Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 9(9)2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38512434

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ataxin-1 (ATXN1) protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockin mouse (f-ATXN1146Q/2Q) with mouse Atxn1 coding exons replaced by human ATXN1 exons encoding 146 glutamines. f-ATXN1146Q/2Q mice manifested SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. Central nervous system (CNS) contributions to disease were revealed using f-ATXN1146Q/2Q;Nestin-Cre mice, which showed improved rotarod, open field, and Barnes maze performance by 6-12 weeks of age. In contrast, striatal contributions to motor deficits using f-ATXN1146Q/2Q;Rgs9-Cre mice revealed that mice lacking ATXN1146Q/2Q in striatal medium-spiny neurons showed a trending improvement in rotarod performance at 30 weeks of age. Surprisingly, a prominent role for muscle contributions to disease was revealed in f-ATXN1146Q/2Q;ACTA1-Cre mice based on their recovery from kyphosis and absence of muscle pathology. Collectively, data from the targeted conditional deletion of the expanded allele demonstrated CNS and peripheral contributions to disease and highlighted the need to consider muscle in addition to the brain for optimal SCA1 therapeutics.


Asunto(s)
Ataxina-1 , Modelos Animales de Enfermedad , Músculo Esquelético , Ataxias Espinocerebelosas , Animales , Ataxina-1/genética , Ataxina-1/metabolismo , Ratones , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología , Músculo Esquelético/patología , Músculo Esquelético/metabolismo , Humanos , Masculino , Ratones Transgénicos , Técnicas de Sustitución del Gen , Femenino , Fenotipo , Neuronas/metabolismo , Neuronas/patología
2.
J Clin Invest ; 133(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37561596

RESUMEN

Adoptive immunotherapy with Tregs is a promising approach for preventing or treating type 1 diabetes. Islet antigen-specific Tregs have more potent therapeutic effects than polyclonal cells, but their low frequency is a barrier for clinical application. To generate Tregs that recognize islet antigens, we engineered a chimeric antigen receptor (CAR) derived from a monoclonal antibody with specificity for the insulin B chain 10-23 peptide presented in the context of the IAg7 MHC class II allele present in NOD mice. Peptide specificity of the resulting InsB-g7 CAR was confirmed by tetramer staining and T cell proliferation in response to recombinant or islet-derived peptide. The InsB-g7 CAR redirected NOD Treg specificity such that insulin B 10-23-peptide stimulation enhanced suppressive function, measured via reduction of proliferation and IL-2 production by BDC2.5 T cells and CD80 and CD86 expression on dendritic cells. Cotransfer of InsB-g7 CAR Tregs prevented adoptive transfer diabetes by BDC2.5 T cells in immunodeficient NOD mice. In WT NOD mice, InsB-g7 CAR Tregs prevented spontaneous diabetes. These results show that engineering Treg specificity for islet antigens using a T cell receptor-like CAR is a promising therapeutic approach for the prevention of autoimmune diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Receptores Quiméricos de Antígenos , Ratones , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/prevención & control , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Ratones Endogámicos NOD , Insulina/metabolismo , Linfocitos T Reguladores
3.
bioRxiv ; 2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36865264

RESUMEN

Adoptive immunotherapy with Tregs is a promising approach for prevention or treatment of type 1 diabetes. Islet antigen-specific Tregs have more potent therapeutic effects than polyclonal cells, but their low frequency is a barrier for clinical application. To generate Tregs that recognize islet antigens, we engineered a chimeric antigen receptor (CAR) derived from a monoclonal antibody with specificity for the insulin B-chain 10-23 peptide presented in the context of the IA g7 MHC class II allele present in NOD mice. Peptide specificity of the resulting InsB-g7 CAR was confirmed by tetramer staining and T cell proliferation in response to recombinant or islet-derived peptide. The InsB-g7 CAR re-directed NOD Treg specificity such that insulin B 10-23-peptide stimulation enhanced suppressive function, measured via reduction of proliferation and IL-2 production by BDC2.5 T cells and CD80 and CD86 expression on dendritic cells. Co-transfer of InsB-g7 CAR Tregs prevented adoptive transfer diabetes by BDC2.5 T cells in immunodeficient NOD mice. In wild type NOD mice, InsB-g7 CAR Tregs stably expressed Foxp3 and prevented spontaneous diabetes. These results show that engineering Treg specificity for islet antigens using a T cell receptor-like CAR is a promising new therapeutic approach for the prevention of autoimmune diabetes. Brief Summary: Chimeric antigen receptor Tregs specific for an insulin B-chain peptide presented by MHC class II prevent autoimmune diabetes.

4.
bioRxiv ; 2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-36798410

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by an expanded polyglutamine tract in the widely expressed ATXN1 protein. To elucidate anatomical regions and cell types that underlie mutant ATXN1-induced disease phenotypes, we developed a floxed conditional knockout mouse model ( f-ATXN1 146Q/2Q ) having mouse Atxn1 coding exons replaced by human exons encoding 146 glutamines. F-ATXN1 146Q/2Q mice manifest SCA1-like phenotypes including motor and cognitive deficits, wasting, and decreased survival. CNS contributions to disease were revealed using ATXN1 146Q/2Q ; Nestin-Cre mice, that showed improved rotarod, open field and Barnes maze performances. Striatal contributions to motor deficits were examined using f-ATXN1 146Q/2Q ; Rgs9-Cre mice. Mice lacking striatal ATXN1 146Q/2Q had improved rotarod performance late in disease. Muscle contributions to disease were revealed in f-ATXN1 146Q/2Q ; ACTA1-Cre mice which lacked muscle pathology and kyphosis seen in f-ATXN1 146Q/2Q mice. Kyphosis was not improved in f-ATXN1 146Q/2Q ;Nestin - Cre mice. Thus, optimal SCA1 therapeutics will require targeting mutant ATXN1 toxic actions in multiple brain regions and muscle.

5.
Neuron ; 111(4): 493-507.e6, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36577403

RESUMEN

Spinocerebellar ataxia type 1 (SCA1) is a dominant trinucleotide repeat neurodegenerative disease characterized by motor dysfunction, cognitive impairment, and premature death. Degeneration of cerebellar Purkinje cells is a frequent and prominent pathological feature of SCA1. We previously showed that transport of ATXN1 to Purkinje cell nuclei is required for pathology, where mutant ATXN1 alters transcription. To examine the role of ATXN1 nuclear localization broadly in SCA1-like disease pathogenesis, CRISPR-Cas9 was used to develop a mouse with an amino acid alteration (K772T) in the nuclear localization sequence of the expanded ATXN1 protein. Characterization of these mice indicates that proper nuclear localization of mutant ATXN1 contributes to many disease-like phenotypes including motor dysfunction, cognitive deficits, and premature lethality. RNA sequencing analysis of genes with expression corrected to WT levels in Atxn1175QK772T/2Q mice indicates that transcriptomic aspects of SCA1 pathogenesis differ between the cerebellum, brainstem, cerebral cortex, hippocampus, and striatum.


Asunto(s)
Ataxina-1 , Ataxias Espinocerebelosas , Transcriptoma , Animales , Ratones , Ataxina-1/genética , Ataxina-1/metabolismo , Encéfalo/metabolismo , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Fenotipo , Transporte de Proteínas/genética , Células de Purkinje/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo
6.
Sci Immunol ; 7(78): eadd3075, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36459542

RESUMEN

Respiratory tract resident memory T cells (TRM), typically generated by local vaccination or infection, can accelerate control of pulmonary infections that evade neutralizing antibody. It is unknown whether mRNA vaccination establishes respiratory TRM. We generated a self-amplifying mRNA vaccine encoding the influenza A virus nucleoprotein that is encapsulated in modified dendron-based nanoparticles. Here, we report how routes of immunization in mice, including contralateral versus ipsilateral intramuscular boosts, or intravenous and intranasal routes, influenced influenza-specific cell-mediated and humoral immunity. Parabiotic surgeries revealed that intramuscular immunization was sufficient to establish CD8 TRM in the lung and draining lymph nodes. Contralateral, compared with ipsilateral, intramuscular boosting broadened the distribution of lymph node TRM and T follicular helper cells but slightly diminished resulting levels of serum antibody. Intranasal mRNA delivery established modest circulating CD8 and CD4 T cell memory but augmented distribution to the respiratory mucosa. Combining intramuscular immunizations with an intranasal mRNA boost achieved high levels of both circulating T cell memory and lung TRM. Thus, routes of mRNA vaccination influence humoral and cell-mediated immunity, and intramuscular prime-boosting establishes lung TRM that can be further expanded by an additional intranasal immunization.


Asunto(s)
Linfocitos T CD4-Positivos , Vacunación , Animales , Ratones , ARN Mensajero , Anticuerpos Neutralizantes , Linfocitos T CD8-positivos , Vacunas de ARNm
7.
JCI Insight ; 7(7)2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35393950

RESUMEN

We investigate how myeloid subsets differentially shape immunity to pancreatic ductal adenocarcinoma (PDA). We show that tumor antigenicity sculpts myeloid cell composition and functionality. Antigenicity promotes accumulation of type 1 dendritic cells (cDC1), which is driven by Xcr1 signaling, and overcomes macrophage-mediated suppression. The therapeutic activity of adoptive T cell therapy or programmed cell death ligand 1 blockade required cDC1s, which sustained splenic Klrg1+ cytotoxic antitumor T cells and functional intratumoral T cells. KLRG1 and cDC1 genes correlated in human tumors, and PDA patients with high intratumoral KLRG1 survived longer than patients with low intratumoral KLRG1. The immunotherapy CD40 agonist also required host cDC1s for maximal therapeutic benefit. However, CD40 agonist exhibited partial therapeutic benefit in cDC1-deficient hosts and resulted in priming of tumor-specific yet atypical CD8+ T cells with a regulatory phenotype and that failed to participate in tumor control. Monocyte/macrophage depletion using clodronate liposomes abrogated T cell priming yet enhanced the antitumor activity of CD40 agonist in cDC1-deficient hosts via engagement of innate immunity. In sum, our study supports that cDC1s are essential for sustaining effective antitumor T cells and supports differential roles for cDC1s and monocytes/macrophages in instructing T cell fate and immunotherapy response.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Antígenos CD40/metabolismo , Linfocitos T CD8-positivos , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/terapia , Células Dendríticas , Humanos , Inmunoterapia/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas
8.
Sci Rep ; 11(1): 17142, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433860

RESUMEN

The notion that T cell insulitis increases as type 1 diabetes (T1D) develops is unsurprising, however, the quantitative analysis of CD4+ and CD8+ T cells within the islet mass is complex and limited with standard approaches. Optical microscopy is an important and widely used method to evaluate immune cell infiltration into pancreatic islets of Langerhans for the study of disease progression or therapeutic efficacy in murine T1D. However, the accuracy of this approach is often limited by subjective and potentially biased qualitative assessment of immune cell subsets. In addition, attempts at quantitative measurements require significant time for manual analysis and often involve sophisticated and expensive imaging software. In this study, we developed and illustrate here a streamlined analytical strategy for the rapid, automated and unbiased investigation of islet area and immune cell infiltration within (insulitis) and around (peri-insulitis) pancreatic islets. To this end, we demonstrate swift and accurate detection of islet borders by modeling cross-sectional islet areas with convex polygons (convex hulls) surrounding islet-associated insulin-producing ß cell and glucagon-producing α cell fluorescent signals. To accomplish this, we used a macro produced with the freeware software ImageJ equipped with the Fiji Is Just ImageJ (FIJI) image processing package. Our image analysis procedure allows for direct quantification and statistical determination of islet area and infiltration in a reproducible manner, with location-specific data that more accurately reflect islet areas as insulitis proceeds throughout T1D. Using this approach, we quantified the islet area infiltrated with CD4+ and CD8+ T cells allowing statistical comparison between different age groups of non-obese diabetic (NOD) mice progressing towards T1D. We found significantly more CD4+ and CD8+ T cells infiltrating the convex hull-defined islet mass of 13-week-old non-diabetic and 17-week-old diabetic NOD mice compared to 4-week-old NOD mice. We also determined a significant and measurable loss of islet mass in mice that developed T1D. This approach will be helpful for the location-dependent quantitative calculation of islet mass and cellular infiltration during T1D pathogenesis and can be combined with other markers of inflammation or activation in future studies.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/patología , Procesamiento de Imagen Asistido por Computador/métodos , Islotes Pancreáticos/patología , Animales , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Movimiento Celular , Diabetes Mellitus Tipo 1/inmunología , Femenino , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Microscopía Fluorescente/métodos
9.
J Immunol ; 205(5): 1449-1460, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32737148

RESUMEN

Optimal ex vivo expansion protocols of tumor-specific T cells followed by adoptive cell therapy must yield T cells able to home to tumors and effectively kill them. Our previous study demonstrated ex vivo activation in the presence of IL-12-induced optimal CD8+ T cell expansion and melanoma regression; however, adverse side effects, including autoimmunity, can occur. This may be due to transfer of high-avidity self-specific T cells. In this study, we compared mouse low- and high-avidity T cells targeting the tumor Ag tyrosinase-related protein 2 (TRP2). Not surprisingly, high-avidity T cells provide superior tumor control, yet low-avidity T cells can promote tumor regression. The addition of IL-12 during in vitro expansion boosts low-avidity T cell responsiveness, tumor regression, and prevents T cell exhaustion. In this study, we demonstrate that IL-12-primed T cells are resistant to PD-1/PD-L1-mediated suppression and retain effector function. Importantly, IL-12 preconditioning prevented exhaustion as LAG-3, PD-1, and TOX were decreased while simultaneously increasing KLRG1. Using intravital imaging, we also determined that high-avidity T cells have sustained contacts with intratumoral dendritic cells and tumor targets compared with low-avidity T cells. However, with Ag overexpression, this defect is overcome, and low-avidity T cells control tumor growth. Taken together, these data illustrate that low-avidity T cells can be therapeutically beneficial if cocultured with IL-12 cytokine during in vitro expansion and highly effective in vivo if Ag is not limiting. Clinically, low-avidity T cells provide a safer alternative to high-avidity, TCR-engineered T cells, as IL-12-primed, low-avidity T cells cause less autoimmune vitiligo.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucina-12/inmunología , Activación de Linfocitos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Antígenos de Neoplasias/inmunología , Autoinmunidad/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunoterapia Adoptiva/métodos , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología
10.
J Exp Med ; 217(8)2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32568362

RESUMEN

Numerous observations indicate that resident memory T cells (TRM) undergo unusually rapid attrition within the lung. Here we demonstrate that contraction of lung CD8+ T cell responses after influenza infection is contemporized with egress of CD69+/CD103+ CD8+ T cells to the draining mediastinal LN via the lymphatic vessels, which we term retrograde migration. Cells within the draining LN retained canonical markers of lung TRM, including CD103 and CD69, lacked Ly6C expression (also a feature of lung TRM), maintained granzyme B expression, and did not equilibrate among immunized parabiotic mice. Investigations of bystander infection or removal of the TCR from established memory cells revealed that the induction of the TRM phenotype was dependent on antigen recognition; however, maintenance was independent. Thus, local lung infection induces CD8+ T cells with a TRM phenotype that nevertheless undergo retrograde migration, yet remain durably committed to the residency program within the draining LN, where they provide longer-lived regional memory while chronicling previous upstream antigen experiences.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Memoria Inmunológica , Virus de la Influenza A/inmunología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/patología , Movimiento Celular/genética , Femenino , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Transgénicos , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
11.
J Immunol ; 203(4): 844-852, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31324724

RESUMEN

Programmed death-1 (PD-1) inhibits T and B cell function upon ligand binding. PD-1 blockade revolutionized cancer treatment, and although numerous patients respond, some develop autoimmune-like symptoms or overt autoimmunity characterized by autoantibody production. PD-1 inhibition accelerates autoimmunity in mice, but its role in regulating germinal centers (GC) is controversial. To address the role of PD-1 in the GC reaction in type 1 diabetes, we used tetramers to phenotype insulin-specific CD4+ T and B cells in NOD mice. PD-1 or PD-L1 deficiency, and PD-1 but not PD-L2 blockade, unleashed insulin-specific T follicular helper CD4+ T cells and enhanced their survival. This was concomitant with an increase in GC B cells and augmented insulin autoantibody production. The effect of PD-1 blockade on the GC was reduced when mice were treated with a mAb targeting the insulin peptide:MHC class II complex. This work provides an explanation for autoimmune side effects following PD-1 pathway inhibition and suggests that targeting the self-peptide:MHC class II complex might limit autoimmunity arising from checkpoint blockade.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Animales , Antígeno B7-H1/inmunología , Diabetes Mellitus Experimental/inmunología , Femenino , Centro Germinal/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos NOD
12.
J Immunol ; 202(9): 2535-2545, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30858199

RESUMEN

Naive CD4+ T lymphocytes differentiate into various Th cell subsets following TCR binding to microbial peptide:MHC class II (p:MHCII) complexes on dendritic cells (DCs). The affinity of the TCR interaction with p:MHCII plays a role in Th differentiation by mechanisms that are not completely understood. We found that low-affinity TCRs biased mouse naive T cells to become T follicular helper (Tfh) cells, whereas higher-affinity TCRs promoted the formation of Th1 or Th17 cells. We explored the basis for this phenomenon by focusing on IL-2R signaling, which is known to promote Th1 and suppress Tfh cell differentiation. SIRP⍺+ DCs produce abundant p:MHCII complexes and consume IL-2, whereas XCR1+ DCs weakly produce p:MHCII but do not consume IL-2. We found no evidence, however, of preferential interactions between Th1 cell-prone, high-affinity T cells and XCR1+ DCs or Tfh cell-prone, low-affinity T cells and SIRP⍺+ DCs postinfection with bacteria expressing the peptide of interest. Rather, high-affinity T cells sustained IL-2R expression longer and expressed two novel Th cell differentiation regulators, Eef1e1 and Gbp2, to a higher level than low-affinity T cells. These results suggest that TCR affinity does not influence Th cell differentiation by biasing T cell interactions with IL-2-consuming DCs, but instead, directly regulates genes in naive T cells that control the differentiation process.


Asunto(s)
Diferenciación Celular/inmunología , Proteínas de Unión al GTP/inmunología , Regulación de la Expresión Génica/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Factores de Elongación de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Células TH1/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular/genética , Células Dendríticas/citología , Células Dendríticas/inmunología , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica/genética , Subunidad alfa del Receptor de Interleucina-2/genética , Ratones , Ratones Noqueados , Factores de Elongación de Péptidos/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células TH1/citología , Células Th2/citología
13.
Cell Rep ; 26(11): 2859-2867.e4, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30865878

RESUMEN

The migratory capacity of adaptive CD8αß T cells dictates their ability to locate target cells and exert cytotoxicity, which is the basis of immune surveillance for the containment of microbes and disease. The small intestine (SI) is the largest mucosal surface and is a primary site of pathogen entrance. Using two-photon laser scanning microscopy, we found that motility of antigen (Ag)-specific CD8αß T cells in the SI is dynamic and varies with the environmental milieu. Pathogen-specific CD8αß T cell movement differed throughout infection, becoming locally confined at memory. Motility was not dependent on CD103 but was influenced by micro-anatomical locations within the SI and by inflammation. CD8 T cells responding to self-protein were initially affected by the presence of self-Ag, but this was altered after complete tolerance induction. These studies identify multiple factors that affect CD8αß T cell movement in the intestinal mucosa and show the adaptability of CD8αß T cell motility.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Movimiento Celular , Intestino Delgado/citología , Animales , Linfocitos T CD8-positivos/inmunología , Inflamación , Intestino Delgado/inmunología , Intestino Delgado/patología , Ratones , Ratones Endogámicos C57BL
14.
J Immunol ; 202(1): 300-308, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30510065

RESUMEN

Advances in imaging have led to the development of powerful multispectral, quantitative imaging techniques, like histo-cytometry. The utility of this approach is limited, however, by the need for time consuming manual image analysis. We therefore developed the software Chrysalis and a group of Imaris Xtensions to automate this process. The resulting automation allowed for high-throughput histo-cytometry analysis of three-dimensional confocal microscopy and two-photon time-lapse images of T cell-dendritic cell interactions in mouse spleens. It was also applied to epi-fluorescence images to quantify T cell localization within splenic tissue by using a "signal absorption" strategy that avoids computationally intensive distance measurements. In summary, this image processing and analysis software makes histo-cytometry more useful for immunology applications by automating image analysis.


Asunto(s)
Células Dendríticas/patología , Procesamiento de Imagen Asistido por Computador/métodos , Sinapsis Inmunológicas/patología , Programas Informáticos , Linfocitos T/patología , Animales , Automatización de Laboratorios , Células Cultivadas , Femenino , Ensayos Analíticos de Alto Rendimiento , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Bazo/patología , Imagen de Lapso de Tiempo
15.
J Clin Invest ; 128(10): 4604-4621, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30106752

RESUMEN

Regulatory T cells (Tregs) are critical for maintaining immune homeostasis. However, current Treg immunotherapies do not optimally treat inflammatory diseases in patients. Understanding the cellular processes that control Treg function may allow for the augmentation of therapeutic efficacy. In contrast to activated conventional T cells, in which protein kinase C-θ (PKC-θ) localizes to the contact point between T cells and antigen-presenting cells, in human and mouse Tregs, PKC-θ localizes to the opposite end of the cell in the distal pole complex (DPC). Here, using a phosphoproteomic screen, we identified the intermediate filament vimentin as a PKC-θ phospho target and show that vimentin forms a DPC superstructure on which PKC-θ accumulates. Treatment of mouse Tregs with either a clinically relevant PKC-θ inhibitor or vimentin siRNA disrupted vimentin and enhanced Treg metabolic and suppressive activity. Moreover, vimentin-disrupted mouse Tregs were significantly better than controls at suppressing alloreactive T cell priming in graft-versus-host disease (GVHD) and GVHD lethality, using a complete MHC-mismatch mouse model of acute GVHD (C57BL/6 donor into BALB/c host). Interestingly, vimentin disruption augmented the suppressor function of PKC-θ-deficient mouse Tregs. This suggests that enhanced Treg activity after PKC-θ inhibition is secondary to effects on vimentin, not just PKC-θ kinase activity inhibition. Our data demonstrate that vimentin is a key metabolic and functional controller of Treg activity and provide proof of principle that disruption of vimentin is a feasible, translationally relevant method to enhance Treg potency.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Filamentos Intermedios/inmunología , Activación de Linfocitos , Linfocitos T Reguladores/inmunología , Vimentina/inmunología , Animales , Células Presentadoras de Antígenos/patología , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Humanos , Filamentos Intermedios/genética , Filamentos Intermedios/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteína Quinasa C-theta/genética , Proteína Quinasa C-theta/inmunología , Linfocitos T Reguladores/patología , Vimentina/genética
16.
J Immunol ; 201(2): 337-342, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29875151

RESUMEN

Lymphocytes enter tissues from blood vessels through a well-characterized three-step process of extravasation. To our knowledge, nonvascular routes of lymphocyte entry have not been described. In this article, we report that Ag-experienced CD8 T cells in mice recirculate from blood through the peritoneal cavity. In the event of infection, Ag-experienced CD8 T cell subsets adhered to visceral organs, indicating potential transcapsular immunosurveillance. Focusing on the male genital tract (MGT), we observed Ag-experienced CD8 T cell migration from the peritoneal cavity directly to the infected MGT across the capsule, which was dependent on the extracellular matrix receptor CD44. We also observed that, following clearance of infection, the MGT retained functional resident memory CD8 T cells. These data suggest that recirculation through body cavities may provide T cells with opportunities for broad immunosurveillance and potential nonvascular mechanisms of entry.


Asunto(s)
Subgrupos de Linfocitos T/inmunología , Animales , Movimiento Celular/inmunología , Matriz Extracelular/inmunología , Genitales Masculinos/inmunología , Receptores de Hialuranos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monitorización Inmunológica/métodos , Cavidad Peritoneal/fisiología , Infecciones del Sistema Genital/inmunología
17.
Sci Rep ; 8(1): 8295, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29844327

RESUMEN

Type 1 diabetes is caused by autoreactive T cell-mediated ß cell destruction. Even though co-inhibitory receptor programmed death-1 (PD-1) restrains autoimmunity, the expression and regulation of its cognate ligands on ß cell remains unknown. Here, we interrogated ß cell-intrinsic programmed death ligand-1 (PD-L1) expression in mouse and human islets. We measured a significant increase in the level of PD-L1 surface expression and the frequency of PD-L1+ ß cells as non-obese diabetic (NOD) mice aged and developed diabetes. Increased ß cell PD-L1 expression was dependent on T cell infiltration, as ß cells from Rag1-deficient mice lacked PD-L1. Using Rag1-deficient NOD mouse islets, we determined that IFN-γ promotes ß cell PD-L1 expression. We performed analogous experiments using human samples, and found a significant increase in ß cell PD-L1 expression in type 1 diabetic samples compared to type 2 diabetic, autoantibody positive, and non-diabetic samples. Among type 1 diabetic samples, ß cell PD-L1 expression correlated with insulitis. In vitro experiments with human islets from non-diabetic individuals showed that IFN-γ promoted ß cell PD-L1 expression. These results suggest that insulin-producing ß cells respond to pancreatic inflammation and IFN-γ production by upregulating PD-L1 expression to limit self-reactive T cells.


Asunto(s)
Antígeno B7-H1/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Interferón gamma/metabolismo , Islotes Pancreáticos/metabolismo , Linfocitos T/inmunología , Animales , Femenino , Humanos , Interferón gamma/biosíntesis , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD
18.
Immunity ; 48(2): 327-338.e5, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466758

RESUMEN

Immunosurveillance of secondary lymphoid organs (SLO) is performed by central memory T cells that recirculate through blood. Resident memory T (Trm) cells remain parked in nonlymphoid tissues and often stably express CD69. We recently identified Trm cells within SLO, but the origin and phenotype of these cells remains unclear. Using parabiosis of "dirty" mice, we found that CD69 expression is insufficient to infer stable residence of SLO Trm cells. Restimulation of nonlymphoid memory CD8+ T cells within the skin or mucosa resulted in a substantial increase in bona fide Trm cells specifically within draining lymph nodes. SLO Trm cells derived from emigrants from nonlymphoid tissues and shared some transcriptional and phenotypic signatures associated with nonlymphoid Trm cells. These data indicate that nonlymphoid cells can give rise to SLO Trm cells and suggest vaccination strategies by which memory CD8+ T cell immunosurveillance can be regionalized to specific lymph nodes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Ganglios Linfáticos/inmunología , Animales , Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Femenino , Lectinas Tipo C/análisis , Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL
19.
Nat Immunol ; 19(2): 173-182, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29311694

RESUMEN

CD8+ T cell immunosurveillance dynamics influence the outcome of intracellular infections and cancer. Here we used two-photon intravital microscopy to visualize the responses of CD8+ resident memory T cells (TRM cells) within the reproductive tracts of live female mice. We found that mucosal TRM cells were highly motile, but paused and underwent in situ division after local antigen challenge. TRM cell reactivation triggered the recruitment of recirculating memory T cells that underwent antigen-independent TRM cell differentiation in situ. However, the proliferation of pre-existing TRM cells dominated the local mucosal recall response and contributed most substantially to the boosted secondary TRM cell population. We observed similar results in skin. Thus, TRM cells can autonomously regulate the expansion of local immunosurveillance independently of central memory or proliferation in lymphoid tissue.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa/inmunología , Memoria Inmunológica/inmunología , Vigilancia Inmunológica/inmunología , Membrana Mucosa/inmunología , Animales , Femenino , Microscopía Intravital , Ratones , Membrana Mucosa/citología , Piel/inmunología
20.
J Immunol ; 191(5): 2372-83, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23918975

RESUMEN

The Ag-specific interactions between T cells and dendritic cells progress through dynamic contact stages in vivo consisting of early long-term stable contacts and later confined, yet motile, short-lived contacts. The signaling pathways that control in vivo interaction dynamics between T cells and dendritic cells during priming remain undefined. Adhesion and degranulation promoting adapter protein (ADAP) is a multifunctional adapter that regulates "inside-out" signaling from the TCR to integrins. Using two-photon microscopy, we demonstrate that, in the absence of ADAP, CD4 T cells make fewer early-stage stable contacts with Ag-laden dendritic cells, and the interactions are characterized by brief repetitive contacts. Furthermore, ADAP-deficient T cells show reduced contacts at the late motile contact phase and display less confinement around dendritic cells. The altered T cell interaction dynamics in the absence of ADAP are associated with defective early proliferation and attenuated TCR signaling in vivo. Regulation of multistage contact behaviors and optimal T cell signaling involves the interaction of ADAP with the adapter src kinase-associated phosphoprotein of 55 kDa (SKAP55). Thus, integrin activation by the ADAP-SKAP55-signaling module controls the stability and duration of T cell-dendritic cell contacts during the progressive phases necessary for optimal T cell activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Linfocitos T CD4-Positivos/inmunología , Comunicación Celular/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Proteínas de la Membrana/inmunología , Fosfoproteínas/inmunología , Animales , Integrinas/inmunología , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Rastreo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...