Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 350
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(5): 1238-1254.e14, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38367616

RESUMEN

CD4+ T cells with latent HIV-1 infection persist despite treatment with antiretroviral agents and represent the main barrier to a cure of HIV-1 infection. Pharmacological disruption of viral latency may expose HIV-1-infected cells to host immune activity, but the clinical efficacy of latency-reversing agents for reducing HIV-1 persistence remains to be proven. Here, we show in a randomized-controlled human clinical trial that the histone deacetylase inhibitor panobinostat, when administered in combination with pegylated interferon-α2a, induces a structural transformation of the HIV-1 reservoir cell pool, characterized by a disproportionate overrepresentation of HIV-1 proviruses integrated in ZNF genes and in chromatin regions with reduced H3K27ac marks, the molecular target sites for panobinostat. By contrast, proviruses near H3K27ac marks were actively selected against, likely due to increased susceptibility to panobinostat. These data suggest that latency-reversing treatment can increase the immunological vulnerability of HIV-1 reservoir cells and accelerate the selection of epigenetically privileged HIV-1 proviruses.


Asunto(s)
Infecciones por VIH , VIH-1 , Inhibidores de Histona Desacetilasas , Interferón-alfa , Panobinostat , Provirus , Humanos , Infecciones por VIH/tratamiento farmacológico , VIH-1/genética , Panobinostat/uso terapéutico , Provirus/efectos de los fármacos , Latencia del Virus , Inhibidores de Histona Desacetilasas/uso terapéutico , Interferón-alfa/uso terapéutico
3.
Cell ; 159(3): 469-70, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25417097

RESUMEN

Thaiss et al. report that the intestinal microbiota undergoes diurnal oscillation, which is controlled by host feeding time. Disruption of the host circadian clock induces dysbiosis, which is associated with host metabolic disorders.


Asunto(s)
Relojes Circadianos , Ritmo Circadiano , Intolerancia a la Glucosa , Microbiota , Animales , Humanos
4.
Nature ; 613(7945): 639-649, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36697862

RESUMEN

Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.


Asunto(s)
Biomasa , Contaminación de ADN , Feto , Microbiota , Animales , Femenino , Humanos , Embarazo , Líquido Amniótico/inmunología , Líquido Amniótico/microbiología , Mamíferos , Microbiota/genética , Placenta/inmunología , Placenta/microbiología , Feto/inmunología , Feto/microbiología , Reproducibilidad de los Resultados
5.
Nature ; 611(7937): 780-786, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36385534

RESUMEN

Enteric pathogens are exposed to a dynamic polymicrobial environment in the gastrointestinal tract1. This microbial community has been shown to be important during infection, but there are few examples illustrating how microbial interactions can influence the virulence of invading pathogens2. Here we show that expansion of a group of antibiotic-resistant, opportunistic pathogens in the gut-the enterococci-enhances the fitness and pathogenesis of Clostridioides difficile. Through a parallel process of nutrient restriction and cross-feeding, enterococci shape the metabolic environment in the gut and reprogramme C. difficile metabolism. Enterococci provide fermentable amino acids, including leucine and ornithine, which increase C. difficile fitness in the antibiotic-perturbed gut. Parallel depletion of arginine by enterococci through arginine catabolism provides a metabolic cue for C. difficile that facilitates increased virulence. We find evidence of microbial interaction between these two pathogenic organisms in multiple mouse models of infection and patients infected with C. difficile. These findings provide mechanistic insights into the role of pathogenic microbiota in the susceptibility to and the severity of C. difficile infection.


Asunto(s)
Clostridioides difficile , Enterococcus , Interacciones Microbianas , Animales , Humanos , Ratones , Antibacterianos/farmacología , Arginina/deficiencia , Arginina/metabolismo , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidad , Clostridioides difficile/fisiología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Enterococcus/efectos de los fármacos , Enterococcus/metabolismo , Enterococcus/patogenicidad , Enterococcus/fisiología , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Intestinos/microbiología , Leucina/metabolismo , Ornitina/metabolismo , Virulencia , Susceptibilidad a Enfermedades
6.
Nature ; 602(7897): 503-509, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110735

RESUMEN

The adoptive transfer of T lymphocytes reprogrammed to target tumour cells has demonstrated potential for treatment of various cancers1-7. However, little is known about the long-term potential and clonal stability of the infused cells. Here we studied long-lasting CD19-redirected chimeric antigen receptor (CAR) T cells in two patients with chronic lymphocytic leukaemia1-4 who achieved a complete remission in 2010. CAR T cells remained detectable more than ten years after infusion, with sustained remission in both patients. Notably, a highly activated CD4+ population emerged in both patients, dominating the CAR T cell population at the later time points. This transition was reflected in the stabilization of the clonal make-up of CAR T cells with a repertoire dominated by a small number of clones. Single-cell profiling demonstrated that these long-persisting CD4+ CAR T cells exhibited cytotoxic characteristics along with ongoing functional activation and proliferation. In addition, longitudinal profiling revealed a population of gamma delta CAR T cells that prominently expanded in one patient concomitant with CD8+ CAR T cells during the initial response phase. Our identification and characterization of these unexpected CAR T cell populations provide novel insight into the CAR T cell characteristics associated with anti-cancer response and long-term remission in leukaemia.


Asunto(s)
Linfocitos T CD4-Positivos , Inmunoterapia Adoptiva , Leucemia , Receptores Quiméricos de Antígenos , Antígenos CD19/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Separación Celular , Humanos , Leucemia/inmunología , Leucemia/terapia , Receptores Quiméricos de Antígenos/inmunología , Factores de Tiempo
7.
Blood ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38949981

RESUMEN

-Thalassemia (AT) is one of the most commonly occurring inherited hematological diseases. However, few treatments are available, and allogeneic bone marrow transplantation (BMT) is the only available therapeutic option for patients with severe AT. Research into AT has remained limited due to a lack of adult mouse models, with severe AT typically resulting in in utero lethality. By using a lipid nanoparticle (LNP) targeting the receptor CD117 and delivering a Cre mRNA (mRNACreLNPCD117), we were able to delete floxed -globin genes at high efficiency in hematopoietic stem cells (HSC) ex vivo. These cells were then engrafted in the absence or presence of a novel α-globin expressing lentiviral vector (ALS20I). Myeloablated mice transplanted with mRNACreLNPCD117-treated HSC showed a complete knockout of -globin genes. They demonstrated a phenotype characterized by the synthesis of hemoglobin H (-tetramers,  or HbH), aberrant erythropoiesis, and abnormal organ morphology, culminating in lethality approximately eight weeks following engraftment. Mice receiving mRNACreLNPCD117-treated HSC with at least one copy of ALS20I survived long-term with normalization of erythropoiesis, decreased the production of HbH, and ameliorated the abnormal organ morphology. Furthermore, we tested ALS20I in erythroid progenitors derived from -globin-KO CD34+ and cells isolated from patients with both deletional and non-deletional HbH disease, demonstrating improvement in -globin/-globin mRNA ratio and reduction in the formation of HbH by HPLC. Our results demonstrate the broad applicability of LNP for disease modeling, characterization of a novel severe mouse model of AT, and the efficacy of ALS20I for treating AT.

8.
Nature ; 581(7809): 470-474, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32461640

RESUMEN

The gut of healthy human neonates is usually devoid of viruses at birth, but quickly becomes colonized, which-in some cases-leads to gastrointestinal disorders1-4. Here we show that the assembly of the viral community in neonates takes place in distinct steps. Fluorescent staining of virus-like particles purified from infant meconium or early stool samples shows few or no particles, but by one month of life particle numbers increase to 109 per gram, and these numbers seem to persist throughout life5-7. We investigated the origin of these viral populations using shotgun metagenomic sequencing of virus-enriched preparations and whole microbial communities, followed by targeted microbiological analyses. Results indicate that, early after birth, pioneer bacteria colonize the infant gut and by one month prophages induced from these bacteria provide the predominant population of virus-like particles. By four months of life, identifiable viruses that replicate in human cells become more prominent. Multiple human viruses were more abundant in stool samples from babies who were exclusively fed on formula milk compared with those fed partially or fully on breast milk, paralleling reports that breast milk can be protective against viral infections8-10. Bacteriophage populations also differed depending on whether or not the infant was breastfed. We show that the colonization of the infant gut is stepwise, first mainly by temperate bacteriophages induced from pioneer bacteria, and later by viruses that replicate in human cells; this second phase is modulated by breastfeeding.


Asunto(s)
Lactancia Materna , Tracto Gastrointestinal/virología , Virus/aislamiento & purificación , Adulto , Bacteriólisis , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Heces/virología , Femenino , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Humanos , Lactante , Recién Nacido , Lisogenia , Masculino , Meconio/virología , Profagos/genética , Profagos/aislamiento & purificación , Virus/genética
9.
PLoS Pathog ; 19(3): e1011097, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36867659

RESUMEN

HIV integrase (IN) inserts viral DNA into the host genome and is the target of the strand transfer inhibitors (STIs), a class of small molecules currently in clinical use. Another potent class of antivirals is the allosteric inhibitors of integrase, or ALLINIs. ALLINIs promote IN aggregation by stabilizing an interaction between the catalytic core domain (CCD) and carboxy-terminal domain (CTD) that undermines viral particle formation in late replication. Ongoing challenges with inhibitor potency, toxicity, and viral resistance motivate research to understand their mechanism. Here, we report a 2.93 Å X-ray crystal structure of the minimal ternary complex between CCD, CTD, and the ALLINI BI-224436. This structure reveals an asymmetric ternary complex with a prominent network of π-mediated interactions that suggest specific avenues for future ALLINI development and optimization.


Asunto(s)
Inhibidores de Integrasa VIH , Integrasa de VIH , VIH-1 , VIH-1/metabolismo , Regulación Alostérica , Inhibidores de Integrasa VIH/farmacología , Antivirales , Dominio Catalítico , Integrasa de VIH/genética
10.
Blood ; 141(22): 2698-2712, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36745870

RESUMEN

Cell therapies that rely on engineered immune cells can be enhanced by achieving uniform and controlled transgene expression in order to maximize T-cell function and achieve predictable patient responses. Although they are effective, current genetic engineering strategies that use γ-retroviral, lentiviral, and transposon-based vectors to integrate transgenes, unavoidably produce variegated transgene expression in addition to posing a risk of insertional mutagenesis. In the setting of chimeric antigen receptor (CAR) therapy, inconsistent and random CAR expression may result in tonic signaling, T-cell exhaustion, and variable T-cell persistence. Here, we report and validate an algorithm for the identification of extragenic genomic safe harbors (GSH) that can be efficiently targeted for DNA integration and can support sustained and predictable CAR expression in human peripheral blood T cells. The algorithm is based on 7 criteria established to minimize genotoxicity by directing transgene integration away from functionally important genomic elements, maximize efficient CRISPR/Cas9-mediated targeting, and avert transgene silencing over time. T cells engineered to express a CD19 CAR at GSH6, which meets all 7 criteria, are curative at low cell dose in a mouse model of acute lymphoblastic leukemia, matching the potency of CAR T cells engineered at the TRAC locus and effectively resisting tumor rechallenge 100 days after their infusion. The identification of functional extragenic GSHs thus expands the human genome available for therapeutic precision engineering.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Linfocitos T , Animales , Ratones , Humanos , Vectores Genéticos , Inmunoterapia Adoptiva , Ingeniería Celular , Genómica , Antígenos CD19
11.
Blood ; 142(15): 1281-1296, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37478401

RESUMEN

Wiskott-Aldrich syndrome (WAS) is a rare X-linked disorder characterized by combined immunodeficiency, eczema, microthrombocytopenia, autoimmunity, and lymphoid malignancies. Gene therapy (GT) to modify autologous CD34+ cells is an emerging alternative treatment with advantages over standard allogeneic hematopoietic stem cell transplantation for patients who lack well-matched donors, avoiding graft-versus-host-disease. We report the outcomes of a phase 1/2 clinical trial in which 5 patients with severe WAS underwent GT using a self-inactivating lentiviral vector expressing the human WAS complementary DNA under the control of a 1.6-kB fragment of the autologous promoter after busulfan and fludarabine conditioning. All patients were alive and well with sustained multilineage vector gene marking (median follow-up: 7.6 years). Clinical improvement of eczema, infections, and bleeding diathesis was universal. Immune function was consistently improved despite subphysiologic levels of transgenic WAS protein expression. Improvements in platelet count and cytoskeletal function in myeloid cells were most prominent in patients with high vector copy number in the transduced product. Two patients with a history of autoimmunity had flares of autoimmunity after GT, despite similar percentages of WAS protein-expressing cells and gene marking to those without autoimmunity. Patients with flares of autoimmunity demonstrated poor numerical recovery of T cells and regulatory T cells (Tregs), interleukin-10-producing regulatory B cells (Bregs), and transitional B cells. Thus, recovery of the Breg compartment, along with Tregs appears to be protective against development of autoimmunity after GT. These results indicate that clinical and laboratory manifestations of WAS are improved with GT with an acceptable safety profile. This trial is registered at clinicaltrials.gov as #NCT01410825.


Asunto(s)
Eccema , Trasplante de Células Madre Hematopoyéticas , Síndrome de Wiskott-Aldrich , Humanos , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/terapia , Proteína del Síndrome de Wiskott-Aldrich/genética , Células Madre Hematopoyéticas/metabolismo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Terapia Genética/métodos , Eccema/etiología , Eccema/metabolismo , Eccema/terapia
12.
Nature ; 566(7742): 120-125, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30700913

RESUMEN

A stable latent reservoir for HIV-1 in resting CD4+ T cells is the principal barrier to a cure1-3. Curative strategies that target the reservoir are being tested4,5 and require accurate, scalable reservoir assays. The reservoir was defined with quantitative viral outgrowth assays for cells that release infectious virus after one round of T cell activation1. However, these quantitative outgrowth assays and newer assays for cells that produce viral RNA after activation6 may underestimate the reservoir size because one round of activation does not induce all proviruses7. Many studies rely on simple assays based on polymerase chain reaction to detect proviral DNA regardless of transcriptional status, but the clinical relevance of these assays is unclear, as the vast majority of proviruses are defective7-9. Here we describe a more accurate method of measuring the HIV-1 reservoir that separately quantifies intact and defective proviruses. We show that the dynamics of cells that carry intact and defective proviruses are different in vitro and in vivo. These findings have implications for targeting the intact proviruses that are a barrier to curing HIV infection.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Portador Sano/virología , Virus Defectuosos/aislamiento & purificación , Infecciones por VIH/virología , VIH-1/aislamiento & purificación , Provirus/aislamiento & purificación , Latencia del Virus , Linfocitos T CD4-Positivos/citología , Portador Sano/terapia , Línea Celular , ADN Viral/análisis , ADN Viral/genética , Virus Defectuosos/genética , Virus Defectuosos/fisiología , Infecciones por VIH/terapia , VIH-1/genética , VIH-1/fisiología , Humanos , Activación de Linfocitos , Reacción en Cadena de la Polimerasa , Provirus/genética , Provirus/fisiología
13.
Nature ; 558(7709): 307-312, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29849141

RESUMEN

Cancer immunotherapy based on genetically redirecting T cells has been used successfully to treat B cell malignancies1-3. In this strategy, the T cell genome is modified by integration of viral vectors or transposons encoding chimaeric antigen receptors (CARs) that direct tumour cell killing. However, this approach is often limited by the extent of expansion and persistence of CAR T cells4,5. Here we report mechanistic insights from studies of a patient with chronic lymphocytic leukaemia treated with CAR T cells targeting the CD19 protein. Following infusion of CAR T cells, anti-tumour activity was evident in the peripheral blood, lymph nodes and bone marrow; this activity was accompanied by complete remission. Unexpectedly, at the peak of the response, 94% of CAR T cells originated from a single clone in which lentiviral vector-mediated insertion of the CAR transgene disrupted the methylcytosine dioxygenase TET2 gene. Further analysis revealed a hypomorphic mutation in this patient's second TET2 allele. TET2-disrupted CAR T cells exhibited an epigenetic profile consistent with altered T cell differentiation and, at the peak of expansion, displayed a central memory phenotype. Experimental knockdown of TET2 recapitulated the potency-enhancing effect of TET2 dysfunction in this patient's CAR T cells. These findings suggest that the progeny of a single CAR T cell induced leukaemia remission and that TET2 modification may be useful for improving immunotherapies.


Asunto(s)
5-Metilcitosina/metabolismo , Antígenos CD19/inmunología , Dioxigenasas/genética , Inmunoterapia/métodos , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante , Traslado Adoptivo , Anciano , Alelos , Diferenciación Celular , Ensayos Clínicos como Asunto , Células Clonales/citología , Células Clonales/inmunología , Dioxigenasas/metabolismo , Epigénesis Genética , Células HEK293 , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Mutación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Transgenes
14.
J Infect Dis ; 228(4): 479-486, 2023 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-37217829

RESUMEN

BACKGROUND: Face masks have been critical in the coronavirus disease 2019 (COVID-19) pandemic, but supplies were sometimes limited and disposable masks contribute greatly to environmental waste. Studies suggest that filtration capacity is retained with repeated use, and surveys indicate many people reuse surgical masks. However, the impact of mask reuse on the host is understudied. METHODS: We applied 16S rRNA gene sequencing to investigate the bacterial microbiome of the facial skin and oropharynx of individuals randomized to wearing fresh surgical masks daily versus masks reused for 1 week. RESULTS: Compared to daily fresh masks, reuse was associated with increased richness (number of taxa) of the skin microbiome and trend towards greater diversity, but no difference in the oropharyngeal microbiome. Used masks had either skin-dominant or oropharynx-dominant bacterial sequences, and reused masks had >100-fold higher bacterial content but no change in composition compared to those used for 1 day. CONCLUSIONS: One week of mask reuse increased the number of low-abundance taxa on the face but did not impact the upper respiratory microbiome. Thus, face mask reuse has little impact on the host microbiome, although whether minor changes to the skin microbiome might relate to reported skin sequelae of masking (maskne) remains to be determined.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , ARN Ribosómico 16S/genética , Máscaras , Pandemias
15.
Blood ; 138(15): 1304-1316, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33974038

RESUMEN

Patients lacking functional adenosine deaminase activity have severe combined immunodeficiency (ADA SCID), which can be treated with ADA enzyme replacement therapy (ERT), allogeneic hematopoietic stem cell transplantation (HSCT), or autologous HSCT with gene-corrected cells (gene therapy [GT]). A cohort of 10 ADA SCID patients, aged 3 months to 15 years, underwent GT in a phase 2 clinical trial between 2009 and 2012. Autologous bone marrow CD34+ cells were transduced ex vivo with the MND (myeloproliferative sarcoma virus, negative control region deleted, dl587rev primer binding site)-ADA gammaretroviral vector (gRV) and infused following busulfan reduced-intensity conditioning. These patients were monitored in a long-term follow-up protocol over 8 to 11 years. Nine of 10 patients have sufficient immune reconstitution to protect against serious infections and have not needed to resume ERT or proceed to secondary allogeneic HSCT. ERT was restarted 6 months after GT in the oldest patient who had no evidence of benefit from GT. Four of 9 evaluable patients with the highest gene marking and B-cell numbers remain off immunoglobulin replacement therapy and responded to vaccines. There were broad ranges of responses in normalization of ADA enzyme activity and adenine metabolites in blood cells and levels of cellular and humoral immune reconstitution. Outcomes were generally better in younger patients and those receiving higher doses of gene-marked CD34+ cells. No patient experienced a leukoproliferative event after GT, despite persisting prominent clones with vector integrations adjacent to proto-oncogenes. These long-term findings demonstrate enduring efficacy of GT for ADA SCID but also highlight risks of genotoxicity with gRVs. This trial was registered at www.clinicaltrials.gov as #NCT00794508.


Asunto(s)
Agammaglobulinemia/terapia , Terapia Genética , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/genética , Adolescente , Agammaglobulinemia/genética , Niño , Preescolar , Estudios de Seguimiento , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Lactante , Inmunodeficiencia Combinada Grave/genética , Trasplante Autólogo/métodos , Resultado del Tratamiento
16.
Cell ; 135(1): 49-60, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18854154

RESUMEN

Human Immunodeficiency Viruses (HIV-1 and HIV-2) rely upon host-encoded proteins to facilitate their replication. Here, we combined genome-wide siRNA analyses with interrogation of human interactome databases to assemble a host-pathogen biochemical network containing 213 confirmed host cellular factors and 11 HIV-1-encoded proteins. Protein complexes that regulate ubiquitin conjugation, proteolysis, DNA-damage response, and RNA splicing were identified as important modulators of early-stage HIV-1 infection. Additionally, over 40 new factors were shown to specifically influence the initiation and/or kinetics of HIV-1 DNA synthesis, including cytoskeletal regulatory proteins, modulators of posttranslational modification, and nucleic acid-binding proteins. Finally, 15 proteins with diverse functional roles, including nuclear transport, prostaglandin synthesis, ubiquitination, and transcription, were found to influence nuclear import or viral DNA integration. Taken together, the multiscale approach described here has uncovered multiprotein virus-host interactions that likely act in concert to facilitate the early steps of HIV-1 infection.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Interacciones Huésped-Patógeno , Proteínas/metabolismo , Replicación Viral , Línea Celular , Humanos , Interferencia de ARN , Técnicas del Sistema de Dos Híbridos
18.
Mol Ther ; 30(8): 2646-2663, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35690906

RESUMEN

On August 18, 2021, the American Society of Gene and Cell Therapy (ASGCT) hosted a virtual roundtable on adeno-associated virus (AAV) integration, featuring leading experts in preclinical and clinical AAV gene therapy, to further contextualize and understand this phenomenon. Recombinant AAV (rAAV) vectors are used to develop therapies for many conditions given their ability to transduce multiple cell types, resulting in long-term expression of transgenes. Although most rAAV DNA typically remains episomal, some rAAV DNA becomes integrated into genomic DNA at a low frequency, and rAAV insertional mutagenesis has been shown to lead to tumorigenesis in neonatal mice. Currently, the risk of rAAV-mediated oncogenesis in humans is theoretical because no confirmed genotoxic events have been reported to date. However, because insertional mutagenesis has been reported in a small number of murine studies, there is a need to characterize this genotoxicity to inform research, regulatory needs, and patient care. The purpose of this white paper is to review the evidence of rAAV-related host genome integration in animal models and possible risks of insertional mutagenesis in patients. In addition, technical considerations, regulatory guidance, and bioethics are discussed.


Asunto(s)
Dependovirus , Vectores Genéticos , Animales , Dependovirus/genética , Vectores Genéticos/genética , Humanos , Ratones , Mutagénesis Insercional , Plásmidos , Transgenes , Integración Viral
19.
Am J Respir Crit Care Med ; 206(12): 1508-1521, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36103583

RESUMEN

Rationale: Primary graft dysfunction (PGD) is the principal cause of early morbidity and mortality after lung transplantation. The lung microbiome has been implicated in later transplantation outcomes but has not been investigated in PGD. Objectives: To define the peritransplant bacterial lung microbiome and relationship to host response and PGD. Methods: This was a single-center prospective cohort study. Airway lavage samples from donor lungs before organ procurement and recipient allografts immediately after implantation underwent bacterial 16S ribosomal ribonucleic acid gene sequencing. Recipient allograft samples were analyzed for cytokines by multiplex array and pepsin by ELISA. Measurements and Main Results: We enrolled 139 transplant subjects and obtained donor lung (n = 109) and recipient allograft (n = 136) samples. Severe PGD (persistent grade 3) developed in 15 subjects over the first 72 hours, and 40 remained without PGD (persistent grade 0). The microbiome of donor lungs differed from healthy lungs, and recipient allograft microbiomes differed from donor lungs. Development of severe PGD was associated with enrichment in the immediate postimplantation lung of oropharyngeal anaerobic taxa, particularly Prevotella. Elevated pepsin, a gastric biomarker, and a hyperinflammatory cytokine profile were present in recipient allografts in severe PGD and strongly correlated with microbiome composition. Together, immediate postimplantation allograft Prevotella/Streptococcus ratio, pepsin, and indicator cytokines were associated with development of severe PGD during the 72-hour post-transplantation period (area under the curve = 0.81). Conclusions: Lung allografts that develop PGD have a microbiome enriched in anaerobic oropharyngeal taxa, elevated gastric pepsin, and hyperinflammatory phenotype. These findings suggest a possible role for peritransplant aspiration in PGD, a potentially actionable mechanism that warrants further investigation.


Asunto(s)
Trasplante de Pulmón , Microbiota , Disfunción Primaria del Injerto , Humanos , Disfunción Primaria del Injerto/etiología , Pepsina A , Estudios Prospectivos , Trasplante de Pulmón/efectos adversos , Citocinas , Pulmón , Inflamación/complicaciones , Aloinjertos
20.
J Infect Dis ; 226(3): 463-473, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35134186

RESUMEN

Some risk factors for severe coronavirus disease 2019 (COVID-19) have been identified, including age, race, and obesity. However, 20%-50% of severe cases occur in the absence of these factors. Cytomegalovirus (CMV) is a herpesvirus that infects about 50% of all individuals worldwide and is among the most significant nongenetic determinants of immune system. We hypothesized that latent CMV infection might influence the severity of COVID-19. Our analyses demonstrate that CMV seropositivity is associated with more than twice the risk of hospitalization due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Immune profiling of blood and CMV DNA quantitative polymerase chain reaction in a subset of patients for whom respiratory tract samples were available revealed altered T-cell activation profiles in absence of extensive CMV replication in the upper respiratory tract. These data suggest a potential role for CMV-driven immune perturbations in affecting the outcome of SARS-CoV-2 infection and may have implications for the discrepancies in COVID-19 severity between different human populations.


Asunto(s)
COVID-19 , Infecciones por Citomegalovirus , Infección Latente , Citomegalovirus , Hospitalización , Humanos , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA