on voluntary locomotor behavior in rats. Our SAR study indicates that trifluoromethyl groups on the aromatic rings are well tolerated by the targeted enzymes when placed at the ortho and para positions; however, they, surprisingly, did not improve metabolic stability in liver microsomes. Our behavioral studies indicate that doses of dual sEH/FAAH inhibitors that alleviate pain do not depress voluntary behavior in naïve rats, which is a common side effect of currently available analgesic drugs (e.g., opioids). Thus, dual sEH/FAAH inhibitors may be a safe and effective approach to treat pain.


Asunto(s)
Inhibidores Enzimáticos , Manejo del Dolor , Animales , Ratas , Benzotiazoles/farmacología , Benzotiazoles/uso terapéutico , Inhibidores Enzimáticos/farmacología , Dolor/tratamiento farmacológico , Relación Estructura-Actividad , Conducta Animal
8.
Proc Natl Acad Sci U S A ; 116(6): 2232-2236, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30674677

RESUMEN

Circulating tumor cells (CTCs) play a fundamental role in cancer progression. However, in mice, limited blood volume and the rarity of CTCs in the bloodstream preclude longitudinal, in-depth studies of these cells using existing liquid biopsy techniques. Here, we present an optofluidic system that continuously collects fluorescently labeled CTCs from a genetically engineered mouse model (GEMM) for several hours per day over multiple days or weeks. The system is based on a microfluidic cell sorting chip connected serially to an unanesthetized mouse via an implanted arteriovenous shunt. Pneumatically controlled microfluidic valves capture CTCs as they flow through the device, and CTC-depleted blood is returned back to the mouse via the shunt. To demonstrate the utility of our system, we profile CTCs isolated longitudinally from animals over 4 days of treatment with the BET inhibitor JQ1 using single-cell RNA sequencing (scRNA-Seq) and show that our approach eliminates potential biases driven by intermouse heterogeneity that can occur when CTCs are collected across different mice. The CTC isolation and sorting technology presented here provides a research tool to help reveal details of how CTCs evolve over time, allowing studies to credential changes in CTCs as biomarkers of drug response and facilitating future studies to understand the role of CTCs in metastasis.


Asunto(s)
Citometría de Flujo , Técnicas Analíticas Microfluídicas , Microfluídica , Neoplasias/diagnóstico , Neoplasias/metabolismo , Células Neoplásicas Circulantes/metabolismo , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Modelos Animales de Enfermedad , Citometría de Flujo/métodos , Perfilación de la Expresión Génica/métodos , Ratones , Microfluídica/métodos , Neoplasias/genética , Células Neoplásicas Circulantes/patología , Análisis de la Célula Individual/métodos , Transcriptoma
9.
J Neurooncol ; 147(3): 671-679, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32221785

RESUMEN

PURPOSE: Although non-enhancing lesions suspicious for glioma are usually assumed to be low grade glioma (LGG), some high grade glioma (HGG) do not enhance, which may lead to a delay in biopsy and/or resection, diagnosis, and treatment initiation. Thus, there is a clear need for a large-sample study that quantifies the rate of malignant, non-enhancing gliomas. METHODS: We retrospectively reviewed our series of 561 consecutive surgically treated gliomas with tissue diagnosis, 111 of which were non-enhancing, to determine the prevalence of high-grade histology in radiographically presumed LGG. Relative expression of tumor markers were also reported for non-enhancing lesions to investigate genetic correlates. RESULTS: We identified 561 surgically treated gliomas with tissue diagnosis from August 2012 to July 2018 and found that 111 patients (19.8%) demonstrated non-enhancing lesions suspicious for glioma on preoperative MRI. Thirty-one (27.9%) of the non-enhancing lesions were classified as HGGs (WHO Grade III or IV). Non-enhancing lesions were four times more likely to be HGG in patients older than 60 years than patients younger than 35 years (41.2% vs. 11.4%, Pearson Chi2 p < 0.001). Binomial logistic regression showed a significant inverse effect of age on the presence of IDH mutation in non-enhancing HGGs (p = 0.007). CONCLUSION: A clinically significant proportion (27.9%) of non-enhancing lesions were found to be HGG on final pathologic diagnosis. Thus, in patients with good functional and health status, especially those older than 60 years, we recommend obtaining tissue diagnosis of all lesions suspected to be glioma, even those that are non-enhancing, to guide diagnosis as well as early initiation of chemotherapy and radiation therapy.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/cirugía , Glioma/diagnóstico por imagen , Glioma/cirugía , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/epidemiología , Neoplasias Encefálicas/patología , Femenino , Glioma/epidemiología , Glioma/patología , Humanos , Incidencia , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
10.
Nature ; 484(7395): 519-23, 2012 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-22446628

RESUMEN

The 2009 H1N1 influenza pandemic showed the speed with which a novel respiratory virus can spread and the ability of a generally mild infection to induce severe morbidity and mortality in a subset of the population. Recent in vitro studies show that the interferon-inducible transmembrane (IFITM) protein family members potently restrict the replication of multiple pathogenic viruses. Both the magnitude and breadth of the IFITM proteins' in vitro effects suggest that they are critical for intrinsic resistance to such viruses, including influenza viruses. Using a knockout mouse model, we now test this hypothesis directly and find that IFITM3 is essential for defending the host against influenza A virus in vivo. Mice lacking Ifitm3 display fulminant viral pneumonia when challenged with a normally low-pathogenicity influenza virus, mirroring the destruction inflicted by the highly pathogenic 1918 'Spanish' influenza. Similar increased viral replication is seen in vitro, with protection rescued by the re-introduction of Ifitm3. To test the role of IFITM3 in human influenza virus infection, we assessed the IFITM3 alleles of individuals hospitalized with seasonal or pandemic influenza H1N1/09 viruses. We find that a statistically significant number of hospitalized subjects show enrichment for a minor IFITM3 allele (SNP rs12252-C) that alters a splice acceptor site, and functional assays show the minor CC genotype IFITM3 has reduced influenza virus restriction in vitro. Together these data reveal that the action of a single intrinsic immune effector, IFITM3, profoundly alters the course of influenza virus infection in mouse and humans.


Asunto(s)
Virus de la Influenza A/patogenicidad , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Proteínas de Unión al ARN/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Citocinas/inmunología , Inglaterra/epidemiología , Eliminación de Gen , Humanos , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/clasificación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza B/clasificación , Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/patogenicidad , Gripe Humana/complicaciones , Gripe Humana/epidemiología , Gripe Humana/mortalidad , Gripe Humana/virología , Leucocitos/inmunología , Pulmón/patología , Pulmón/virología , Proteínas de la Membrana/química , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/etiología , Neumonía Viral/patología , Neumonía Viral/prevención & control , Polimorfismo de Nucleótido Simple/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Escocia/epidemiología , Replicación Viral
11.
PLoS Pathog ; 10(4): e1004048, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699674

RESUMEN

Interferon-induced transmembrane proteins (IFITMs) inhibit infection of diverse enveloped viruses, including the influenza A virus (IAV) which is thought to enter from late endosomes. Recent evidence suggests that IFITMs block virus hemifusion (lipid mixing in the absence of viral content release) by altering the properties of cell membranes. Consistent with this mechanism, excess cholesterol in late endosomes of IFITM-expressing cells has been reported to inhibit IAV entry. Here, we examined IAV restriction by IFITM3 protein using direct virus-cell fusion assay and single virus imaging in live cells. IFITM3 over-expression did not inhibit lipid mixing, but abrogated the release of viral content into the cytoplasm. Although late endosomes of IFITM3-expressing cells accumulated cholesterol, other interventions leading to aberrantly high levels of this lipid did not inhibit virus fusion. These results imply that excess cholesterol in late endosomes is not the mechanism by which IFITM3 inhibits the transition from hemifusion to full fusion. The IFITM3's ability to block fusion pore formation at a post-hemifusion stage shows that this protein stabilizes the cytoplasmic leaflet of endosomal membranes without adversely affecting the lumenal leaflet. We propose that IFITM3 interferes with pore formation either directly, through partitioning into the cytoplasmic leaflet of a hemifusion intermediate, or indirectly, by modulating the lipid/protein composition of this leaflet. Alternatively, IFITM3 may redirect IAV fusion to a non-productive pathway, perhaps by promoting fusion with intralumenal vesicles within multivesicular bodies/late endosomes.


Asunto(s)
Endosomas/metabolismo , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Fusión de Membrana , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Internalización del Virus , Animales , Células CHO , Colesterol/genética , Colesterol/inmunología , Colesterol/metabolismo , Cricetinae , Cricetulus , Endosomas/genética , Endosomas/inmunología , Células HEK293 , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Gripe Humana/genética , Gripe Humana/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología
12.
J Shoulder Elbow Surg ; 25(11): 1774-1779, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27288274

RESUMEN

BACKGROUND: Research on optimal timing of bilateral anatomic total shoulder arthroplasty (TSA) is lacking. The purpose of this study was to investigate functional outcomes in patients undergoing bilateral anatomic TSA to understand the ideal timing for the second arthroplasty. METHODS: Patients who underwent bilateral TSA for osteoarthritis between 2000 and 2012 with a minimum follow-up of 12 months since their most recent surgery were evaluated. Postoperative patient-reported outcomes (University of California-Los Angeles [UCLA] shoulder rating scale, Constant score, and Simple Shoulder Test [SST]), biometrics (strength and range of motion), and a subjective questionnaire were compared for 4 "interval groups" based on timing between surgeries: <6 months, 6 to 12 months, 12 to 24 months, and >24 months. RESULTS: Eighty-two shoulders (41 patients, 70 ± 9 years old) were analyzed. Mean postoperative UCLA, Constant, and SST scores were 29, 72, and 9 points, respectively; 83% of patients reported satisfaction with both shoulders. Patients with <6 months between surgeries demonstrated significantly better UCLA scores than 6- to 12-month interval patients (P = .04), greater Constant scores compared with all other groups (P < .001), and greater SST scores compared with 6- to 12-month and 12- to 24-month interval patients (P = .002), with no differences in length of follow-up between groups. CONCLUSION: In the absence of extrinsic factors, such as convenience, changes in social support structure, or changes in health status, patients may be advised that having the second surgery within 6 months of the first might optimize their postoperative functional outcomes and satisfaction compared with waiting a longer interval between surgeries.


Asunto(s)
Artroplastía de Reemplazo de Hombro , Osteoartritis/cirugía , Articulación del Hombro/cirugía , Tiempo de Tratamiento , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Medición de Resultados Informados por el Paciente , Satisfacción del Paciente , Estudios Retrospectivos
13.
J Craniofac Surg ; 27(4): e348-50, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27171958

RESUMEN

Chronic rhinosinusitis (CRS) is a common health problem in the Western world. CRS is classified as CRS with (CRSwNP) and without (CRSsNP) nasal polyps. A less common third type is allergic fungal sinusitis, which often presents with polyps and, not infrequently, skull base erosion. Most patients are successfully managed with maximal medical therapy or endoscopic approaches. There are currently no reports of CRSwNPs resulting in fibro-osseous thickening and proptosis in the English literature. As such, the authors report a case of a 33-year-old man who underwent a craniofacial resection with drilling of the hyperostosed bone, which led to resolution of the proptosis and nasal symptoms. In an era where endoscopic surgery is the standard surgical approach for CRSwNP, this case highlights the need for open skullbase approaches for this condition due to the extensive and recalcitrant nature of the disease. While the majority of patients can be dealt with endoscopically, the authors highlight the importance of having the open approach in the otolaryngologists' armamentarium for patients of recalcitrant and extensive CRSwNP.


Asunto(s)
Craneotomía/métodos , Exoftalmia/etiología , Exoftalmia/cirugía , Pólipos Nasales/cirugía , Rinitis/cirugía , Sinusitis/cirugía , Adulto , Enfermedad Crónica , Endoscopía/métodos , Humanos , Imagen por Resonancia Magnética , Masculino , Base del Cráneo/cirugía
14.
J Craniofac Surg ; 27(6): 1532-4, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27428902

RESUMEN

With the use and efficacy of the vascularized nasoseptal flap, its indications are also expanding. Due to its relative ease of harvesting and no significant impairment in the long-term sinonasal quality of life, the flap has been used for a number of other purposes apart from its originally proposed use in reconstruction of the anterior cranial fossa, sella, and the clivus. Its use may negate the need of another incision to obtain fat or fascia. The authors describe the case of a 47-year-old lady who underwent endoscopic excision of a medially placed orbital intraconal hemangioma who presented to us with very poor vision in the left eye. The large medial orbital defect was reconstructed with a vascularized pedicled nasoseptal flap from the ipsilateral side. The patient made an excellent visual and sino-nasal recovery. This patient highlights a unique use for the proliferating indications for the use of the nasoseptal flap.


Asunto(s)
Tabique Nasal/cirugía , Órbita/cirugía , Procedimientos de Cirugía Plástica/métodos , Colgajos Quirúrgicos/cirugía , Oftalmopatías/cirugía , Femenino , Hemangioma/cirugía , Humanos , Persona de Mediana Edad
15.
J Virol ; 88(4): 2279-90, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24335308

RESUMEN

Human cytomegalovirus (HCMV) genome replication requires host DNA damage responses (DDRs) and raises the possibility that DNA repair pathways may influence viral replication. We report here that a nucleotide excision repair (NER)-associated-factor is required for efficient HCMV DNA replication. Mutations in genes encoding NER factors are associated with xeroderma pigmentosum (XP). One of the XP complementation groups, XPE, involves mutation in ddb2, which encodes DNA damage binding protein 2 (DDB2). Infectious progeny virus production was reduced by >2 logs in XPE fibroblasts compared to levels in normal fibroblasts. The levels of immediate early (IE) (IE2), early (E) (pp65), and early/late (E/L) (gB55) proteins were decreased in XPE cells. These replication defects were rescued by infection with a retrovirus expressing DDB2 cDNA. Similar patterns of reduced viral gene expression and progeny virus production were also observed in normal fibroblasts that were depleted for DDB2 by RNA interference (RNAi). Mature replication compartments (RCs) were nearly absent in XPE cells, and there were 1.5- to 2.0-log reductions in viral DNA loads in infected XPE cells relative to those in normal fibroblasts. The expression of viral genes (UL122, UL44, UL54, UL55, and UL84) affected by DDB2 status was also sensitive to a viral DNA replication inhibitor, phosphonoacetic acid (PAA), suggesting that DDB2 affects gene expression upstream of or events associated with the initiation of DNA replication. Finally, a novel, infection-associated feedback loop between DDB2 and ataxia telangiectasia mutated (ATM) was observed in infected cells. Together, these results demonstrate that DDB2 and a DDB2-ATM feedback loop influence HCMV replication.


Asunto(s)
Citomegalovirus/fisiología , Proteínas de Unión al ADN/genética , Retroalimentación Fisiológica/fisiología , Regulación Viral de la Expresión Génica/fisiología , Replicación Viral/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Citomegalovirus/genética , Técnica del Anticuerpo Fluorescente , Regulación Viral de la Expresión Génica/genética , Genes Inmediatos-Precoces/genética , Humanos , Immunoblotting , Mutación/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción Genética , Transfección , Replicación Viral/genética
16.
Arthroscopy ; 31(5): 896-900, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25660011

RESUMEN

PURPOSE: The purpose of the study was to (1) investigate the rate of return to play among Major League Baseball (MLB) athletes after anterior cruciate ligament reconstruction (ACLR), (2) determine the impact of ACL injury on ability to perform baseball-specific planting and pivoting tasks (batting and stealing bases), and (3) to explore the effect of the injured side on these metrics. METHODS: ACL injury data from 1999 to 2012 were compiled, along with player performance statistics recorded for players with at least 30 games before ACL injury. Predictor variables included side of injury and outcome variables focused on batting average, stolen bases, and number of times caught stealing before injury and after surgery. RESULTS: Twenty-three of 26 (88%) players were able to return to at least 30 games after ACLR, although they experienced a decline of 21.2% in number of games played (P = .004). Those who had a ACLR for a rear batting leg injury averaged a 12.3% decline in batting average, whereas those who had ACLR for a lead leg injury had a 6.4% increase in batting average (P = .04). Side of injury was not predictive of stolen base metrics. CONCLUSIONS: The overall rate of return to play among MLB position players after ACLR was 88%, although there was a 21.2% decline in the number of games played postoperatively. Injury to the rear batting leg resulted in a lower returning batting average compared with an injury to the lead batting leg. Side of injury had no effect on stolen bases or on the number of times a player was caught stealing. LEVEL OF EVIDENCE: Level IV, therapeutic case series.


Asunto(s)
Reconstrucción del Ligamento Cruzado Anterior , Ligamento Cruzado Anterior/cirugía , Béisbol/lesiones , Volver al Deporte , Adulto , Lesiones del Ligamento Cruzado Anterior , Atletas , Rendimiento Atlético , Humanos , Masculino , Estudios Retrospectivos
17.
J Biol Chem ; 288(24): 17261-71, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23649619

RESUMEN

Reoviruses are double-stranded RNA viruses that infect the mammalian respiratory and gastrointestinal tract. Reovirus infection elicits production of type I interferons (IFNs), which trigger antiviral pathways through the induction of interferon-stimulated genes (ISGs). Although hundreds of ISGs have been identified, the functions of many of these genes are unknown. The interferon-inducible transmembrane (IFITM) proteins are one class of ISGs that restrict the cell entry of some enveloped viruses, including influenza A virus. One family member, IFITM3, localizes to late endosomes, where reoviruses undergo proteolytic disassembly; therefore, we sought to determine whether IFITM3 also restricts reovirus entry. IFITM3-expressing cell lines were less susceptible to infection by reovirus, as they exhibited significantly lower percentages of infected cells in comparison to control cells. Reovirus replication was also significantly reduced in IFITM3-expressing cells. Additionally, cells expressing an shRNA targeting IFITM3 exhibited a smaller decrease in infection after IFN treatment than the control cells, indicating that endogenous IFITM3 restricts reovirus infection. However, IFITM3 did not restrict entry of reovirus infectious subvirion particles (ISVPs), which do not require endosomal proteolysis, indicating that restriction occurs in the endocytic pathway. Proteolysis of outer capsid protein µ1 was delayed in IFITM3-expressing cells in comparison to control cells, suggesting that IFITM3 modulates the function of late endosomal compartments either by reducing the activity of endosomal proteases or delaying the proteolytic processing of virions. These data provide the first evidence that IFITM3 restricts infection by a nonenveloped virus and suggest that IFITM3 targets an increasing number of viruses through a shared requirement for endosomes during cell entry.


Asunto(s)
Orthoreovirus Mamífero 3/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Unión al ARN/fisiología , Internalización del Virus , Cápside/metabolismo , Endocitosis , Endosomas/virología , Expresión Génica , Técnicas de Silenciamiento del Gen , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Interferón-alfa/fisiología , Cinética , Orthoreovirus de los Mamíferos/fisiología , ARN Interferente Pequeño/genética , Virión/fisiología , Ensamble de Virus , Replicación Viral
18.
Gastroenterology ; 144(7): 1438-49, 1449.e1-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23462180

RESUMEN

BACKGROUND & AIMS: Hepatitis C virus (HCV) infection is a leading cause of end-stage liver disease. Interferon-α (IFNα) is an important component of anti-HCV therapy; it up-regulates transcription of IFN-stimulated genes, many of which have been investigated for their antiviral effects. However, all of the genes required for the antiviral function of IFNα (IFN effector genes [IEGs]) are not known. IEGs include not only IFN-stimulated genes, but other nontranscriptionally induced genes that are required for the antiviral effect of IFNα. In contrast to candidate approaches based on analyses of messenger RNA (mRNA) expression, identification of IEGs requires a broad functional approach. METHODS: We performed an unbiased genome-wide small interfering RNA screen to identify IEGs that inhibit HCV. Huh7.5.1 hepatoma cells were transfected with small interfering RNAs incubated with IFNα and then infected with JFH1 HCV. Cells were stained using HCV core antibody, imaged, and analyzed to determine the percent infection. Candidate IEGs detected in the screen were validated and analyzed further. RESULTS: The screen identified 120 previously unreported IEGs. From these, we more fully evaluated the following: asparagine-linked glycosylation 10 homolog (yeast, α-1,2-glucosyltransferase); butyrylcholinesterase; dipeptidyl-peptidase 4 (CD26, adenosine deaminase complexing protein 2); glucokinase (hexokinase 4) regulator; guanylate cyclase 1, soluble, ß 3; MYST histone acetyltransferase 1; protein phosphatase 3 (formerly 2B), catalytic subunit, ß isoform; peroxisomal proliferator-activated receptor-γ-DBD-interacting protein 1; and solute carrier family 27 (fatty acid transporter), member 2; and demonstrated that they enabled IFNα-mediated suppression of HCV at multiple steps of its life cycle. Expression of these genes had more potent effects against flaviviridae because a subset was required for IFNα to suppress dengue virus but not influenza A virus. In addition, many of the host genes detected in this screen (92%) were not transcriptionally stimulated by IFNα; these genes represent a heretofore unknown class of non-IFN-stimulated gene IEGs. CONCLUSIONS: We performed a whole-genome loss-of-function screen to identify genes that mediate the effects of IFNα against human pathogenic viruses. We found that IFNα restricts HCV via actions of general and specific IEGs.


Asunto(s)
Antivirales/uso terapéutico , Hepacivirus/genética , Hepatitis C/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Replicación Viral/genética , Hepacivirus/efectos de los fármacos , Humanos , ARN Viral/genética , Replicación Viral/efectos de los fármacos
19.
J Virol ; 87(14): 7837-52, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23658454

RESUMEN

The interferon-induced transmembrane protein 3 (IFITM3) gene is an interferon-stimulated gene that inhibits the replication of multiple pathogenic viruses in vitro and in vivo. IFITM3 is a member of a large protein superfamily, whose members share a functionally undefined area of high amino acid conservation, the CD225 domain. We performed mutational analyses of IFITM3 and identified multiple residues within the CD225 domain, consisting of the first intramembrane domain (intramembrane domain 1 [IM1]) and a conserved intracellular loop (CIL), that are required for restriction of both influenza A virus (IAV) and dengue virus (DENV) infection in vitro. Two phenylalanines within IM1 (F75 and F78) also mediate a physical association between IFITM proteins, and the loss of this interaction decreases IFITM3-mediated restriction. By extension, similar IM1-mediated associations may contribute to the functions of additional members of the CD225 domain family. IFITM3's distal N-terminal domain is also needed for full antiviral activity, including a tyrosine (Y20), whose alteration results in mislocalization of a portion of IFITM3 to the cell periphery and surface. Comparative analyses demonstrate that similar molecular determinants are needed for IFITM3's restriction of both IAV and DENV. However, a portion of the CIL including Y99 and R87 is preferentially needed for inhibition of the orthomyxovirus. Several IFITM3 proteins engineered with rare single-nucleotide polymorphisms demonstrated reduced expression or mislocalization, and these events were associated with enhanced viral replication in vitro, suggesting that possessing such alleles may impact an individual's risk for viral infection. On the basis of this and other data, we propose a model for IFITM3-mediated restriction.


Asunto(s)
Virus del Dengue/fisiología , Virus de la Influenza A/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Replicación Viral/fisiología , Secuencia de Aminoácidos , Animales , Técnicas de Cultivo de Célula , Clonación Molecular , Secuencia Conservada/genética , Análisis Mutacional de ADN , ADN Complementario/genética , Perros , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Células de Riñón Canino Madin Darby , Espectrometría de Masas , Microscopía Confocal , Modelos Biológicos , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple/genética , Estructura Terciaria de Proteína/genética , Replicación Viral/genética
20.
Methods ; 59(2): 217-24, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23036328

RESUMEN

The use of genome wide RNA interference (RNAi) screens to investigate host-virals interactions has revealed unexpected connections that have improved our understanding of viral pathogenesis and cell biology. This work describes the use of an RNAi screening method employing an immunofluorescence image-based strategy and influenza A virus. We find this approach to be readily implemented, scalable and amenable to the direct evaluation of a variety of viral lifecycles.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Interferencia de ARN , Replicación Viral/genética , Células HeLa , Factores Celulares Derivados del Huésped/genética , Humanos , Virus de la Influenza A , ARN Interferente Pequeño/genética , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda