Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Anim Sci ; 1012023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37226633

RESUMEN

A total of 2,184 pigs (337 × 1,050, PIC; initially 12.4 ± 0.17 kg) were used in a 143-d study to evaluate the effects of feeding varying analyzed calcium to phosphorus ratios (Ca:P) at two standardized total tract digestible (STTD) phosphorus to net energy ratios (STTD P:NE). Pens of pigs (26 pigs per pen) were assigned to 1 of the 6 dietary treatments in a 2 × 3 factorial with main effects of STTD P:NE and Ca:P ratio. Diets consisted of two levels of STTD P:NE; High (1.80, 1.62, 1.43, 1.25, 1.10, and 0.99 g STTD P/Mcal NE from 11 to 22, 22 to 40, 40 to 58, 58 to 81, 81 to 104, and 104 to 129 kg, respectively); or Low (75% of the High levels), and three analyzed Ca:P ratios (0.90:1, 1.30:1, and 1.75:1). There were 14 pens per treatment. Diets were corn-soybean meal-based and contained a constant phytase concentration within each dietary phase with levels decreasing throughout the trial (phases 1 through 3, 500 FTU/kg, assumed release of 0.13% STTD P; phase 4, 400 FTU/kg, assumed release of 0.11% STTD P; phase 5, 290 FTU/kg, assumed release of 0.09% STTD P; and phase 6, 210 FTU/kg, assumed release of 0.07% STTD P). Overall, there was a Ca:P × STTD P:NE interaction (P < 0.05) observed for average daily gain (ADG), feed efficiency (G:F), final body weight (BW), hot carcass weight (HCW), bone mineral density, bone mineral content, and bone-breaking strength. When feeding Low STTD P:NE levels, increasing the analyzed Ca:P ratio decreased (linear, P < 0.001) ADG final BW, HCW, and tended to worsen G:F, bone mineral density, and bone mineral content (linear, P < 0.10). However, when feeding High STTD P:NE levels, increasing the analyzed Ca:P ratio significantly improved bone mineral content and bone mineral density (linear, P < 0.05), and tended to improve ADG and final BW (linear, P < 0.10) and G:F (quadratic P < 0.10). Additionally, increasing the analyzed Ca:P ratio worsened ADG, G:F, and bone mineralization with Low STTD P:NE but had marginal impacts when adequate STTD P:NE was fed.


Calcium (Ca) and phosphorus (P) are the most abundant minerals in the pig and are involved in lean tissue deposition and synthesis and maintenance of the skeletal structure. Swine diets are typically formulated with low margins of safety for P and excess P in the diet can lead to increased P excretion, which can result in negative environmental effects. To have an adequate utilization of both Ca and P, it is important to consider the Ca:P ratio when formulating pig diets. Research has shown that a wide Ca:P is detrimental to pig growth performance and bone mineralization when diets are low in STTD P. Therefore, the objective of this study was to evaluate the impact of varying Ca:P ratios fed at two levels of STTD P:NE on growth performance, bone, and carcass characteristics of pigs from 12 to 129 kg. When P levels were below requirement estimates, widening the Ca:P ratio from 0.90:1 to 1.75:1 reduced growth performance and bone mineralization; however, widening the Ca:P ratio improved performance and bone mineralization when P levels of the diet were above requirement estimates.


Asunto(s)
Dieta , Fósforo Dietético , Animales , 6-Fitasa/farmacología , Calcio/farmacología , Calcio de la Dieta/farmacología , Dieta/veterinaria , Fósforo Dietético/farmacología , Porcinos , Aumento de Peso
2.
Proc Natl Acad Sci U S A ; 120(12): e2215914120, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36917674

RESUMEN

How bacterial strains within a complex human microbiota collectively shape intestinal T cell homeostasis is not well understood. Methods that quickly identify effector strains or species that drive specific mucosal T cell phenotypes are needed to define general principles for how the microbiota modulates host immunity. We colonize germ-free mice with defined communities of cultured strains and profile antigen-specific responses directed toward individual strains ex vivo. We find that lamina propria T cells are specific to bacterial strains at the species level and can discriminate between strains of the same species. Ex vivo restimulations consistently identify the strains within complex communities that induce Th17 responses in vivo, providing the potential to shape baseline immune tone via community composition. Using an adoptive transfer model of colitis, we find that lamina propria T cells respond to different bacterial strains in conditions of inflammation versus homeostasis. Collectively, our approach represents a unique method for efficiently predicting the relative impact of individual bacterial strains within a complex community and for parsing microbiota-dependent phenotypes into component fractions.


Asunto(s)
Intestinos , Microbiota , Humanos , Animales , Ratones , Intestinos/microbiología , Membrana Mucosa , Bacterias , Linfocitos T CD4-Positivos , Fenotipo , Mucosa Intestinal
3.
Cell Host Microbe ; 30(10): 1481-1498.e5, 2022 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-36099923

RESUMEN

The potential of commensal bacteria to modulate host immunity remains largely uncharacterized, largely due to the vast number of strains that comprise the human gut microbiota. We have developed a screening platform to measure the innate immune responses of myeloid cells to 277 bacterial strains isolated from the gut microbiota of healthy individuals and those with inflammatory bowel diseases. The innate immune responses to gut-derived bacteria are as strong as those toward pathogenic bacteria, and they vary from phylum to strain. Myeloid cells differentially rely upon innate receptors TLR2 or TLR4 to sense taxa, with differential sensing of Bacteroidetes and Proteobacteria that predict in vivo functions. These innate immune responses can be modeled using combinations of up to 8 Toll-like receptor (TLR) agonists. Furthermore, the immunogenicity of strains is stable over time and following fecal microbiota transplantation into new human recipients. Collectively, this high-throughput approach provides an insight into how commensal microorganisms shape innate immune phenotypes.


Asunto(s)
Microbioma Gastrointestinal , Bacterias , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiología , Humanos , Inmunidad Innata , Receptor Toll-Like 2 , Receptor Toll-Like 4
4.
Pathogens ; 11(7)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35890050

RESUMEN

Conventionally, hyperimmune globulin drugs manufactured from pooled immunoglobulins from vaccinated or convalescent donors have been used in treating infections where no treatment is available. This is especially important where multi-epitope neutralization is required to prevent the development of immune-evading viral mutants that can emerge upon treatment with monoclonal antibodies. Using microfluidics, flow sorting, and a targeted integration cell line, a first-in-class recombinant hyperimmune globulin therapeutic against SARS-CoV-2 (GIGA-2050) was generated. Using processes similar to conventional monoclonal antibody manufacturing, GIGA-2050, comprising 12,500 antibodies, was scaled-up for clinical manufacturing and multiple development/tox lots were assessed for consistency. Antibody sequence diversity, cell growth, productivity, and product quality were assessed across different manufacturing sites and production scales. GIGA-2050 was purified and tested for good laboratory procedures (GLP) toxicology, pharmacokinetics, and in vivo efficacy against natural SARS-CoV-2 infection in mice. The GIGA-2050 master cell bank was highly stable, producing material at consistent yield and product quality up to >70 generations. Good manufacturing practices (GMP) and development batches of GIGA-2050 showed consistent product quality, impurity clearance, potency, and protection in an in vivo efficacy model. Nonhuman primate toxicology and pharmacokinetics studies suggest that GIGA-2050 is safe and has a half-life similar to other recombinant human IgG1 antibodies. These results supported a successful investigational new drug application for GIGA-2050. This study demonstrates that a new class of drugs, recombinant hyperimmune globulins, can be manufactured consistently at the clinical scale and presents a new approach to treating infectious diseases that targets multiple epitopes of a virus.

5.
Sci Immunol ; 7(73): eabg3208, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35857580

RESUMEN

Despite being the most abundantly secreted immunoglobulin isotype, the pattern of reactivity of immunoglobulin A (IgA) antibodies toward each individual's own gut commensal bacteria still remains elusive. By colonizing germ-free mice with defined commensal bacteria, we found that the binding specificity of bulk fecal and serum IgA toward resident gut bacteria resolves well at the species level and has modest strain-level specificity. IgA hybridomas generated from lamina propria B cells of gnotobiotic mice showed that most IgA clones recognized a single bacterial species, whereas a small portion displayed cross-reactivity. Orally administered hybridoma-produced IgAs still retained bacterial antigen binding capability, implying the potential for a new class of therapeutic antibodies. Species-specific IgAs had a range of strain specificities. Given the distinctive bacterial species and strain composition found in each individual's gut, our findings suggest the IgA antibody repertoire is shaped uniquely to bind "self" gut bacteria.


Asunto(s)
Microbioma Gastrointestinal , Animales , Linfocitos B , Células Clonales , Hibridomas , Inmunoglobulina A , Ratones
6.
Nucleic Acids Res ; 50(12): e68, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35325179

RESUMEN

The study and manipulation of T cell receptors (TCRs) is central to multiple fields across basic and translational immunology research. Produced by V(D)J recombination, TCRs are often only recorded in the literature and data repositories as a combination of their V and J gene symbols, plus their hypervariable CDR3 amino acid sequence. However, numerous applications require full-length coding nucleotide sequences. Here we present Stitchr, a software tool developed to specifically address this limitation. Given minimal V/J/CDR3 information, Stitchr produces complete coding sequences representing a fully spliced TCR cDNA. Due to its modular design, Stitchr can be used for TCR engineering using either published germline or novel/modified variable and constant region sequences. Sequences produced by Stitchr were validated by synthesizing and transducing TCR sequences into Jurkat cells, recapitulating the expected antigen specificity of the parental TCR. Using a companion script, Thimble, we demonstrate that Stitchr can process a million TCRs in under ten minutes using a standard desktop personal computer. By systematizing the production and modification of TCR sequences, we propose that Stitchr will increase the speed, repeatability, and reproducibility of TCR research. Stitchr is available on GitHub.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Programas Informáticos , Secuencia de Aminoácidos , Secuencia de Bases , ADN Complementario , Humanos , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/genética , Reproducibilidad de los Resultados
8.
Nat Commun ; 12(1): 6324, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732726

RESUMEN

Mutations in the cardiac splicing factor RBM20 lead to malignant dilated cardiomyopathy (DCM). To understand the mechanism of RBM20-associated DCM, we engineered isogenic iPSCs with DCM-associated missense mutations in RBM20 as well as RBM20 knockout (KO) iPSCs. iPSC-derived engineered heart tissues made from these cell lines recapitulate contractile dysfunction of RBM20-associated DCM and reveal greater dysfunction with missense mutations than KO. Analysis of RBM20 RNA binding by eCLIP reveals a gain-of-function preference of mutant RBM20 for 3' UTR sequences that are shared with amyotrophic lateral sclerosis (ALS) and processing-body associated RNA binding proteins (FUS, DDX6). Deep RNA sequencing reveals that the RBM20 R636S mutant has unique gene, splicing, polyadenylation and circular RNA defects that differ from RBM20 KO. Super-resolution microscopy verifies that mutant RBM20 maintains very limited nuclear localization potential; rather, the mutant protein associates with cytoplasmic processing bodies (DDX6) under basal conditions, and with stress granules (G3BP1) following acute stress. Taken together, our results highlight a pathogenic mechanism in cardiac disease through splicing-dependent and -independent pathways.


Asunto(s)
Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Mutación con Ganancia de Función , Mutación , Empalme del ARN , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/metabolismo , Cardiomiopatía Dilatada/genética , ARN Helicasas DEAD-box , ADN Helicasas , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Mutación Missense , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Proto-Oncogénicas , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo
9.
Sci Rep ; 11(1): 13308, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172783

RESUMEN

Gastrointestinal symptoms are common in COVID-19 patients but the nature of the gut immune response to SARS-CoV-2 remains poorly characterized, partly due to the difficulty of obtaining biopsy specimens from infected individuals. In lieu of tissue samples, we measured cytokines, inflammatory markers, viral RNA, microbiome composition, and antibody responses in stool samples from a cohort of 44 hospitalized COVID-19 patients. SARS-CoV-2 RNA was detected in stool of 41% of patients and more frequently in patients with diarrhea. Patients who survived had lower fecal viral RNA than those who died. Strains isolated from stool and nasopharynx of an individual were the same. Compared to uninfected controls, COVID-19 patients had higher fecal levels of IL-8 and lower levels of fecal IL-10. Stool IL-23 was higher in patients with more severe COVID-19 disease, and we found evidence of intestinal virus-specific IgA responses associated with more severe disease. We provide evidence for an ongoing humeral immune response to SARS-CoV-2 in the gastrointestinal tract, but little evidence of overt inflammation.


Asunto(s)
COVID-19 , Heces , Microbioma Gastrointestinal , Nasofaringe/virología , ARN Viral/aislamiento & purificación , Anciano , Biomarcadores/metabolismo , COVID-19/epidemiología , COVID-19/inmunología , Estudios de Cohortes , Citocinas/metabolismo , Heces/virología , Femenino , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Masculino , Persona de Mediana Edad , Ciudad de Nueva York/epidemiología , SARS-CoV-2/aislamiento & purificación
10.
Nat Biotechnol ; 39(8): 989-999, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33859400

RESUMEN

Plasma-derived polyclonal antibody therapeutics, such as intravenous immunoglobulin, have multiple drawbacks, including low potency, impurities, insufficient supply and batch-to-batch variation. Here we describe a microfluidics and molecular genomics strategy for capturing diverse mammalian antibody repertoires to create recombinant multivalent hyperimmune globulins. Our method generates of diverse mixtures of thousands of recombinant antibodies, enriched for specificity and activity against therapeutic targets. Each hyperimmune globulin product comprised thousands to tens of thousands of antibodies derived from convalescent or vaccinated human donors or from immunized mice. Using this approach, we generated hyperimmune globulins with potent neutralizing activity against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in under 3 months, Fc-engineered hyperimmune globulins specific for Zika virus that lacked antibody-dependent enhancement of disease, and hyperimmune globulins specific for lung pathogens present in patients with primary immune deficiency. To address the limitations of rabbit-derived anti-thymocyte globulin, we generated a recombinant human version and demonstrated its efficacy in mice against graft-versus-host disease.


Asunto(s)
Linfocitos B/inmunología , COVID-19/terapia , Globulinas/biosíntesis , SARS-CoV-2/inmunología , Animales , Anticuerpos Antivirales/inmunología , Células CHO , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Globulinas/inmunología , Humanos , Inmunización Pasiva , Ratones , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Virus Zika/inmunología , Sueroterapia para COVID-19
11.
Gastroenterology ; 160(7): 2435-2450.e34, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33676971

RESUMEN

BACKGROUND & AIMS: Given that gastrointestinal (GI) symptoms are a prominent extrapulmonary manifestation of COVID-19, we investigated intestinal infection with SARS-CoV-2, its effect on pathogenesis, and clinical significance. METHODS: Human intestinal biopsy tissues were obtained from patients with COVID-19 (n = 19) and uninfected control individuals (n = 10) for microscopic examination, cytometry by time of flight analyses, and RNA sequencing. Additionally, disease severity and mortality were examined in patients with and without GI symptoms in 2 large, independent cohorts of hospitalized patients in the United States (N = 634) and Europe (N = 287) using multivariate logistic regressions. RESULTS: COVID-19 case patients and control individuals in the biopsy cohort were comparable for age, sex, rates of hospitalization, and relevant comorbid conditions. SARS-CoV-2 was detected in small intestinal epithelial cells by immunofluorescence staining or electron microscopy in 15 of 17 patients studied. High-dimensional analyses of GI tissues showed low levels of inflammation, including down-regulation of key inflammatory genes including IFNG, CXCL8, CXCL2, and IL1B and reduced frequencies of proinflammatory dendritic cells compared with control individuals. Consistent with these findings, we found a significant reduction in disease severity and mortality in patients presenting with GI symptoms that was independent of sex, age, and comorbid illnesses and despite similar nasopharyngeal SARS-CoV-2 viral loads. Furthermore, there was reduced levels of key inflammatory proteins in circulation in patients with GI symptoms. CONCLUSIONS: These data highlight the absence of a proinflammatory response in the GI tract despite detection of SARS-CoV-2. In parallel, reduced mortality in patients with COVID-19 presenting with GI symptoms was observed. A potential role of the GI tract in attenuating SARS-CoV-2-associated inflammation needs to be further examined.


Asunto(s)
COVID-19/virología , Enfermedades Gastrointestinales/virología , Inmunidad Mucosa , Mucosa Intestinal/virología , SARS-CoV-2/patogenicidad , Anciano , Anciano de 80 o más Años , COVID-19/diagnóstico , COVID-19/inmunología , COVID-19/mortalidad , Estudios de Casos y Controles , Células Cultivadas , Citocinas/sangre , Femenino , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/inmunología , Enfermedades Gastrointestinales/mortalidad , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/sangre , Mucosa Intestinal/inmunología , Italia , Masculino , Persona de Mediana Edad , Ciudad de Nueva York , Pronóstico , Medición de Riesgo , Factores de Riesgo , SARS-CoV-2/inmunología , Carga Viral
12.
medRxiv ; 2020 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-32909002

RESUMEN

We sought to characterize the role of the gastrointestinal immune system in the pathogenesis of the inflammatory response associated with COVID-19. We measured cytokines, inflammatory markers, viral RNA, microbiome composition and antibody responses in stool from a cohort of 44 hospitalized COVID-19 patients. SARS-CoV-2 RNA was detected in stool of 41% of patients and more frequently in patients with diarrhea. Patients who survived had lower fecal viral RNA than those who died. Strains isolated from stool and nasopharynx of an individual were the same. Compared to uninfected controls, COVID-19 patients had higher fecal levels of IL-8 and lower levels of fecal IL-10. Stool IL-23 was higher in patients with more severe COVID-19 disease, and we found evidence of intestinal virus-specific IgA responses associated with more severe disease. We provide evidence for an ongoing humeral immune response to SARS-CoV-2 in the gastrointestinal tract, but little evidence of overt inflammation.

13.
medRxiv ; 2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-32935117

RESUMEN

Given that gastrointestinal (GI) symptoms are a prominent extrapulmonary manifestation of coronavirus disease 2019 (COVID-19), we investigated intestinal infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its effect on disease pathogenesis. SARS-CoV-2 was detected in small intestinal enterocytes by immunofluorescence staining or electron microscopy, in 13 of 15 patients studied. High dimensional analyses of GI tissues revealed low levels of inflammation in general, including active downregulation of key inflammatory genes such as IFNG, CXCL8, CXCL2 and IL1B and reduced frequencies of proinflammatory dendritic cell subsets. To evaluate the clinical significance of these findings, examination of two large, independent cohorts of hospitalized patients in the United States and Europe revealed a significant reduction in disease severity and mortality that was independent of gender, age, and examined co-morbid illnesses. The observed mortality reduction in COVID-19 patients with GI symptoms was associated with reduced levels of key inflammatory proteins including IL-6, CXCL8, IL-17A and CCL28 in circulation but was not associated with significant differences in nasopharyngeal viral loads. These data draw attention to organ-level heterogeneity in disease pathogenesis and highlight the role of the GI tract in attenuating SARS-CoV-2-associated inflammation with related mortality benefit. ONE SENTENCE SUMMARY: Intestinal infection with SARS-CoV-2 is associated with a mild inflammatory response and improved clinical outcomes.

14.
Proc Natl Acad Sci U S A ; 117(35): 21536-21545, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817490

RESUMEN

The building evidence for the contribution of microbiota to human disease has spurred an effort to develop therapies that target the gut microbiota. This is particularly evident in inflammatory bowel diseases (IBDs), where clinical trials of fecal microbiota transplantation have shown some efficacy. To aid the development of novel microbiota-targeted therapies and to better understand the biology underpinning such treatments, we have used gnotobiotic mice to model microbiota manipulations in the context of microbiotas from humans with inflammatory bowel disease. Mice colonized with IBD donor-derived microbiotas exhibit a stereotypical set of phenotypes, characterized by abundant mucosal Th17 cells, a deficit in the tolerogenic RORγt+ regulatory T (Treg) cell subset, and susceptibility to disease in colitis models. Transplanting healthy donor-derived microbiotas into mice colonized with human IBD microbiotas led to induction of RORγt+ Treg cells, which was associated with an increase in the density of the microbiotas following transplant. Microbiota transplant reduced gut Th17 cells in mice colonized with a microbiota from a donor with Crohn's disease. By culturing strains from this microbiota and screening them in vivo, we identified a specific strain that potently induces Th17 cells. Microbiota transplants reduced the relative abundance of this strain in the gut microbiota, which was correlated with a reduction in Th17 cells and protection from colitis.


Asunto(s)
Trasplante de Microbiota Fecal , Enfermedades Inflamatorias del Intestino/microbiología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Colitis/prevención & control , Colon/microbiología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/microbiología , Citocinas/inmunología , Modelos Animales de Enfermedad , Heces/microbiología , Femenino , Microbioma Gastrointestinal/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores/microbiología , Células Th17/microbiología
15.
Immunity ; 52(6): 910-941, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32505227

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected millions of people worldwide, igniting an unprecedented effort from the scientific community to understand the biological underpinning of COVID19 pathophysiology. In this Review, we summarize the current state of knowledge of innate and adaptive immune responses elicited by SARS-CoV-2 infection and the immunological pathways that likely contribute to disease severity and death. We also discuss the rationale and clinical outcome of current therapeutic strategies as well as prospective clinical trials to prevent or treat SARS-CoV-2 infection.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Animales , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/terapia , Susceptibilidad a Enfermedades , Humanos , Inmunidad Innata , Memoria Inmunológica , Inflamación/inmunología , Inflamación/virología , Linfocitos/inmunología , Células Mieloides/inmunología , Pandemias , Neumonía Viral/diagnóstico , Neumonía Viral/patología , Neumonía Viral/terapia , SARS-CoV-2
16.
Nat Biotechnol ; 38(5): 609-619, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32393905

RESUMEN

T cells engineered to express antigen-specific T cell receptors (TCRs) are potent therapies for viral infections and cancer. However, efficient identification of clinical candidate TCRs is complicated by the size and complexity of T cell repertoires and the challenges of working with primary T cells. Here we present a high-throughput method to identify TCRs with high functional avidity from diverse human T cell repertoires. The approach used massively parallel microfluidics to generate libraries of natively paired, full-length TCRαß clones, from millions of primary T cells, which were then expressed in Jurkat cells. The TCRαß-Jurkat libraries enabled repeated screening and panning for antigen-reactive TCRs using peptide major histocompatibility complex binding and cellular activation. We captured more than 2.9 million natively paired TCRαß clonotypes from six healthy human donors and identified rare (<0.001% frequency) viral-antigen-reactive TCRs. We also mined a tumor-infiltrating lymphocyte sample from a patient with melanoma and identified several tumor-specific TCRs, which, after expression in primary T cells, led to tumor cell killing.


Asunto(s)
Antígenos/análisis , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/citología , Ingeniería Celular , Biblioteca de Genes , Humanos , Células Jurkat , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Linfocitos T/inmunología , Virus/inmunología
17.
Diabetes ; 69(8): 1692-1707, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32381645

RESUMEN

A failure in self-tolerance leads to autoimmune destruction of pancreatic ß-cells and type 1 diabetes (T1D). Low-molecular-weight dextran sulfate (DS) is a sulfated semisynthetic polysaccharide with demonstrated cytoprotective and immunomodulatory properties in vitro. However, whether DS can protect pancreatic ß-cells, reduce autoimmunity, and ameliorate T1D is unknown. In this study, we report that DS, but not dextran, protects human ß-cells against cytokine-mediated cytotoxicity in vitro. DS also protects mitochondrial function and glucose-stimulated insulin secretion and reduces chemokine expression in human islets in a proinflammatory environment. Interestingly, daily treatment with DS significantly reduces diabetes incidence in prediabetic NOD mice and, most importantly, reverses diabetes in early-onset diabetic NOD mice. DS decreases ß-cell death, enhances islet heparan sulfate (HS)/HS proteoglycan expression, and preserves ß-cell mass and plasma insulin in these mice. DS administration also increases the expression of the inhibitory costimulatory molecule programmed death-1 (PD-1) in T cells, reduces interferon-γ+CD4+ and CD8+ T cells, and enhances the number of FoxP3+ cells. Collectively, these studies demonstrate that the action of one single molecule, DS, on ß-cell protection, extracellular matrix preservation, and immunomodulation can reverse diabetes in NOD mice, highlighting its therapeutic potential for the treatment of T1D.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Sulfato de Dextran/uso terapéutico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Animales , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Quimiocinas/metabolismo , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Humanos , Inmunohistoquímica , Células Secretoras de Insulina/metabolismo , Ratones , Óxidos de Nitrógeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/metabolismo
18.
Antibodies (Basel) ; 8(1)2019 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-31544823

RESUMEN

To discover therapeutically relevant antibody candidates, many groups use mouse immunization followed by hybridoma generation or B cell screening. One modern approach is to screen B cells by generating natively paired single chain variable fragment (scFv) display libraries in yeast. Such methods typically rely on soluble antigens for scFv library screening. However, many therapeutically relevant cell-surface targets are difficult to express in a soluble protein format, complicating discovery. In this study, we developed methods to screen humanized mouse-derived yeast scFv libraries using recombinant OX40 protein in cell lysate. We used deep sequencing to compare screening with cell lysate to screening with soluble OX40 protein, in the context of mouse immunizations using either soluble OX40 or OX40-expressing cells and OX40-encoding DNA vector. We found that all tested methods produce a unique diversity of scFv binders. However, when we reformatted forty-one of these scFv as full-length monoclonal antibodies (mAbs), we observed that mAbs identified using soluble antigen immunization with cell lysate sorting always bound cell surface OX40, whereas other methods had significant false positive rates. Antibodies identified using soluble antigen immunization and cell lysate sorting were also significantly more likely to activate OX40 in a cellular assay. Our data suggest that sorting with OX40 protein in cell lysate is more likely than other methods to retain the epitopes required for antibody-mediated OX40 agonism.

19.
MAbs ; 11(5): 870-883, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30898066

RESUMEN

Immunization of mice followed by hybridoma or B-cell screening is one of the most common antibody discovery methods used to generate therapeutic monoclonal antibody (mAb) candidates. There are a multitude of different immunization protocols that can generate an immune response in animals. However, an extensive analysis of the antibody repertoires that these alternative immunization protocols can generate has not been performed. In this study, we immunized mice that transgenically express human antibodies with either programmed cell death 1 protein or cytotoxic T-lymphocyte associated protein 4 using four different immunization protocols, and then utilized a single cell microfluidic platform to generate tissue-specific, natively paired immunoglobulin (Ig) repertoires from each method and enriched for target-specific binders using yeast single-chain variable fragment (scFv) display. We deep sequenced the scFv repertoires from both the pre-sort and post-sort libraries. All methods and both targets yielded similar oligoclonality, variable (V) and joining (J) gene usage, and divergence from germline of enriched libraries. However, there were differences between targets and/or immunization protocols for overall clonal counts, complementarity-determining region 3 (CDR3) length, and antibody/CDR3 sequence diversity. Our data suggest that, although different immunization protocols may generate a response to an antigen, performing multiple immunization protocols in parallel can yield greater Ig diversity. We conclude that modern microfluidic methods, followed by an extensive molecular genomic analysis of antibody repertoires, can be used to quickly analyze new immunization protocols or mouse platforms.


Asunto(s)
Anticuerpos Monoclonales Humanizados/genética , Diversidad de Anticuerpos , Inmunización/métodos , Microfluídica/métodos , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Linfocitos B/inmunología , Antígeno CTLA-4/inmunología , Regiones Determinantes de Complementariedad/genética , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hibridomas , Ratones , Ratones Transgénicos , Biblioteca de Péptidos , Receptor de Muerte Celular Programada 1/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología
20.
MAbs ; 10(3): 431-443, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29376776

RESUMEN

Deep sequencing and single-chain variable fragment (scFv) yeast display methods are becoming more popular for discovery of therapeutic antibody candidates in mouse B cell repertoires. In this study, we compare a deep sequencing and scFv display method that retains native heavy and light chain pairing with a related method that randomly pairs heavy and light chain. We performed the studies in a humanized mouse, using interleukin 21 receptor (IL-21R) as a test immunogen. We identified 44 high-affinity binder scFv with the native pairing method and 100 high-affinity binder scFv with the random pairing method. 30% of the natively paired scFv binders were also discovered with the randomly paired method, and 13% of the randomly paired binders were also discovered with the natively paired method. Additionally, 33% of the scFv binders discovered only in the randomly paired library were initially present in the natively paired pre-sort library. Thus, a significant proportion of "randomly paired" scFv were actually natively paired. We synthesized and produced 46 of the candidates as full-length antibodies and subjected them to a panel of binding assays to characterize their therapeutic potential. 87% of the antibodies were verified as binding IL-21R by at least one assay. We found that antibodies with native light chains were more likely to bind IL-21R than antibodies with non-native light chains, suggesting a higher false positive rate for antibodies from the randomly paired library. Additionally, the randomly paired method failed to identify nearly half of the true natively paired binders, suggesting a higher false negative rate. We conclude that natively paired libraries have critical advantages in sensitivity and specificity for antibody discovery programs.


Asunto(s)
Linfocitos B/inmunología , Biblioteca de Genes , Cadenas Ligeras de Inmunoglobulina , Subunidad alfa del Receptor de Interleucina-21 , Anticuerpos de Cadena Única , Animales , Humanos , Cadenas Ligeras de Inmunoglobulina/biosíntesis , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/inmunología , Subunidad alfa del Receptor de Interleucina-21/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-21/inmunología , Ratones , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...