Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 40(11): e107333, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33950524

RESUMEN

To examine global changes in breast heterogeneity across different states, we determined the single-cell transcriptomes of > 340,000 cells encompassing normal breast, preneoplastic BRCA1+/- tissue, the major breast cancer subtypes, and pairs of tumors and involved lymph nodes. Elucidation of the normal breast microenvironment revealed striking changes in the stroma of post-menopausal women. Single-cell profiling of 34 treatment-naive primary tumors, including estrogen receptor (ER)+ , HER2+ , and triple-negative breast cancers, revealed comparable diversity among cancer cells and a discrete subset of cycling cells. The transcriptomes of preneoplastic BRCA1+/- tissue versus tumors highlighted global changes in the immune microenvironment. Within the tumor immune landscape, proliferative CD8+ T cells characterized triple-negative and HER2+ cancers but not ER+ tumors, while all subtypes comprised cycling tumor-associated macrophages, thus invoking potentially different immunotherapy targets. Copy number analysis of paired ER+ tumors and lymph nodes indicated seeding by genetically distinct clones or mass migration of primary tumor cells into axillary lymph nodes. This large-scale integration of patient samples provides a high-resolution map of cell diversity in normal and cancerous human breast.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Glándulas Mamarias Humanas/metabolismo , Análisis de la Célula Individual , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Linfocitos T CD8-positivos/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/patología , RNA-Seq , Microambiente Tumoral
2.
Genomics ; 116(2): 110793, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38220132

RESUMEN

Single-cell RNA sequencing (scRNA-Seq) has emerged as a powerful tool for understanding cellular heterogeneity and function. However the choice of sample multiplexing reagents can impact data quality and experimental outcomes. In this study, we compared various multiplexing reagents, including MULTI-Seq, Hashtag antibody, and CellPlex, across diverse sample types such as human peripheral blood mononuclear cells (PBMCs), mouse embryonic brain and patient-derived xenografts (PDXs). We found that all multiplexing reagents worked well in cell types robust to ex vivo manipulation but suffered from signal-to-noise issues in more delicate sample types. We compared multiple demultiplexing algorithms which differed in performance depending on data quality. We find that minor improvements to laboratory workflows such as titration and rapid processing are critical to optimal performance. We also compared the performance of fixed scRNA-Seq kits and highlight the advantages of the Parse Biosciences kit for fragile samples. Highly multiplexed scRNA-Seq experiments require more sequencing resources, therefore we evaluated CRISPR-based destruction of non-informative genes to enhance sequencing value. Our comprehensive analysis provides insights into the selection of appropriate sample multiplexing reagents and protocols for scRNA-Seq experiments, facilitating more accurate and cost-effective studies.


Asunto(s)
Leucocitos Mononucleares , Análisis de la Célula Individual , Humanos , Animales , Ratones , RNA-Seq , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Algoritmos , Perfilación de la Expresión Génica/métodos
3.
Clin Exp Rheumatol ; 38 Suppl 126(4): 271-282, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33025890

RESUMEN

Primary Sjögren's syndrome (pSS) is often considered a B cell-mediated disease, yet the precise role of B cells in the pathogenesis is not fully understood. This is exemplified by the failure of multiple clinical trials directed at B cell depletion or inhibition. To date, most prognostic markers for severe disease outcomes are autoantibodies, but the underlying mechanisms by which B cells drive diverse disease presentations in pSS likely extend beyond autoantibody production. Here we outline an expanded role of B cells in disease pathogenesis drawing on examples from animal models of SS, and from other autoimmune diseases that share similar clinical or immunological abnormalities. We focus on recent findings from the detailed analysis of pathogenic B cells in patients with pSS to propose strategies for patient stratification to improve clinical trial outcomes. We conclude that an integrated cellular, molecular and genetic analysis of patients with pSS will reveal the underlying pathogenic mechanisms and guide precision medicine.


Asunto(s)
Enfermedades Autoinmunes , Síndrome de Sjögren , Animales , Autoanticuerpos , Linfocitos B , Humanos , Síndrome de Sjögren/diagnóstico
4.
J Clin Immunol ; 39(3): 324-335, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30927119

RESUMEN

Common variable immunodeficiency is the most prevalent of the primary immunodeficiency diseases, yet its pathogenesis is largely poorly understood. Of the cases that are monogenic, many arise due to pathogenic variants in NFKB1 and NFKB2. Here, we report enteroviral encephalomyelitis as the cause of a fatal neurodegenerative condition in a patient with a novel heterozygous mutation in NFKB2 (c.2543insG, p.P850Sfs36*) that disrupts non-canonical NF-κB signaling. Investigations of primary and secondary lymphoid tissue demonstrated a complete absence of B cells and germinal centers. Despite multiple negative viral PCR testing of cerebrospinal fluid during her disease progression, post-mortem analysis of cerebral tissue revealed a chronic lymphocytic meningoencephalitis, in the presence of Cocksackie A16 virus, as the cause of death. The clinical features, and progression of disease reported here, demonstrate divergent clinical and immunological phenotypes of individuals within a single family. This is the first reported case of fatal enteroviral encephalomyelitis in a patient with NF-κB2 deficiency and mandates a low threshold for early brain biopsy and the administration of increased immunoglobulin replacement in any patient with a defect in this pathway and deterioration of neurological status.


Asunto(s)
Inmunodeficiencia Variable Común/diagnóstico , Encefalomielitis/diagnóstico , Infecciones por Enterovirus/diagnóstico , Enterovirus/fisiología , Subunidad p52 de NF-kappa B/genética , Enfermedades Neurodegenerativas/diagnóstico , Eliminación de Secuencia/genética , Biopsia , Células Cultivadas , Niño , Inmunodeficiencia Variable Común/genética , Encefalomielitis/genética , Infecciones por Enterovirus/genética , Resultado Fatal , Femenino , Humanos , Enfermedades Neurodegenerativas/genética , Linaje
5.
Am J Hum Genet ; 97(3): 389-403, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26279205

RESUMEN

Common variable immunodeficiency (CVID), characterized by recurrent infections, is the most prevalent symptomatic antibody deficiency. In ∼90% of CVID-affected individuals, no genetic cause of the disease has been identified. In a Dutch-Australian CVID-affected family, we identified a NFKB1 heterozygous splice-donor-site mutation (c.730+4A>G), causing in-frame skipping of exon 8. NFKB1 encodes the transcription-factor precursor p105, which is processed to p50 (canonical NF-κB pathway). The altered protein bearing an internal deletion (p.Asp191_Lys244delinsGlu; p105ΔEx8) is degraded, but is not processed to p50ΔEx8. Altered NF-κB1 proteins were also undetectable in a German CVID-affected family with a heterozygous in-frame exon 9 skipping mutation (c.835+2T>G) and in a CVID-affected family from New Zealand with a heterozygous frameshift mutation (c.465dupA) in exon 7. Given that residual p105 and p50­translated from the non-mutated alleles­were normal, and altered p50 proteins were absent, we conclude that the CVID phenotype in these families is caused by NF-κB1 p50 haploinsufficiency.


Asunto(s)
Inmunodeficiencia Variable Común/genética , Haploinsuficiencia/genética , Subunidad p50 de NF-kappa B/genética , Australia , Secuencia de Bases , Western Blotting , Cartilla de ADN/genética , Exoma/genética , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Países Bajos , Nueva Zelanda , Análisis de Secuencia de ADN
6.
Immunol Cell Biol ; 96(2): 149-159, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29363164

RESUMEN

The discovery of cell division tracking properties of 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester (CFSE) by Lyons and Parish in 1994 led to a broad range of new methods and numerous important biological discoveries. After labeling, CFSE is attached to free amine groups and intracellular proteins in the cytoplasm and nucleus of a cell, and halves in fluorescence intensity with each round of cell division, enabling enumeration of the number of divisions a cell has undergone. A range of popular division tracking dyes were subsequently developed, including CellTrace Violet (CTV), making available the green fluorescent channel previously occupied by CFSE. More recently, CellTrace Yellow (CTY) and CellTrace Far Red (CTFR), each with unique fluorescence properties, were introduced. In a comparison, we found that the fluorescence values of both dyes were well separated from autofluorescence, and enabled a greater number of divisions to be identified than CTV, before this limit was reached. These new dyes provided clear and well-separated peaks for both murine and human B lymphocytes, and should find wide application. The range of excitation/emission spectra available for division tracking dyes now also facilitates multiplexing, that is, the labeling of cells with different combinations of dyes to give a unique fluorescence signature, allowing single cell in vitro and in vivo tracking. The combinatorial possibilities are significantly increased with these additional dyes.


Asunto(s)
División Celular , Rastreo Celular , Colorantes Fluorescentes/química , Linfocitos/citología , Animales , Linfocitos B/metabolismo , Femenino , Citometría de Flujo , Fluorescencia , Humanos , Memoria Inmunológica , Masculino , Ratones Endogámicos C57BL , Coloración y Etiquetado
7.
Hum Mol Genet ; 22(4): 769-81, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23161749

RESUMEN

Mendelian susceptibility to mycobacterial diseases (MSMD) is a rare syndrome, the known genetic etiologies of which impair the production of, or the response to interferon-gamma (IFN-γ). We report here a patient (P1) with MSMD whose cells display mildly impaired responses to IFN-γ, at levels, however, similar to those from MSMD patients with autosomal recessive (AR) partial IFN-γR2 or STAT1 deficiency. Whole-exome sequencing (WES) and Sanger sequencing revealed only one candidate variation for both MSMD-causing and IFN-γ-related genes. P1 carried a heterozygous frame-shift IFNGR2 mutation inherited from her father. We show that the mutant allele is intrinsically loss-of-function and not dominant-negative, suggesting haploinsufficiency at the IFNGR2 locus. We also show that Epstein-Barr virus transformed B lymphocyte cells from 10 heterozygous relatives of patients with AR complete IFN-γR2 deficiency respond poorly to IFN-γ, in some cases as poorly as the cells of P1. Naive CD4(+) T cells and memory IL-4-producing T cells from these individuals also responded poorly to IFN-γ, whereas monocytes and monocyte-derived macrophages (MDMs) did not. This is consistent with the lower levels of expression of IFN-γR2 in lymphoid than in myeloid cells. Overall, MSMD in this patient is probably due to autosomal dominant (AD) IFN-γR2 deficiency, resulting from haploinsufficiency, at least in lymphoid cells. The clinical penetrance of AD IFN-γR2 deficiency is incomplete, possibly due, at least partly, to the variability of cellular responses to IFN-γ in these individuals.


Asunto(s)
Haploinsuficiencia , Infecciones por Mycobacterium no Tuberculosas/genética , Receptores de Interferón/genética , Adolescente , Linfocitos B/inmunología , Linfocitos B/metabolismo , Secuencia de Bases , Estudios de Casos y Controles , Células Cultivadas , Femenino , Expresión Génica , Genes Dominantes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Interferón gamma/fisiología , Infecciones por Mycobacterium/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores de Interferón/deficiencia , Análisis de Secuencia de ADN , Eliminación de Secuencia
8.
Immunol Cell Biol ; 93(4): 364-71, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25895814

RESUMEN

Chemokines are a highly specialized group of cytokines that coordinate trafficking and homing of leucocytes between bone marrow, lymphoid organs and sites of infection or inflammation. They are also responsible for structural organization within lymphoid organs. Aberrant expression or function of these molecules, or their receptors, has been linked to protection or susceptibility to specific infectious diseases, as well as the risk of autoimmune disease and malignancy, revealing critical roles of chemokines and their receptors in human health, disease and therapeutics. In this review, we focus on human diseases that provide lessons regarding the critical role of these specialized and complex cytokines.


Asunto(s)
Quimiocinas/inmunología , Infecciones/inmunología , Inflamación/inmunología , Prurito/inmunología , Receptores de Quimiocina/metabolismo , Linfocitos T/fisiología , Animales , Movimiento Celular/genética , Quimiocinas/genética , Predisposición Genética a la Enfermedad , Humanos , Inmunidad Innata/genética , Mutación/genética , Receptores de Quimiocina/genética
9.
Blood ; 122(14): 2390-401, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23963039

RESUMEN

We report a molecular study of the two known patients with autosomal recessive, partial interferon-γ receptor (IFN-γR)2 deficiency (homozygous for mutations R114C and G227R), and three novel, unrelated children, homozygous for S124F (P1) and G141R (P2 and P3). IFN-γR2 levels on the surface of the three latter patients' cells are slightly lower than those on control cells. The patients' cells also display impaired, but not abolished, response to IFN-γ. Moreover, the R114C, S124F, G141R and G227R IFNGR2 hypomorphic alleles all encode misfolded proteins with abnormal N-glycosylation. The mutants are largely retained in the endoplasmic reticulum, although a small proportion reach and function at the cell surface. Strikingly, the IFN-γ response of the patients' cells is enhanced by chemical modifiers of N-glycosylation, as previously shown for patients with gain-of-glysosylation T168N and misfolding 382-387dup null mutations. All four in-frame IFNGR2 hypomorphic mutant alleles encoding surface-expressed receptors are thus deleterious by a mechanism involving abnormal N-glycosylation and misfolding of the IFN-γR2 protein. The diagnosis of partial IFN-γR2 deficiency is clinically useful, as affected patients should be treated with IFN-γ, [corrected] unlike patients with complete IFN-γR2 deficiency. Moreover, inhibitors of glycosylation might be beneficial in patients with complete or partial IFN-γR2 deficiency due to misfolding or gain-of-glycosylation receptors.


Asunto(s)
Deficiencias en la Proteostasis/genética , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Alcaloides/farmacología , Secuencia de Bases , Western Blotting , Niño , Inhibidores Enzimáticos/farmacología , Femenino , Citometría de Flujo , Predisposición Genética a la Enfermedad/genética , Glicosilación/efectos de los fármacos , Humanos , Masculino , Microscopía Confocal , Datos de Secuencia Molecular , Mutación , Infecciones por Mycobacterium/genética , Linaje , Transfección
10.
Vaccines (Basel) ; 12(2)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38400197

RESUMEN

Coeliac disease (CD) is associated with hyposplenism, an acquired impairment of spleen function associated with reduced IgM memory B cells and increased susceptibility to serious pneumococcal infection. Little is known about the immune implications of hyposplenism in CD or the optimal pneumococcal vaccination strategy. In this study, the immune effects of hyposplenism in CD, and the accuracy of screening approaches and protective responses induced by two different pneumococcal vaccines were examined. Active and treated CD cohorts, and healthy and surgically splenectomised controls underwent testing for the presence of Howell-Jolly bodies and pitted red cells, spleen ultrasound, and immune assessment of IgM memory B cell frequency and IgM memory B cell responses to T cell-dependent (TD) or T cell-independent (TI) stimulation. Responses following conjugate (TD) and polysaccharide (TI) pneumococcal vaccination were compared using ELISA and opsonophagocytic assays. Although hyposplenism is rare in treated CD (5.1%), functional B cell defects are common (28-61%) and are not detected by current clinical tests. Conjugate pneumococcal vaccination induced superior and sustained protection against clinically relevant serotypes. Clinical practice guidelines in CD should recommend routine pneumococcal vaccination, ideally with a conjugate vaccine, of all patients in lieu of hyposplenism screening.

11.
J Allergy Clin Immunol Glob ; 3(2): 100241, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38585448

RESUMEN

Background: Reported outcomes in patients with primary immunodeficiency (PID) infected by coronavirus disease 2019 (COVID-19) have been variable owing to a combination of viral strain heterogeneity, differences in patient populations and health systems, and local availability of vaccination and specific COVID-19 therapies. There are few reports on the experience of Australian patients with PID during the pandemic. Objectives: In this retrospective study, we describe the baseline characteristics and short-term outcomes of patients with PID who were infected by COVID-19 and known to the Royal Melbourne Hospital, a major tertiary center in Victoria, Australia. Methods: Between April 2021 and April 2022, a total of 31 of 138 patients with PID were affected by COVID-19. More than half of them had 3 vaccine doses at the time of infection (which at the time was considered being fully vaccinated) and received COVID-19-targeted treatment. Results: All of the infected patients had ambulatory disease, with no cases of morbidity or mortality. In line with the current literature, the PID subtypes described did not appear to independently predict worse outcomes. Conclusions: Some protective factors include this cohort's relatively younger average age and its high uptake of vaccination and COVID-19 therapies.

12.
Nat Commun ; 15(1): 4286, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769332

RESUMEN

The function and phenotype of γδ T cells in the context of common variable immunodeficiency (CVID) has not been explored. CVID is a primary immunodeficiency disorder characterized by impaired antibody responses resulting in increased susceptibility to infections. γδ T cells are a subset of unconventional T cells that play crucial roles in host defence against infections. In this study, we aim to determine the roles and functions of γδ T cells in CVID. We observe a higher frequency of Vδ1+ γδ T cells compared to healthy controls, particularly in older patients. We also find a higher proportion of effector-memory Vδ1+ γδ T cells and a more clonal T cell receptor (TCR) repertoire in CVID. The most significant driver of the Vδ1+ γδ T cell expansion and phenotype in CVID patients is persistent cytomegalovirus (CMV) viremia. These findings provide valuable insights into γδ T cell biology and their contribution to immune defence in CVID.


Asunto(s)
Inmunodeficiencia Variable Común , Infecciones por Citomegalovirus , Citomegalovirus , Receptores de Antígenos de Linfocitos T gamma-delta , Humanos , Inmunodeficiencia Variable Común/inmunología , Inmunodeficiencia Variable Común/virología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Masculino , Femenino , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Adulto , Citomegalovirus/inmunología , Persona de Mediana Edad , Anciano , Adulto Joven , Subgrupos de Linfocitos T/inmunología , Viremia/inmunología , Adolescente , Estudios de Casos y Controles
13.
J Exp Med ; 220(7)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37273190

RESUMEN

B cells develop from hematopoietic stem cells in the bone marrow. Once generated, they serve multiple roles in immune regulation and host defense. However, their most important function is producing antibodies (Ab) that efficiently clear invading pathogens. This is achieved by generating memory B cells that rapidly respond to subsequent Ag exposure, and plasma cells (PCs) that continually secrete Ab. These B cell subsets maintain humoral immunity and host protection against recurrent infections for extended periods of time. Thus, the generation of antigen (Ag)-specific memory cells and PCs underlies long-lived serological immunity, contributing to the success of most vaccines. Our understanding of immunity is often derived from animal models. However, analysis of individuals with monogenic defects that disrupt immune cell function are unprecedented models to link genotypes to clinical phenotypes, establish mechanisms of disease pathogenesis, and elucidate critical pathways for immune cell development and differentiation. Here, we review fundamental breakthroughs in unraveling the complexities of humoral immunity in humans that have come from the discovery of inborn errors disrupting B cell function.


Asunto(s)
Subgrupos de Linfocitos B , Linfocitos B , Animales , Humanos , Células Plasmáticas , Diferenciación Celular , Inmunidad Humoral , Anticuerpos/metabolismo
14.
J Allergy Clin Immunol Pract ; 11(6): 1646-1664, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36796510

RESUMEN

The understanding of common variable immunodeficiency disorders (CVID) is in evolution. CVID was previously a diagnosis of exclusion. New diagnostic criteria have allowed the disorder to be identified with greater precision. With the advent of next-generation sequencing (NGS), it has become apparent that an increasing number of patients with a CVID phenotype have a causative genetic variant. If a pathogenic variant is identified, these patients are removed from the overarching diagnosis of CVID and are deemed to have a CVID-like disorder. In populations where consanguinity is more prevalent, the majority of patients with severe primary hypogammaglobulinemia will have an underlying inborn error of immunity, usually an early-onset autosomal recessive disorder. In nonconsanguineous societies, pathogenic variants are identified in approximately 20% to 30% of patients. These are often autosomal dominant mutations with variable penetrance and expressivity. To add to the complexity of CVID and CVID-like disorders, some genetic variants such as those in TNFSF13B (transmembrane activator calcium modulator cyclophilin ligand interactor) predispose to, or enhance, disease severity. These variants are not causative but can have epistatic (synergistic) interactions with more deleterious mutations to worsen disease severity. This review is a description of the current understanding of genes associated with CVID and CVID-like disorders. This information will assist clinicians in interpreting NGS reports when investigating the genetic basis of disease in patients with a CVID phenotype.


Asunto(s)
Agammaglobulinemia , Inmunodeficiencia Variable Común , Humanos , Inmunodeficiencia Variable Común/diagnóstico , Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/complicaciones , Mutación/genética , Fenotipo , Agammaglobulinemia/complicaciones
15.
Clin Transl Immunology ; 12(10): e1470, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37799772

RESUMEN

Objectives: B cells drive the production of autoreactive antibody-secreting cells (ASCs) in autoimmune diseases such as Systemic Lupus Erythematosus (SLE) and Sjögren's syndrome, causing long-term organ damage. Current treatments for antibody-mediated autoimmune diseases target B cells or broadly suppress the immune system. However, pre-existing long-lived ASCs are often refractory to treatment, leaving a reservoir of autoreactive cells that continue to produce antibodies. Therefore, the development of novel treatment methods targeting ASCs is vital to improve patient outcomes. Our objective was to test whether targeting the epigenetic regulator BMI-1 could deplete ASCs in autoimmune conditions in vivo and in vitro. Methods: Use of a BMI-1 inhibitor in both mouse and human autoimmune settings was investigated. Lyn -/- mice, a model of SLE, were treated with the BMI-1 small molecule inhibitor PTC-028, before assessment of ASCs, serum antibody and immune complexes. To examine human ASC survival, a novel human fibroblast-based assay was established, and the impact of PTC-028 on ASCs derived from Sjögren's syndrome patients was evaluated. Results: BMI-1 inhibition significantly decreased splenic and bone marrow ASCs in Lyn -/- mice. The decline in ASCs was linked to aberrant cell cycle gene expression and led to a significant decrease in serum IgG3, immune complexes and anti-DNA IgG. PTC-028 was also efficacious in reducing ex vivo plasma cell survival from both Sjögren's syndrome patients and age-matched healthy donors. Conclusion: These data provide evidence that inhibiting BMI-1 can deplete ASC in a variety of contexts and thus BMI-1 is a viable therapeutic target for antibody-mediated autoimmune diseases.

16.
Blood Adv ; 7(12): 2733-2745, 2023 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-36521105

RESUMEN

Venetoclax is an effective treatment for certain blood cancers, such as chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML). However, most patients relapse while on venetoclax and further treatment options are limited. Combining venetoclax with immunotherapies is an attractive approach; however, a detailed understanding of how venetoclax treatment impacts normal immune cells in patients is lacking. In this study, we performed deep profiling of peripheral blood (PB) cells from patients with CLL and AML before and after short-term treatment with venetoclax using mass cytometry (cytometry by time of flight) and found no impact on the concentrations of key T-cell subsets or their expression of checkpoint molecules. We also analyzed PB from patients with breast cancer receiving venetoclax long-term using a single-cell multiomics approach (cellular indexing of transcriptomes and epitopes by sequencing) and functional assays. We found significant depletion of B-cell populations with low expression of MCL-1 relative to other immune cells, attended by extensive transcriptomic changes. By contrast, there was less impact on circulating T cells and natural killer (NK) cells, with no changes in their subset composition, transcriptome, or function following venetoclax treatment. Our data indicate that venetoclax has minimal impact on circulating T or NK cells, supporting the rationale of combining this BH3 mimetic drug with cancer immunotherapies for more durable antitumor responses.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Leucemia Mieloide Aguda , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Células Asesinas Naturales , Leucemia Mieloide Aguda/tratamiento farmacológico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico
17.
Front Immunol ; 13: 815193, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35242131

RESUMEN

BACKGROUND: Common Variable Immunodeficiency (CVID) is classified as a 'Predominantly Antibody Deficiency' (PAD), but there is emerging evidence of cellular immunodeficiency in a subset of patients. This evidence includes CVID patients diagnosed with cytomegalovirus (CMV) infection, a hallmark of 'combined immunodeficiency'. CMV infection also has the potential to drive immune dysregulation contributing to significant morbidity and mortality in CVID. We aim to determine the extent of cellular immune dysfunction in CVID patients, and whether this correlates with CMV infection status. METHODS: We conducted a single-center retrospective cohort study of individuals with CVID at the Royal Melbourne Hospital, and identified patients with and without CMV disease or viraemia. We then isolated T-cells from patient and healthy donor blood samples and examined T-cell proliferation and function. RESULTS: Six patients (7.6%, 6/79) had either CMV disease (pneumonitis or gastrointestinal disease), or symptomatic CMV viraemia. A high mortality rate in the cohort of patients with CVID and CMV disease was observed, with 4 deaths in the period of analysis (66.6%, 4/6). Individuals with CMV infection showed reduced T-cell division in response to T-cell receptor (TCR) stimulation when compared with CMV-negative patients. DISCUSSION: This study demonstrates the morbidity and mortality associated with CMV in CVID, and highlights the need for focused interventions for patients with CVID at risk of CMV disease.


Asunto(s)
Inmunodeficiencia Variable Común , Infecciones por Citomegalovirus , Enfermedades de Inmunodeficiencia Primaria , Citomegalovirus , Humanos , Morbilidad , Estudios Retrospectivos , Viremia/complicaciones
18.
Cell Death Differ ; 29(12): 2519-2530, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35831623

RESUMEN

High-throughput methodologies are the cornerstone of screening approaches to identify novel compounds that regulate immune cell function. To identify novel targeted therapeutics to treat immune disorders and haematological malignancies, there is a need to integrate functional cellular information with the molecular mechanisms that regulate changes in immune cell phenotype. We facilitate this goal by combining quantitative methods for dissecting complex simultaneous cell phenotypic effects with genomic analysis. This combination strategy we term Multiplexed Analysis of Cells sequencing (MAC-seq), a modified version of Digital RNA with perturbation of Genes (DRUGseq). We applied MAC-seq to screen compounds that target the epigenetic machinery of B cells and assess altered humoral immunity by measuring changes in proliferation, survival, differentiation and transcription. This approach revealed that polycomb repressive complex 2 (PRC2) inhibitors promote antibody secreting cell (ASC) differentiation in both murine and human B cells in vitro. This is further validated using T cell-dependent immunization in mice. Functional dissection of downstream effectors of PRC2 using arrayed CRISPR screening uncovered novel regulators of B cell differentiation, including Mybl1, Myof, Gas7 and Atoh8. Together, our findings demonstrate that integrated phenotype-transcriptome analyses can be effectively combined with drug screening approaches to uncover the molecular circuitry that drives lymphocyte fate decisions.


Asunto(s)
Linfocitos B , Epigénesis Genética , Animales , Humanos , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Perfilación de la Expresión Génica , Fenotipo , Complejo Represivo Polycomb 2/metabolismo
20.
Transl Lung Cancer Res ; 10(6): 2830-2841, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34295681

RESUMEN

Metastatic lung cancer represents a significant global issue where it is responsible for the most cancer diagnoses and deaths worldwide. Treatment for advanced lung cancer has undergone a series of paradigm shifts from chemotherapy to targeted molecular agents to the most recent immunotherapy strategies. The most successful of the latter involves antibodies that block inhibitory receptors on tumor infiltrating T cells, thereby enhancing T cell activity against tumor cells. However, only a subset of patients demonstrate durable responses to these drugs and treatment resistance is common. Emerging evidence suggests that a critical role exists for B cells as more than a bystander immune cell in the tumor microenvironment (TME). However, this role is likely context-specific where B cells comprise distinct subtypes with unique effector functions that may result in anti- or pro-tumor effects. As such, the balance between various B cell subtypes affects the net B cell impact upon tumor immunity. To date, the factors needed to polarize B cell function toward anti-tumor activity are unclear. Understanding B cell biology in the lung cancer setting will help redefine and refine treatment strategies to augment anti-tumor immunity. This article presents a review of the literature describing the current knowledge of the development and function of B cells, and explores their role in lung cancer and potential as an immunotherapeutic strategy and as a predictive marker for response to immune checkpoint blockade.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA