Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
STAR Protoc ; 3(4): 101784, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36386869

ABSTRACT

It is technically challenging to generate large doses of regulatory T cells (Tregs) engineered to express a chimeric antigen receptor (CAR) in non-human primates (NHP). Here, we have optimized the manufacturing of CAR Tregs by stringent sorting of Tregs, stimulation by artificial antigen-presenting cells, transduction by simian tropic lentiviral vectors, and antigen-specific expansion. The result of this method is highly suppressive CAR Tregs for use in a pre-clinical, large animal model of transplant tolerance. For complete details on the use and execution of this protocol, please refer to Ellis et al. (2022).


Subject(s)
Receptors, Chimeric Antigen , Animals , Receptors, Chimeric Antigen/genetics , Antigen-Presenting Cells , T-Lymphocytes, Regulatory , Primates , Transplantation Tolerance
2.
J Immunol ; 209(5): 864-873, 2022 09 01.
Article in English | MEDLINE | ID: mdl-36130133

ABSTRACT

HIV-1-specific CD4+ T cells (TCD4+s) play a critical role in controlling HIV-1 infection. Canonically, TCD4+s are activated by peptides derived from extracellular ("exogenous") Ags displayed in complex with MHC class II (MHC II) molecules on the surfaces of "professional" APCs such as dendritic cells (DCs). In contrast, activated human TCD4+s, which express MHC II, are not typically considered for their APC potential because of their low endocytic capacity and the exogenous Ag systems historically used for assessment. Using primary TCD4+s and monocyte-derived DCs from healthy donors, we show that activated human TCD4+s are highly effective at MHC II-restricted presentation of an immunodominant HIV-1-derived epitope postinfection and subsequent noncanonical processing and presentation of endogenously produced Ag. Our results indicate that, in addition to marshalling HIV-1-specific immune responses during infection, TCD4+s also act as APCs, leading to the activation of HIV-1-specific TCD4+s.


Subject(s)
HIV Seropositivity , HIV-1 , Antigen Presentation , CD4-Positive T-Lymphocytes , Dendritic Cells , Epitopes , Histocompatibility Antigens Class II , Humans , Peptides , T-Lymphocytes
3.
Cell Rep Med ; 3(5): 100614, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35551746

ABSTRACT

Adoptive transfer of chimeric antigen receptor regulatory T cells (CAR Tregs) is a promising way to prevent allograft loss without the morbidity associated with current therapies. Non-human primates (NHPs) are a clinically relevant model to develop transplant regimens, but manufacturing and engraftment of NHP CAR Tregs have not been demonstrated yet. Here, we describe a culture system that massively expands CAR Tregs specific for the Bw6 alloantigen. In vitro, these Tregs suppress in an antigen-specific manner without pro-inflammatory cytokine secretion or cytotoxicity. In vivo, Bw6-specific CAR Tregs preferentially traffic to and persist in bone marrow for at least 1 month. Following transplant of allogeneic Bw6+ islets and autologous CAR Tregs into the bone marrow of diabetic recipients, CAR Tregs traffic to the site of islet transplantation and maintain a phenotype of suppressive Tregs. Our results establish a framework for the optimization of CAR Treg therapy in NHP disease models.


Subject(s)
Isoantigens , Receptors, Chimeric Antigen , Adoptive Transfer , Animals , Macaca , Receptors, Chimeric Antigen/genetics , T-Lymphocytes, Regulatory
4.
Nat Rev Genet ; 22(7): 427-447, 2021 07.
Article in English | MEDLINE | ID: mdl-33603158

ABSTRACT

Genetically engineered T cell immunotherapies have provided remarkable clinical success to treat B cell acute lymphoblastic leukaemia by harnessing a patient's own T cells to kill cancer, and these approaches have the potential to provide therapeutic benefit for numerous other cancers, infectious diseases and autoimmunity. By introduction of either a transgenic T cell receptor or a chimeric antigen receptor, T cells can be programmed to target cancer cells. However, initial studies have made it clear that the field will need to implement more complex levels of genetic regulation of engineered T cells to ensure both safety and efficacy. Here, we review the principles by which our knowledge of genetics and genome engineering will drive the next generation of adoptive T cell therapies.


Subject(s)
Genetic Engineering , Immunotherapy , T-Lymphocytes/immunology , Animals , Genetic Engineering/trends , Humans , Transgenes
5.
Blood ; 136(15): 1722-1734, 2020 10 08.
Article in English | MEDLINE | ID: mdl-32614969

ABSTRACT

Chimeric antigen receptor (CAR) T cells targeting CD19+ hematologic malignancies have rapidly emerged as a promising, novel therapy. In contrast, results from the few CAR T-cell studies for infectious diseases such as HIV-1 have been less convincing. These challenges are likely due to the low level of antigen present in antiretroviral therapy (ART)-suppressed patients in contrast to those with hematologic malignancies. Using our well-established nonhuman primate model of ART-suppressed HIV-1 infection, we tested strategies to overcome these limitations and challenges. We first optimized CAR T-cell production to maintain central memory subsets, consistent with current clinical paradigms. We hypothesized that additional exogenous antigen might be required in an ART-suppressed setting to aid expansion and persistence of CAR T cells. Thus, we studied 4 simian/HIV-infected, ART-suppressed rhesus macaques infused with virus-specific CD4CAR T cells, followed by supplemental infusion of cell-associated HIV-1 envelope (Env). Env boosting led to significant and unprecedented expansion of virus-specific CAR+ T cells in vivo; after ART treatment interruption, viral rebound was significantly delayed compared with controls (P = .014). In 2 animals with declining CAR T cells, rhesusized anti-programmed cell death protein 1 (PD-1) antibody was administered to reverse PD-1-dependent immune exhaustion. Immune checkpoint blockade triggered expansion of exhausted CAR T cells and concordantly lowered viral loads to undetectable levels. These results show that supplemental cell-associated antigen enables robust expansion of CAR T cells in an antigen-sparse environment. To our knowledge, this is the first study to show expansion of virus-specific CAR T cells in infected, suppressed hosts, and delay/control of viral recrudescence.


Subject(s)
Antigens, Viral/immunology , HIV Infections/immunology , HIV-1/immunology , Immunocompromised Host , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antiretroviral Therapy, Highly Active/adverse effects , Antiretroviral Therapy, Highly Active/methods , Disease Models, Animal , HIV Infections/drug therapy , HIV Infections/virology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/genetics , Immune Checkpoint Proteins/metabolism , Macaca mulatta , Simian Immunodeficiency Virus/immunology , T-Lymphocytes/drug effects
7.
Nat Rev Immunol ; 18(10): 605-616, 2018 10.
Article in English | MEDLINE | ID: mdl-30046149

ABSTRACT

Chimeric antigen receptors (CARs) have shown remarkable ability to re-direct T cells to target CD19-expressing tumours, resulting in remission rates of up to 90% in individuals with paediatric acute lymphoblastic lymphoma. Lessons learned from these clinical trials of adoptive T cell therapy for cancer, as well as investments made in manufacturing T cells at commercial scale, have inspired researchers to develop CARs for additional applications. Here, we explore the challenges and opportunities of using this technology to target infectious diseases such as with HIV and undesired immune responses such as autoimmunity and transplant rejection. Despite substantial obstacles, the potential of CAR T cells to enable cures for a wide array of disease settings could be transformational for the medical field.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , HIV Infections/immunology , HIV Infections/therapy , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Chimeric Antigen/therapeutic use
8.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27178244

ABSTRACT

BACKGROUND: Nociceptive and neuropathic pain occurs as part of the disease process after traumatic brain injury (TBI) in humans. Central and peripheral inflammation, a major secondary injury process initiated by the traumatic brain injury event, has been implicated in the potentiation of peripheral nociceptive pain. We hypothesized that the inflammatory response to diffuse traumatic brain injury potentiates persistent pain through prolonged immune dysregulation. RESULTS: To test this, adult, male C57BL/6 mice were subjected to midline fluid percussion brain injury or to sham procedure. One cohort of mice was analyzed for inflammation-related cytokine levels in cortical biopsies and serum along an acute time course. In a second cohort, peripheral inflammation was induced seven days after surgery/injury with an intraplantar injection of carrageenan. This was followed by measurement of mechanical hyperalgesia, glial fibrillary acidic protein and Iba1 immunohistochemical analysis of neuroinflammation in the brain, and flow cytometric analysis of T-cell differentiation in mucosal lymph. Traumatic brain injury increased interleukin-6 and chemokine ligand 1 levels in the cortex and serum that peaked within 1-9 h and then resolved. Intraplantar carrageenan produced mechanical hyperalgesia that was potentiated by traumatic brain injury. Further, mucosal T cells from brain-injured mice showed a distinct deficiency in the ability to differentiate into inflammation-suppressing regulatory T cells (Tregs). CONCLUSIONS: We conclude that traumatic brain injury increased the inflammatory pain associated with cutaneous inflammation by contributing to systemic immune dysregulation. Regulatory T cells are immune suppressors and failure of T cells to differentiate into regulatory T cells leads to unregulated cytokine production which may contribute to the potentiation of peripheral pain through the excitation of peripheral sensory neurons. In addition, regulatory T cells are identified as a potential target for therapeutic rebalancing of peripheral immune homeostasis to improve functional outcome and decrease the incidence of peripheral inflammatory pain following traumatic brain injury.


Subject(s)
Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/immunology , Hyperalgesia/etiology , Hyperalgesia/immunology , Animals , Inflammation/complications , Inflammation/pathology , Male , Mice, Inbred C57BL , Models, Biological , T-Lymphocytes, Regulatory/immunology
9.
Dig Dis Sci ; 60(10): 2976-84, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25956706

ABSTRACT

BACKGROUND: Inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), are characterized by chronic intestinal inflammation due to immunological, microbial, and environmental factors in genetically predisposed individuals. Advances in the diagnosis, prognosis, and treatment of IBD require the identification of robust biomarkers that can be used for molecular classification of diverse disease presentations. We previously identified five genes, RELA, TNFAIP3 (A20), PIGR, TNF, and IL8, whose mRNA levels in colonic mucosal biopsies could be used in a multivariate analysis to classify patients with CD based on disease behavior and responses to therapy. AIM: We compared expression of these five biomarkers in IBD patients classified as having CD or UC, and in healthy controls. RESULTS: Patients with CD were characterized as having decreased median expression of TNFAIP3, PIGR, and TNF in non-inflamed colonic mucosa as compared to healthy controls. By contrast, UC patients exhibited decreased expression of PIGR and elevated expression of IL8 in colonic mucosa compared to healthy controls. A multivariate analysis combining mRNA levels for all five genes resulted in segregation of individuals based on disease presentation (CD vs. UC) as well as severity, i.e., patients in remission versus those with acute colitis at the time of biopsy. CONCLUSION: We propose that this approach could be used as a model for molecular classification of IBD patients, which could further be enhanced by the inclusion of additional genes that are identified by functional studies, global gene expression analyses, and genome-wide association studies.


Subject(s)
Colitis, Ulcerative/genetics , Crohn Disease/genetics , DNA-Binding Proteins/genetics , Interleukin-8/genetics , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , RNA, Messenger/genetics , Receptors, Polymeric Immunoglobulin/genetics , Transcription Factor RelA/genetics , Adolescent , Adult , Aged , Chi-Square Distribution , Colitis, Ulcerative/pathology , Crohn Disease/pathology , DNA-Binding Proteins/metabolism , Female , Gene Expression , Genetic Markers , Humans , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Multivariate Analysis , Nuclear Proteins/metabolism , Phenotype , Transcription Factor RelA/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/genetics , Young Adult
10.
Eur J Immunol ; 43(12): 3355-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24037540

ABSTRACT

Mutations in PTEN-induced kinase 1 (PINK1), a serine/threonine kinase linked to familial early-onset Parkinsonism, compromise mitochondrial integrity and metabolism and impair AKT signaling. As the activation of a naïve T cell requires an AKT-dependent reorganization of a cell's metabolic machinery, we sought to determine if PINK1-deficient T cells lack the ability to undergo activation and differentiation. We show that CD4(+) T cells from PINK1 knockout mice fail to properly phosphorylate AKT upon activation, resulting in reduced expression of the IL-2 receptor subunit CD25. Following, deficient IL-2 signaling mutes the activation-induced increase in respiratory capacity and mitochondrial membrane potential. Under polarization conditions favoring the development of induced regulatory T cells, PINK1(-/-) T cells exhibit a reduced ability to suppress bystander T-cell proliferation despite normal FoxP3 expression kinetics. Our results describe a critical role for PINK1 in integrating extracellular signals with metabolic state during T-cell fate determination, and may have implications for the understanding of altered T-cell populations and immunity during the progression of active Parkinson's disease or other immunopathologies.


Subject(s)
Cell Differentiation/immunology , Cytosol/immunology , Lymphocyte Activation , Mitochondria/immunology , Protein Kinases/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Knockout , Mitochondria/genetics , Parkinson Disease/genetics , Parkinson Disease/immunology , Parkinson Disease/pathology , Phosphorylation/genetics , Phosphorylation/immunology , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , T-Lymphocytes, Regulatory/pathology
11.
J Asthma ; 50(3): 223-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23259774

ABSTRACT

Introduction. A dysregulation of regulatory T cells (Tregs) could play a major role in the pathogenesis of bronchial asthma. Sex-dependent differences as well as the impact of hormonal changes in the incidence and severity of asthma are widely recognized. Emerging evidence suggests that asthma symptoms are alleviated in female patients taking hormone oral contraceptives (OCs). The impact of OCs on the generation of induced Tregs (iTregs) was assessed in a cohort of female patients with asthma. Methods. Thirteen patients were included in this pilot study. During three distinct phases of their menstrual cycles, we measured exhaled nitric oxide (eNO) levels, forced expiratory volume at 1 second (FEV1s), asthma control test (ACT) score, sex steroid hormone levels in serum, natural Tregs in peripheral blood, and the ability of CD4(+) T cells to generate iTregs ex vivo. Results. The luteal serum levels of estradiol and progesterone negatively correlated with the proportion of iTregs generated ex vivo in patients not taking OCs. In addition, physiological doses of estradiol and progesterone prevented the acquisition of a suppressor T cell phenotype in vitro. Interestingly, patients taking OCs had reduced serum sex hormone levels associated with higher iTreg induction, a better ACT score, and a tendency toward lower eNO levels. Conclusions. Our results identify an impact of sex hormones on the capacity of T cells to polarize towards a regulatory phenotype and suggest the regulation of peripheral T cell lineage plasticity as a potential mechanism underlying the beneficial effects of OCs in women with asthma.


Subject(s)
Asthma/immunology , Contraceptives, Oral/pharmacology , T-Lymphocytes, Regulatory/drug effects , Adult , Asthma/blood , Breath Tests , Cohort Studies , Estradiol/blood , Estradiol/immunology , Female , Flow Cytometry , Forced Expiratory Volume/drug effects , Forced Expiratory Volume/immunology , Humans , Menstrual Cycle/drug effects , Menstrual Cycle/immunology , Nitric Oxide/immunology , Pilot Projects , Progesterone/blood , Progesterone/immunology , Statistics, Nonparametric , T-Lymphocytes, Regulatory/immunology , Young Adult
12.
J Vis Exp ; (62)2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22525890

ABSTRACT

The development and maintenance of immunosuppressive CD4+ regulatory T cells (Tregs) contribute to the peripheral tolerance needed to remain in immunologic homeostasis with the vast amount of self and commensal antigens in and on the human body. Perturbations in the balance between Tregs and inflammatory conventional T cells can result in immunopathology or cancer. Although therapeutic injection of Tregs has been shown to be efficacious in murine models of colitis, type I diabetes, rheumatoid arthritis and graft versus host disease, several fundamental differences in human versus mouse Treg biology has thus far precluded clinical use. The lack of sufficient number, purity, stability and homing specificity of therapeutic Tregs necessitated a dynamic platform of human Treg development on which to optimize conditions for their ex vivo expansion. Here we describe a method for the differentiation of induced Tregs (iTregs) from a single human peripheral blood donor which can be broken down into four stages: isolation of peripheral blood mononuclear cells, magnetic selection of CD4+ T cells, in vitro cell culture and fluorescence activated cell sorting (FACS) of T cell subsets. Since the Treg signature transcription factor forkhead box P3 (FoxP3) is an activation-induced transcription factor in humans and no other unique marker exists, a combinatorial panel of markers must be used to identify T cells with suppressor activity. After six days in culture, cells in our system can be demarcated into naïve T cells, memory T cells or iTregs based on their relative expression of CD25 and CD45RA. As memory and naïve T cells have different reported polarization requirements and plasticities, pre-sorting of the initial T cell population into CD45RA(+) and CD45RO(+) subsets can be used to examine these discrepancies. Consistent with others, our CD25(Hi)CD45RA(-) iTregs express high levels of FoxP3, GITR and CTLA-4 and low levels of CD127. Following FACS of each population, resultant cells can be used in a suppressor assay which evaluates the relative ability to retard the proliferation of carboxyfluorescein succinimidyl ester (CFSE)-labeled autologous T cells.


Subject(s)
Immunologic Memory , T-Lymphocytes, Regulatory/immunology , Cell Culture Techniques , Cell Differentiation/immunology , Flow Cytometry/methods , Fluorescent Dyes/chemistry , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Common Antigens/biosynthesis , Leukocyte Common Antigens/immunology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Magnetics/methods , T-Lymphocytes, Regulatory/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...