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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(1): e2217883120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36574685

RESUMEN

Antibody heavy chain (HC) and light chain (LC) variable region exons are assembled by V(D)J recombination. V(D)J junctional regions encode complementarity-determining-region 3 (CDR3), an antigen-contact region immensely diversified through nontemplated nucleotide additions ("N-regions") by terminal deoxynucleotidyl transferase (TdT). HIV-1 vaccine strategies seek to elicit human HIV-1 broadly neutralizing antibodies (bnAbs), such as the potent CD4-binding site VRC01-class bnAbs. Mice with primary B cells that express receptors (BCRs) representing bnAb precursors are used as vaccination models. VRC01-class bnAbs uniformly use human HC VH1-2 and commonly use human LCs Vκ3-20 or Vκ1-33 associated with an exceptionally short 5-amino-acid (5-aa) CDR3. Prior VRC01-class models had nonphysiological precursor levels and/or limited precursor diversity. Here, we describe VRC01-class rearranging mice that generate more physiological primary VRC01-class BCR repertoires via rearrangement of VH1-2, as well as Vκ1-33 and/or Vκ3-20 in association with diverse CDR3s. Human-like TdT expression in mouse precursor B cells increased LC CDR3 length and diversity and also promoted the generation of shorter LC CDR3s via N-region suppression of dominant microhomology-mediated Vκ-to-Jκ joins. Priming immunization with eOD-GT8 60mer, which strongly engages VRC01 precursors, induced robust VRC01-class germinal center B cell responses. Vκ3-20-based responses were enhanced by N-region addition, which generates Vκ3-20-to-Jκ junctional sequence combinations that encode VRC01-class 5-aa CDR3s with a critical E residue. VRC01-class-rearranging models should facilitate further evaluation of VRC01-class prime and boost immunogens. These new VRC01-class mouse models establish a prototype for the generation of vaccine-testing mouse models for other HIV-1 bnAb lineages that employ different HC or LC Vs.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH-1 , Vacunas , Ratones , Humanos , Animales , Anticuerpos ampliamente neutralizantes , Anticuerpos Neutralizantes , VIH-1/genética , Anticuerpos Anti-VIH , ADN Nucleotidilexotransferasa , Regiones Determinantes de Complementariedad/genética , Infecciones por VIH/prevención & control
2.
Blood ; 142(12): 1082-1098, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37363865

RESUMEN

Antibodies against fetal red blood cell (RBC) antigens can cause hemolytic disease of the fetus and newborn (HDFN). Reductions in HDFN due to anti-RhD antibodies have been achieved through use of Rh immune globulin (RhIg), a polyclonal antibody preparation that causes antibody-mediated immunosuppression (AMIS), thereby preventing maternal immune responses against fetal RBCs. Despite the success of RhIg, it is only effective against 1 alloantigen. The lack of similar interventions that mitigate immune responses toward other RBC alloantigens reflects an incomplete understanding of AMIS mechanisms. AMIS has been previously attributed to rapid antibody-mediated RBC removal, resulting in B-cell ignorance of the RBC alloantigen. However, our data demonstrate that antibody-mediated RBC removal can enhance de novo alloimmunization. In contrast, inclusion of antibodies that possess the ability to rapidly remove the target antigen in the absence of detectable RBC clearance can convert an augmented antibody response to AMIS. These results suggest that the ability of antibodies to remove target antigens from the RBC surface can trigger AMIS in situations in which enhanced immunity may otherwise occur. In doing so, these results hold promise in identifying key antibody characteristics that can drive AMIS, thereby facilitating the design of AMIS approaches toward other RBC antigens to eliminate all forms of HDFN.


Asunto(s)
Eritroblastosis Fetal , Eritrocitos , Femenino , Recién Nacido , Humanos , Eritrocitos/metabolismo , Anticuerpos , Tolerancia Inmunológica , Terapia de Inmunosupresión , Globulina Inmune rho(D) , Isoantígenos , Isoanticuerpos
3.
N Engl J Med ; 384(3): 205-215, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33283990

RESUMEN

BACKGROUND: Sickle cell disease is characterized by hemolytic anemia, pain, and progressive organ damage. A high level of erythrocyte fetal hemoglobin (HbF) comprising α- and γ-globins may ameliorate these manifestations by mitigating sickle hemoglobin polymerization and erythrocyte sickling. BCL11A is a repressor of γ-globin expression and HbF production in adult erythrocytes. Its down-regulation is a promising therapeutic strategy for induction of HbF. METHODS: We enrolled patients with sickle cell disease in a single-center, open-label pilot study. The investigational therapy involved infusion of autologous CD34+ cells transduced with the BCH-BB694 lentiviral vector, which encodes a short hairpin RNA (shRNA) targeting BCL11A mRNA embedded in a microRNA (shmiR), allowing erythroid lineage-specific knockdown. Patients were assessed for primary end points of engraftment and safety and for hematologic and clinical responses to treatment. RESULTS: As of October 2020, six patients had been followed for at least 6 months after receiving BCH-BB694 gene therapy; median follow-up was 18 months (range, 7 to 29). All patients had engraftment, and adverse events were consistent with effects of the preparative chemotherapy. All the patients who could be fully evaluated achieved robust and stable HbF induction (percentage HbF/(F+S) at most recent follow-up, 20.4 to 41.3%), with HbF broadly distributed in red cells (F-cells 58.9 to 93.6% of untransfused red cells) and HbF per F-cell of 9.0 to 18.6 pg per cell. Clinical manifestations of sickle cell disease were reduced or absent during the follow-up period. CONCLUSIONS: This study validates BCL11A inhibition as an effective target for HbF induction and provides preliminary evidence that shmiR-based gene knockdown offers a favorable risk-benefit profile in sickle cell disease. (Funded by the National Institutes of Health; ClinicalTrials.gov number, NCT03282656).


Asunto(s)
Anemia de Células Falciformes/terapia , Hemoglobina Fetal/biosíntesis , Terapia Genética , Interferencia de ARN , Proteínas Represoras/genética , gamma-Globinas/metabolismo , Adolescente , Adulto , Anemia de Células Falciformes/genética , Niño , Regulación hacia Abajo , Femenino , Hemoglobina Fetal/genética , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Humanos , Masculino , Proyectos Piloto , ARN Interferente Pequeño , Proteínas Represoras/metabolismo , Trasplante Autólogo , Adulto Joven , gamma-Globinas/genética
4.
Mol Ther ; 30(8): 2693-2708, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35526095

RESUMEN

A promising treatment for ß-hemoglobinopathies is the de-repression of γ-globin expression leading to increased fetal hemoglobin (HbF) by targeting BCL11A. Here, we aim to improve a lentivirus vector (LV) containing a single BCL11A shmiR (SS) to further increase γ-globin induction. We engineered a novel LV to express two shmiRs simultaneously targeting BCL11A and the γ-globin repressor ZNF410. Erythroid cells derived from human HSCs transduced with the double shmiR (DS) showed up to a 70% reduction of both BCL11A and ZNF410 proteins. There was a consistent and significant additional 10% increase in HbF compared to targeting BCL11A alone in erythroid cells. Erythrocytes differentiated from SCD HSCs transduced with the DS demonstrated significantly reduced in vitro sickling phenotype compared to the SS. Erythrocytes differentiated from transduced HSCs from ß-thalassemia major patients demonstrated improved globin chain balance by increased γ-globin with reduced microcytosis. Reconstitution of DS-transduced cells from Berkeley SCD mice was associated with a statistically larger reduction in peripheral blood hemolysis markers compared with the SS vector. Overall, these results indicate that the DS LV targeting BCL11A and ZNF410 can enhance HbF induction for treating ß-hemoglobinopathies and could be used as a model to simultaneously and efficiently target multiple gene products.


Asunto(s)
Hemoglobina Fetal , Hemoglobinopatías , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , gamma-Globinas/genética
5.
Nature ; 525(7567): 134-139, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26308889

RESUMEN

During B-cell development, RAG endonuclease cleaves immunoglobulin heavy chain (IgH) V, D, and J gene segments and orchestrates their fusion as deletional events that assemble a V(D)J exon in the same transcriptional orientation as adjacent Cµ constant region exons. In mice, six additional sets of constant region exons (CHs) lie 100-200 kilobases downstream in the same transcriptional orientation as V(D)J and Cµ exons. Long repetitive switch (S) regions precede Cµ and downstream CHs. In mature B cells, class switch recombination (CSR) generates different antibody classes by replacing Cµ with a downstream CH (ref. 2). Activation-induced cytidine deaminase (AID) initiates CSR by promoting deamination lesions within Sµ and a downstream acceptor S region; these lesions are converted into DNA double-strand breaks (DSBs) by general DNA repair factors. Productive CSR must occur in a deletional orientation by joining the upstream end of an Sµ DSB to the downstream end of an acceptor S-region DSB. However, the relative frequency of deletional to inversional CSR junctions has not been measured. Thus, whether orientation-specific joining is a programmed mechanistic feature of CSR as it is for V(D)J recombination and, if so, how this is achieved is unknown. To address this question, we adapt high-throughput genome-wide translocation sequencing into a highly sensitive DSB end-joining assay and apply it to endogenous AID-initiated S-region DSBs in mouse B cells. We show that CSR is programmed to occur in a productive deletional orientation and does so via an unprecedented mechanism that involves in cis Igh organizational features in combination with frequent S-region DSBs initiated by AID. We further implicate ATM-dependent DSB-response factors in enforcing this mechanism and provide an explanation of why CSR is so reliant on the 53BP1 DSB-response factor.


Asunto(s)
Linfocitos B/metabolismo , Citidina Desaminasa/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Cambio de Clase de Inmunoglobulina/genética , Regiones Constantes de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Linfocitos B/enzimología , Linfocitos B/inmunología , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Desaminación , Ratones , Eliminación de Secuencia/genética , Proteína 1 de Unión al Supresor Tumoral P53 , Exones VDJ/genética
6.
Transfus Apher Sci ; 60(1): 103059, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33541761

RESUMEN

Emerging cellular therapies require the collection of peripheral blood hematopoietic stem cells (HSC) by apheresis for in vitro manipulation to accomplish gene addition or gene editing. These therapies require relatively large numbers of HSCs within a short time frame to generate an efficacious therapeutic product. This review focuses on the principal factors that affect collection outcomes, especially relevant to gene therapy for sickle cell disease.


Asunto(s)
Anemia de Células Falciformes/terapia , Edición Génica/métodos , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/metabolismo , Acondicionamiento Pretrasplante/métodos , Anemia de Células Falciformes/patología , Humanos
7.
8.
PLoS Genet ; 12(11): e1006410, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27832076

RESUMEN

A proportion of homologous recombination (HR) events in mammalian cells resolve by "long tract" gene conversion, reflecting copying of several kilobases from the donor sister chromatid prior to termination. Cells lacking the major hereditary breast/ovarian cancer predisposition genes, BRCA1 or BRCA2, or certain other HR-defective cells, reveal a bias in favor of long tract gene conversion, suggesting that this aberrant HR outcome might be connected with genomic instability. If termination of gene conversion occurs in regions lacking homology with the second end of the break, the normal mechanism of HR termination by annealing (i.e., homologous pairing) is not available and termination must occur by as yet poorly defined non-canonical mechanisms. Here we use a previously described HR reporter to analyze mechanisms of non-canonical termination of long tract gene conversion in mammalian cells. We find that non-canonical HR termination can occur in the absence of the classical non-homologous end joining gene XRCC4. We observe obligatory use of microhomology (MH)-mediated end joining and/or nucleotide addition during rejoining with the second end of the break. Notably, non-canonical HR termination is associated with complex breakpoints. We identify roles for homology-mediated template switching and, potentially, MH-mediated template switching/microhomology-mediated break-induced replication, in the formation of complex breakpoints at sites of non-canonical HR termination. This work identifies non-canonical HR termination as a potential contributor to genomic instability and to the formation of complex breakpoints in cancer.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Unión al ADN/genética , Recombinación Homóloga/genética , Neoplasias Ováricas/genética , Animales , Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama/patología , Cromátides/genética , Reparación del ADN por Unión de Extremidades/genética , Femenino , Conversión Génica/genética , Inestabilidad Genómica/genética , Humanos , Ratones , Células Madre Embrionarias de Ratones , Neoplasias Ováricas/patología
9.
Blood ; 127(14): 1740-1, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27056995

RESUMEN

In this issue of Blood, Arthur et al uncover that HLA alloantibodies cannot solely account for the immune mechanism in platelet refractoriness.


Asunto(s)
Plaquetas/inmunología , Linfocitos T CD8-positivos/inmunología , Transfusión de Plaquetas , Animales , Humanos
10.
Mol Cell ; 39(6): 873-85, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20864035

RESUMEN

During an immune response, B cells undergo rapid proliferation and activation-induced cytidine deaminase (AID)-dependent remodeling of immunoglobulin (IG) genes within germinal centers (GCs) to generate memory B and plasma cells. Unfortunately, the genotoxic stress associated with the GC reaction also promotes most B cell malignancies. Here, we report that exogenous and intrinsic AID-induced DNA strand breaks activate ATM, which signals through an LKB1 intermediate to inactivate CRTC2, a transcriptional coactivator of CREB. Using genome-wide location analysis, we determined that CRTC2 inactivation unexpectedly represses a genetic program that controls GC B cell proliferation, self-renewal, and differentiation while opposing lymphomagenesis. Inhibition of this pathway results in increased GC B cell proliferation, reduced antibody secretion, and impaired terminal differentiation. Multiple distinct pathway disruptions were also identified in human GC B cell lymphoma patient samples. Combined, our data show that CRTC2 inactivation, via physiologic DNA damage response signaling, promotes B cell differentiation in response to genotoxic stress.


Asunto(s)
Linfocitos B/citología , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/inmunología , Citidina Desaminasa/genética , Daño del ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de la radiación , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/efectos de la radiación , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Expresión Génica/efectos de la radiación , Regulación de la Expresión Génica/inmunología , Centro Germinal/citología , Humanos , Cambio de Clase de Inmunoglobulina/fisiología , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Metformina/farmacología , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
11.
Am J Hematol ; 97(3): 253-255, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35007360
12.
Mol Cell ; 36(1): 110-20, 2009 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-19818714

RESUMEN

Although the BBAP E3 ligase and its binding partner BAL are overexpressed in chemotherapy-resistant lymphomas, the role of these proteins in DNA damage responses remains undefined. Because BAL proteins modulate promoter-coupled transcription and contain structural motifs associated with chromatin remodeling and DNA repair, we reasoned that the BBAP E3 ligase might target nucleosomal proteins. Herein, we demonstrate that BBAP selectively monoubiquitylates histone H4 lysine 91 and protects cells exposed to DNA-damaging agents. Disruption of BBAP-mediated monoubiquitylation of histone H4K91 is associated with the loss of chromatin-associated H4K20 methylase, mono- and dimethyl H4K20, and a delay in the kinetics of 53BP1 foci formation at sites of DNA damage. Because 53BP1 localizes to DNA damage sites, in part, via an interaction with dimethyl H4K20, these data directly implicate BBAP in the monoubiquitylation and additional posttranslational modification of histone H4 and an associated DNA damage response.


Asunto(s)
Daño del ADN/fisiología , Histonas/metabolismo , Lisina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Acetilación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Doxorrubicina/farmacología , Células HeLa , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Hidroxiurea/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metilación/efectos de los fármacos , Nucleosomas/metabolismo , Unión Proteica/fisiología , Procesamiento Proteico-Postraduccional/fisiología , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/fisiología
13.
Proc Natl Acad Sci U S A ; 108(7): 2873-8, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282644

RESUMEN

We utilized gene expression profiling of a comprehensive panel of purified developmentally defined normal murine B cells to identify unique transcriptional signatures for each subset. To elucidate transcription factor activities that function in a stage-specific fashion, we used gene sets that share transcription factor targets and found that germinal center B cells had a robust enrichment of up-regulated and down-regulated signatures compared with the other B-cell subsets. Notably, we found Yy1 and its targets to be central regulators of the germinal center B (GCB)-specific transcriptional program with binding of Yy1 to select signature genes in GCB cells, and translation of the Yy1 signatures to human GCB cells. We then tested whether our newly generated, stage-specific transcriptional signatures could be used to link murine lymphoma models to stages of normal B-cell development. Although each of the molecularly defined murine lymphoma models conserved certain stage-specific features of normal B-cell development, there was a significant alteration of the normal differentiation signature following malignant transformation. These findings offer important tools and insights for elucidating differences between normal and malignant B cells.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Regulación de la Expresión Génica/inmunología , Centro Germinal/metabolismo , Linfoma/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Subgrupos de Linfocitos B/citología , Diferenciación Celular/inmunología , Inmunoprecipitación de Cromatina , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Ratones
14.
Blood Adv ; 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39293082

RESUMEN

Hematopoietic stem cell transplantation with lentiviral vector (LVV) transduced autologous cells has proven an effective therapeutic strategy for sickle cell disease (SCD). However, ex vivo culture or proliferative stress associated with in vivo reconstitution may amplify any underlying genetic risk of leukemia. We aimed to minimize culture-induced stress and reduce genomic damage during ex vivo culture, enhance stem cell fitness and reconstitution of SCD CD34+ cells transduced with BCL11A shmiR-encoding LVV currently in clinical trials (NCT NCT03282656). UM171, a pyrimidoindole derivative can expand normal hematopoietic stem cells (HSCs) during in vitro culture and has been shown to be safe and effective in clinical trials using umbilical cord blood (NCT02668315). We examined the effect of UM171 during ex vivo LVV transduction of SCD HSCs. Culture of SCD CD34+ HSCs with UM171 during transduction reduced DNA damage and reactive oxygen species (ROS), decreased apoptosis, and was associated with increased numbers of immunophenotypically defined long-term HSCs. UM171 increased the engraftment of LVV transduced human HSCs in immunodeficient mice and barcode tracing revealed increased clonal diversity of engrafting cells. In competitive transplantation assays, analysis of BM showed that cells transduced in the presence of UM171 consistently outcompeted those transduced under control conditions. In summary, exposure of SCD peripheral blood CD34+ cells to UM171 during LVV transduction enhances stem cell fitness. These findings suggest manufacturing of genetically modified HSCs in the presence of UM171 may improve efficacy, safety and sustainability of gene therapy utilizing ex vivo approaches.

15.
Nat Aging ; 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39117982

RESUMEN

Although cancer is an age-related disease, how the processes of aging contribute to cancer progression is not well understood. In this study, we uncovered how mouse B cell lymphoma develops as a consequence of a naturally aged system. We show here that this malignancy is associated with an age-associated clonal B cell (ACBC) population that likely originates from age-associated B cells. Driven by c-Myc activation, promoter hypermethylation and somatic mutations, IgM+ ACBCs clonally expand independently of germinal centers and show increased biological age. ACBCs become self-sufficient and support malignancy when transferred into young recipients. Inhibition of mTOR or c-Myc in old mice attenuates pre-malignant changes in B cells during aging. Although the etiology of mouse and human B cell lymphomas is considered distinct, epigenetic changes in transformed mouse B cells are enriched for changes observed in human B cell lymphomas. Together, our findings characterize the spontaneous progression of cancer during aging through both cell-intrinsic and microenvironmental changes and suggest interventions for its prevention.

16.
Nature ; 449(7161): 478-82, 2007 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-17713479

RESUMEN

Immunoglobulin variable region exons are assembled in developing B cells by V(D)J recombination. Once mature, these cells undergo class-switch recombination (CSR) when activated by antigen. CSR changes the heavy chain constant region exons (Ch) expressed with a given variable region exon from Cmu to a downstream Ch (for example, Cgamma, Cepsilon or Calpha), thereby switching expression from IgM to IgG, IgE or IgA. Both V(D)J recombination and CSR involve the introduction of DNA double-strand breaks and their repair by means of end joining. For CSR, double-strand breaks are introduced into switch regions that flank Cmu and a downstream Ch, followed by fusion of the broken switch regions. In mammalian cells, the 'classical' non-homologous end joining (C-NHEJ) pathway repairs both general DNA double-strand breaks and programmed double-strand breaks generated by V(D)J recombination. C-NHEJ, as observed during V(D)J recombination, joins ends that lack homology to form 'direct' joins, and also joins ends with several base-pair homologies to form microhomology joins. CSR joins also display direct and microhomology joins, and CSR has been suggested to use C-NHEJ. Xrcc4 and DNA ligase IV (Lig4), which cooperatively catalyse the ligation step of C-NHEJ, are the most specific C-NHEJ factors; they are absolutely required for V(D)J recombination and have no known functions other than C-NHEJ. Here we assess whether C-NHEJ is also critical for CSR by assaying CSR in Xrcc4- or Lig4-deficient mouse B cells. C-NHEJ indeed catalyses CSR joins, because C-NHEJ-deficient B cells had decreased CSR and substantial levels of IgH locus (immunoglobulin heavy chain, encoded by Igh) chromosomal breaks. However, an alternative end-joining pathway, which is markedly biased towards microhomology joins, supports CSR at unexpectedly robust levels in C-NHEJ-deficient B cells. In the absence of C-NHEJ, this alternative end-joining pathway also frequently joins Igh locus breaks to other chromosomes to generate translocations.


Asunto(s)
Genes de las Cadenas Pesadas de las Inmunoglobulinas/genética , Cambio de Clase de Inmunoglobulina/genética , Recombinación Genética/genética , Translocación Genética/genética , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Linfocitos B/efectos de la radiación , Secuencia de Bases , Proliferación Celular , Células Cultivadas , Rotura Cromosómica , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Hibridación Fluorescente in Situ , Ratones , Radiación Ionizante , Telómero/genética
17.
Proc Natl Acad Sci U S A ; 107(7): 3040-5, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20133637

RESUMEN

Ig heavy chain (IgH) class-switch recombination (CSR) replaces the IgH C mu constant region exons with one of several sets of downstream IgH constant region exons (e.g., C gamma, C epsilon, or C alpha), which affects switching from IgM to another IgH class (e.g., IgG, IgE, or IgA). Activation-induced cytidine deaminase (AID) initiates CSR by promoting DNA double-strand breaks (DSBs) within switch (S) regions flanking the donor C mu (S mu) and a downstream acceptor C(H) (e.g., S gamma, S epsilon, S alpha) that are then joined to complete CSR. DSBs generated in S mu frequently are joined within S mu to form internal switch region deletions (ISD). AID-induced ISD and mutations have been considered rare in downstream S regions, suggesting that AID targeting to these S regions requires its prior recruitment to S mu. We have now assayed for CSR and ISD in B cells lacking S mu (S mu(-/-) B cells). In S mu(-/-) B cells activated for CSR to IgG1 and IgE, CSR to IgG1 was greatly reduced; but, surprisingly, CSR to IgE occurred at nearly normal levels. Moreover, normal B cells had substantial S gamma1 ISD and increased mutations in and near S gamma1, and levels of both were greatly increased in S mu(-/-) B cells. Finally, S mu(-/-) B cells underwent downstream CSR between S gamma1 and S epsilon on alleles that lacked S mu CSR to these sequences. Our findings show that AID targets downstream S regions independently of CSR with Smu and implicate an alternative pathway for IgE class switching that involves generation and joining of DSBs within two different downstream S regions before S mu joining.


Asunto(s)
Linfocitos B/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina E/biosíntesis , Cadenas Pesadas de Inmunoglobulina/genética , Animales , Southern Blotting , Citidina Desaminasa/metabolismo , Análisis Mutacional de ADN , Cartilla de ADN/genética , Citometría de Flujo , Hibridomas/inmunología , Hibridomas/metabolismo , Inmunoglobulina E/inmunología , Cadenas Pesadas de Inmunoglobulina/inmunología , Inmunoglobulina M/genética , Región de Cambio de la Inmunoglobulina/genética , Ratones , Ratones Noqueados
18.
bioRxiv ; 2023 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-37292647

RESUMEN

Gene editing the BCL11A erythroid enhancer is a validated approach to fetal hemoglobin (HbF) induction for ß-hemoglobinopathy therapy, though heterogeneity in edit allele distribution and HbF response may impact its safety and efficacy. Here we compared combined CRISPR-Cas9 endonuclease editing of the BCL11A +58 and +55 enhancers with leading gene modification approaches under clinical investigation. We found that combined targeting of the BCL11A +58 and +55 enhancers with 3xNLS-SpCas9 and two sgRNAs resulted in superior HbF induction, including in engrafting erythroid cells from sickle cell disease (SCD) patient xenografts, attributable to simultaneous disruption of core half E-box/GATA motifs at both enhancers. We corroborated prior observations that double strand breaks (DSBs) could produce unintended on- target outcomes in hematopoietic stem and progenitor cells (HSPCs) such as long deletions and centromere-distal chromosome fragment loss. We show these unintended outcomes are a byproduct of cellular proliferation stimulated by ex vivo culture. Editing HSPCs without cytokine culture bypassed long deletion and micronuclei formation while preserving efficient on-target editing and engraftment function. These results indicate that nuclease editing of quiescent hematopoietic stem cells (HSCs) limits DSB genotoxicity while maintaining therapeutic potency and encourages efforts for in vivo delivery of nucleases to HSCs.

20.
Cancer Cell ; 6(1): 85-9, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15261144

RESUMEN

The recent discoveries of p63 and p73, homologs of the tumor suppressor p53, raised the possibility of a network of these family members governing cell cycle arrest and apoptosis in response to stress. However, mice lacking p73 show no tendency for spontaneous tumors, and mutations in p63 or p73 are rare in human tumors, rendering any obligate role of these genes in cell death and tumor suppression unclear. In an effort to reconcile these incongruent data, we examined the genetic interactions between p53, p63, and p73 in well-established paradigms of p53-dependent and -independent T cell death using primary, genetically defined lymphocytes. Our findings challenge the generality of the notion that p63 and p73 are required for p53 function or for apoptosis in T cells.


Asunto(s)
Apoptosis , Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Linfocitos T/metabolismo , Transactivadores/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Apoptosis/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Heterocigoto , Homocigoto , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Nucleares/genética , Fosfoproteínas/genética , Linfocitos T/citología , Timo/citología , Timo/metabolismo , Transactivadores/genética , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA