Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nature ; 629(8010): 201-210, 2024 May.
Article in English | MEDLINE | ID: mdl-38600376

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of haematological malignancies such as acute lymphoblastic leukaemia, B cell lymphoma and multiple myeloma1-4, but the efficacy of CAR T cell therapy in solid tumours has been limited5. This is owing to a number of factors, including the immunosuppressive tumour microenvironment that gives rise to poorly persisting and metabolically dysfunctional T cells. Analysis of anti-CD19 CAR T cells used clinically has shown that positive treatment outcomes are associated with a more 'stem-like' phenotype and increased mitochondrial mass6-8. We therefore sought to identify transcription factors that could enhance CAR T cell fitness and efficacy against solid tumours. Here we show that overexpression of FOXO1 promotes a stem-like phenotype in CAR T cells derived from either healthy human donors or patients, which correlates with improved mitochondrial fitness, persistence and therapeutic efficacy in vivo. This work thus reveals an engineering approach to genetically enforce a favourable metabolic phenotype that has high translational potential to improve the efficacy of CAR T cells against solid tumours.


Subject(s)
Forkhead Box Protein O1 , Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Stem Cells , T-Lymphocytes , Humans , Mice , Cell Line, Tumor , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Mitochondria/metabolism , Phenotype , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/cytology , Tumor Microenvironment/immunology , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy
2.
EMBO J ; 39(2): e103637, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31803974

ABSTRACT

Although adoptive T-cell therapy has shown remarkable clinical efficacy in haematological malignancies, its success in combating solid tumours has been limited. Here, we report that PTPN2 deletion in T cells enhances cancer immunosurveillance and the efficacy of adoptively transferred tumour-specific T cells. T-cell-specific PTPN2 deficiency prevented tumours forming in aged mice heterozygous for the tumour suppressor p53. Adoptive transfer of PTPN2-deficient CD8+ T cells markedly repressed tumour formation in mice bearing mammary tumours. Moreover, PTPN2 deletion in T cells expressing a chimeric antigen receptor (CAR) specific for the oncoprotein HER-2 increased the activation of the Src family kinase LCK and cytokine-induced STAT-5 signalling, thereby enhancing both CAR T-cell activation and homing to CXCL9/10-expressing tumours to eradicate HER-2+ mammary tumours in vivo. Our findings define PTPN2 as a target for bolstering T-cell-mediated anti-tumour immunity and CAR T-cell therapy against solid tumours.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Neoplasms/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 2/physiology , Receptor, ErbB-2/physiology , Receptors, Antigen, T-Cell/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction
3.
Trends Immunol ; 42(12): 1128-1142, 2021 12.
Article in English | MEDLINE | ID: mdl-34750058

ABSTRACT

Tumor necrosis factor (TNF) is a proinflammatory cytokine that is produced and secreted by cytotoxic lymphocytes upon tumor target recognition. Depending on the context, TNF can mediate either pro-survival or pro-death signals. The potential cytotoxicity of T cell-produced TNF, particularly in the context of T cell-directed immunotherapies, has been largely overlooked. However, a spate of recent studies investigating tumor immune evasion through the application of CRISPR-based gene-editing screens have highlighted TNF-mediated killing as an important component of the mammalian T cell antitumor repertoire. In the context of the current understanding of the role of TNF in antitumor immunity, we discuss these studies and touch on their therapeutic implications. Collectively, we provide an enticing prospect to augment immunotherapy responses through TNF cytotoxicity.


Subject(s)
Immunotherapy , Neoplasms , Animals , Cytotoxicity, Immunologic , Humans , Immunotherapy/methods , Mammals , Neoplasms/therapy , T-Lymphocytes , Tumor Escape , Tumor Necrosis Factors
4.
EMBO Rep ; 22(11): e53391, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34467615

ABSTRACT

The success of cancer immunotherapy is limited to a subset of patients, highlighting the need to identify the processes by which tumors evade immunity. Using CRISPR/Cas9 screening, we reveal that melanoma cells lacking HOIP, the catalytic subunit of LUBAC, are highly susceptible to both NK and CD8+ T-cell-mediated killing. We demonstrate that HOIP-deficient tumor cells exhibit increased sensitivity to the combined effect of the inflammatory cytokines, TNF and IFN-γ, released by NK and CD8+ T cells upon target recognition. Both genetic deletion and pharmacological inhibition of HOIP augment tumor cell sensitivity to combined TNF and IFN-γ. Together, we unveil a protective regulatory axis, involving HOIP, which limits a transcription-dependent form of cell death that engages both intrinsic and extrinsic apoptotic machinery upon exposure to TNF and IFN-γ. Our findings highlight HOIP inhibition as a potential strategy to harness and enhance the killing capacity of TNF and IFN-γ during immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Ubiquitin-Protein Ligases , Apoptosis/genetics , Humans , Interferon-gamma/pharmacology , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/metabolism
5.
J Immunol ; 204(8): 2308-2315, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32152070

ABSTRACT

CRISPR/Cas9 technologies have revolutionized our understanding of gene function in complex biological settings, including T cell immunology. Current CRISPR-mediated gene editing strategies in T cells require in vitro stimulation or culture that can both preclude the study of unmanipulated naive T cells and alter subsequent differentiation. In this study, we demonstrate highly efficient gene editing within uncultured primary naive murine CD8+ T cells by electroporation of recombinant Cas9/sgRNA ribonucleoprotein immediately prior to in vivo adoptive transfer. Using this approach, we generated single and double gene knockout cells within multiple mouse infection models. Strikingly, gene deletion occurred even when the transferred cells were left in a naive state, suggesting that gene deletion occurs independent of T cell activation. Finally, we demonstrate that targeted mutations can be introduced into naive CD8+ T cells using CRISPR-based homology-directed repair. This protocol thus expands CRISPR-based gene editing approaches beyond models of robust T cell activation to encompass both naive T cell homeostasis and models of weak activation, such as tolerance and tumor models.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing , Animals , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , Electroporation , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Single Nucleotide/genetics , Polymorphism, Single Nucleotide/immunology
6.
Eur J Immunol ; 49(5): 770-781, 2019 05.
Article in English | MEDLINE | ID: mdl-30729501

ABSTRACT

Mutation of Dedicator of cytokinesis 8 (DOCK8) has previously been reported to provide resistance to the Th17 cell dependent EAE in mice. Contrary to expectation, we observed an elevation of Th17 cells in two different DOCK8 mutant mouse strains in the steady state. This was specific for Th17 cells with no change in Th1 or Th2 cell populations. In vitro Th cell differentiation assays revealed that the elevated Th17 cell population was not due to a T cell intrinsic differentiation bias. Challenging these mutant mice in the EAE model, we confirmed a resistance to this autoimmune disease with Th17 cells remaining elevated systemically while cellular infiltration in the CNS was reduced. Infiltrating T cells lost the bias toward Th17 cells indicating a relative reduction of Th17 cells in the CNS and a Th17 cell specific migration disadvantage. Adoptive transfers of Th1 and Th17 cells in EAE-affected mice further supported the Th17 cell-specific migration defect, however, DOCK8-deficient Th17 cells expressed normal Th17 cell-specific CCR6 levels and migrated toward chemokine gradients in transwell assays. This study shows that resistance to EAE in DOCK8 mutant mice is achieved despite a systemic Th17 bias.


Subject(s)
Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/etiology , Guanine Nucleotide Exchange Factors/genetics , Lymphocyte Count , Mutation , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Biomarkers , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression , Genetic Predisposition to Disease , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
Immunity ; 34(4): 492-504, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21497118

ABSTRACT

Polarized segregation of proteins in T cells is thought to play a role in diverse cellular functions including signal transduction, migration, and directed secretion of cytokines. Persistence of this polarization can result in asymmetric segregation of fate-determining proteins during cell division, which may enable a T cell to generate diverse progeny. Here, we provide evidence that a lineage-determining transcription factor, T-bet, underwent asymmetric organization in activated T cells preparing to divide and that it was unequally partitioned into the two daughter cells. This unequal acquisition of T-bet appeared to result from its asymmetric destruction during mitosis by virtue of concomitant asymmetric segregation of the proteasome. These results suggest a mechanism by which a cell may unequally localize cellular activities during division, thereby imparting disparity in the abundance of cell fate regulators in the daughter cells.


Subject(s)
Mitosis , Proteasome Endopeptidase Complex/metabolism , T-Box Domain Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Polarity , Cells, Cultured , Mice , Mice, Inbred C57BL , Phosphorylation , T-Box Domain Proteins/metabolism , T-Lymphocytes/enzymology
8.
J Immunol ; 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28794229

ABSTRACT

Mutations in the dedicator of cytokinesis 8 (DOCK8) gene cause an autosomal recessive form of hyper-IgE syndrome, characterized by chronic immunodeficiency with persistent microbial infection and increased incidence of malignancy. These manifestations suggest a defect in cytotoxic lymphocyte function and immune surveillance. However, how DOCK8 regulates NK cell-driven immune responses remains unclear. In this article, we demonstrate that DOCK8 regulates NK cell cytotoxicity and cytokine production in response to target cell engagement or receptor ligation. Genetic ablation of DOCK8 in human NK cells attenuated cytokine transcription and secretion through inhibition of Src family kinase activation, particularly Lck, downstream of target cell engagement or NKp30 ligation. PMA/Ionomycin treatment of DOCK8-deficient NK cells rescued cytokine production, indicating a defect proximal to receptor ligation. Importantly, NK cells from DOCK8-deficient patients had attenuated production of IFN-γ and TNF-α upon NKp30 stimulation. Taken together, we reveal a novel molecular mechanism by which DOCK8 regulates NK cell-driven immunity.

9.
Immunol Cell Biol ; 94(3): 306-11, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26391810

ABSTRACT

Lethal giant larvae-1 (Lgl-1) is an evolutionary conserved protein that regulates cell polarity in diverse lineages; however, the role of Lgl-1 in the polarity and function of immune cells remains to be elucidated. To assess the role of Lgl-1 in T cells, we generated chimeric mice with a hematopoietic system deficient for Lgl-1. Lgl-1 deficiency did not impair the activation or function of peripheral CD8(+) T cells in response to antigen presentation in vitro, but did skew effector and memory T-cell differentiation. When challenged with antigen-expressing virus or tumor, Lgl-1-deficient mice displayed altered T-cell responses. This manifested in a stronger antiviral and antitumor effector CD8(+) T-cell response, the latter resulting in enhanced control of MC38-OVA tumors. These results reveal a novel role for Lgl-1 in the regulation of virus-specific T-cell responses and antitumor immunity.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Glycoproteins/deficiency , Animals , Antigen Presentation/immunology , Immunophenotyping , Influenza A virus/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Phenotype
10.
Crit Rev Immunol ; 35(4): 325-47, 2015.
Article in English | MEDLINE | ID: mdl-26757394

ABSTRACT

A synapse is a specialized structure that forms when the plasma membrane of two cells come into close contact to facilitate communication and signaling. Cells of the immune system form 'immunological' synapses that have an ordered structure and are essential for immune cell activation, function and homeostasis. Optimal synapse formation is not only critical for the generation of effective immunity against pathogens but is also essential for immune surveillance against cancer and for the prevention of immune disorders. Not surprisingly, defective synapse formation can therefore have severe consequences for human health, culminating in poor immune function leading to immunodeficiency disease or failure to detect and control infected or cancerous cells. Here, we discuss the immunological synapse formed by cytotoxic lymphocytes in both immunodeficiency diseases and anticancer immunity and touch on novel therapies that may alter or enhance synapse formation.


Subject(s)
Immunologic Deficiency Syndromes/immunology , Immunological Synapses/metabolism , Immunotherapy , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Homeostasis , Humans , Immunity, Cellular , Immunologic Deficiency Syndromes/therapy , Immunologic Surveillance , Neoplasms/therapy
11.
J Immunol ; 192(2): 553-7, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24337740

ABSTRACT

DNAX accessory molecule 1 (DNAM-1) is expressed on all CD8(+) T cells and promotes their activation and effector function. DNAM-1 interacts with LFA-1, a critical molecule for immunological synapse formation between T cells and APCs, and for cytotoxic killing of target cells. Mice that lack DNAM-1 display abnormal T cell responses and antitumor activity; however, the mechanism involved is unclear. In this article, we show that DNAM-1 deficiency results in reduced proliferation of CD8(+) T cells after Ag presentation and impaired cytotoxic activity. We also demonstrate that DNAM-1-deficient T cells show reduced conjugations with tumor cells and decreased recruitment of both LFA-1 and lipid rafts to the immunological synapse, which correlates with reduced tumor cell killing in vitro. This synapse defect may explain why DNAM-1-deficient mice cannot clear tumors in vivo, and highlights the importance of DNAM-1 and the immunological synapse in T cell-mediated antitumor immunity.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunological Synapses/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation , Immunological Synapses/genetics , Immunological Synapses/metabolism , Lipids/genetics , Lipids/immunology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL
12.
J Immunol ; 193(11): 5744-50, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25348626

ABSTRACT

Cytotoxic lymphocytes destroy pathogen-infected and transformed cells through the cytotoxic granule exocytosis death pathway, which is dependent on the delivery of proapoptotic granzymes into the target cell cytosol by the pore-forming protein, perforin. Despite the importance of mouse models in understanding the role of cytotoxic lymphocytes in immune-mediated disease and their role in cancer immune surveillance, no reliable intracellular detection method exists for mouse perforin. Consequently, rapid, flow-based assessment of cytotoxic potential has been problematic, and complex assays of function are generally required. In this study, we have developed a novel method for detecting perforin in primary mouse cytotoxic T lymphocytes by immunofluorescence and flow cytometry. We used this new technique to validate perforin colocalization with granzyme B in cytotoxic granules polarized to the immunological synapse, and to assess the expression of perforin in cytotoxic T lymphocytes at various stages of activation. The sensitivity of this technique also allowed us to distinguish perforin levels in Prf1(+/+) and Prf1(+/-) mice. This new methodology will have broad applications and contribute to advances within the fields of lymphocyte biology, infectious disease, and cancer.


Subject(s)
Granzymes/metabolism , Immunological Synapses/metabolism , Intracellular Space/metabolism , Perforin/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Separation , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Perforin/genetics , Protein Transport
13.
J Immunol ; 185(1): 367-75, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530266

ABSTRACT

Asymmetric cell division is a potential means by which cell fate choices during an immune response are orchestrated. Defining the molecular mechanisms that underlie asymmetric division of T cells is paramount for determining the role of this process in the generation of effector and memory T cell subsets. In other cell types, asymmetric cell division is regulated by conserved polarity protein complexes that control the localization of cell fate determinants and spindle orientation during division. We have developed a tractable, in vitro model of naive CD8(+) T cells undergoing initial division while attached to dendritic cells during Ag presentation to investigate whether similar mechanisms might regulate asymmetric division of T cells. Using this system, we show that direct interactions with APCs provide the cue for polarization of T cells. Interestingly, the immunological synapse disseminates before division even though the T cells retain contact with the APC. The cue from the APC is translated into polarization of cell fate determinants via the polarity network of the Par3 and Scribble complexes, and orientation of the mitotic spindle during division is orchestrated by the partner of inscuteable/G protein complex. These findings suggest that T cells have selectively adapted a number of evolutionarily conserved mechanisms to generate diversity through asymmetric cell division.


Subject(s)
Antigen Presentation/immunology , Cell Division/immunology , Conserved Sequence/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Adhesion/immunology , Cell Polarity/immunology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocyte Subsets/metabolism
14.
Clin Dev Immunol ; 2012: 417485, 2012.
Article in English | MEDLINE | ID: mdl-22461835

ABSTRACT

Polarity refers to the asymmetric distribution of different cellular components within a cell and is central to many cell functions. In T-cells, polarity regulates the activation, migration, and effector function of cytotoxic T-cells (CTLs) during an immune response. The regulation of asymmetric cell division by polarity proteins may also dictate CTL effector and memory differentiation following antigen presentation. Small GTPases, along with their associated polarity and adaptor proteins, are critical for mediating the polarity changes necessary for T-cell activation and function, and in turn, are regulated by guanine exchange factors (GEFS) and GTPase activating proteins (GAPS). For example, a novel GEF, dedicator of cytokinesis 8 (DOCK8) was recently identified as a regulator of immune cell function and mutations in DOCK8 have been detected in patients with severe combined immunodeficiency. Both B and T-cells from DOCK8 mutant mice form defective immunological synapses and have abnormal functions, in addition to impaired immune memory development. This paper will discuss the interplay between polarity proteins and GTPases, and their role in T-cell function.


Subject(s)
Cell Polarity/immunology , Guanine Nucleotide Exchange Factors/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation/immunology , Cell Division/immunology , Cell Movement/immunology , Guanine Nucleotide Exchange Factors/genetics , Humans , Immunological Synapses/metabolism , Mice , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism
15.
Front Immunol ; 13: 931630, 2022.
Article in English | MEDLINE | ID: mdl-35874669

ABSTRACT

Cytotoxic lymphocytes are essential for anti-tumor immunity, and for effective responses to cancer immunotherapy. Natural killer cell granule protein 7 (NKG7) is expressed at high levels in cytotoxic lymphocytes infiltrating tumors from patients treated with immunotherapy, but until recently, the role of this protein in cytotoxic lymphocyte function was largely unknown. Unexpectedly, we found that highly CD8+ T cell-immunogenic murine colon carcinoma (MC38-OVA) tumors grew at an equal rate in Nkg7+/+ and Nkg7-/- littermate mice, suggesting NKG7 may not be necessary for effective CD8+ T cell anti-tumor activity. Mechanistically, we found that deletion of NKG7 reduces the ability of CD8+ T cells to degranulate and kill target cells in vitro. However, as a result of inefficient cytotoxic activity, NKG7 deficient T cells form a prolonged immune synapse with tumor cells, resulting in increased secretion of inflammatory cytokines, including tumor necrosis factor alpha (TNF). By deleting the TNF receptor, TNFR1, from MC38-OVA tumors, we demonstrate that this hyper-secretion of TNF compensates for reduced synapse-mediated cytotoxic activity against MC38-OVA tumors in vivo, via increased TNF-mediated tumor cell death. Taken together, our results demonstrate that NKG7 enhances CD8+ T cell immune synapse efficiency, which may serve as a mechanism to accelerate direct cytotoxicity and limit potentially harmful inflammatory responses.


Subject(s)
CD8-Positive T-Lymphocytes , Immunological Synapses , Membrane Proteins , Neoplasms , Animals , Immunotherapy/methods , Inflammation/metabolism , Membrane Proteins/metabolism , Mice , Neoplasms/therapy , Tumor Necrosis Factor-alpha/metabolism
16.
Cancer Immunol Res ; 10(1): 87-107, 2022 01.
Article in English | MEDLINE | ID: mdl-34782346

ABSTRACT

Targeting chromatin binding proteins and modifying enzymes can concomitantly affect tumor cell proliferation and survival, as well as enhance antitumor immunity and augment cancer immunotherapies. By screening a small-molecule library of epigenetics-based therapeutics, BET (bromo- and extra-terminal domain) inhibitors (BETi) were identified as agents that sensitize tumor cells to the antitumor activity of CD8+ T cells. BETi modulated tumor cells to be sensitized to the cytotoxic effects of the proinflammatory cytokine TNF. By preventing the recruitment of BRD4 to p65-bound cis-regulatory elements, BETi suppressed the induction of inflammatory gene expression, including the key NF-κB target genes BIRC2 (cIAP1) and BIRC3 (cIAP2). Disruption of prosurvival NF-κB signaling by BETi led to unrestrained TNF-mediated activation of the extrinsic apoptotic cascade and tumor cell death. Administration of BETi in combination with T-cell bispecific antibodies (TCB) or immune-checkpoint blockade increased bystander killing of tumor cells and enhanced tumor growth inhibition in vivo in a TNF-dependent manner. This novel epigenetic mechanism of immunomodulation may guide future use of BETi as adjuvants for immune-oncology agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
17.
Immunol Cell Biol ; 89(4): 549-57, 2011 May.
Article in English | MEDLINE | ID: mdl-20956985

ABSTRACT

The movement of proteins within cells can provide dynamic indications of cell signaling and cell polarity, but methods are needed to track and quantify subcellular protein movement within tissue environments. Here we present a semiautomated approach to quantify subcellular protein location for hundreds of migrating cells within intact living tissue using retrovirally expressed fluorescent fusion proteins and time-lapse two-photon microscopy of intact thymic lobes. We have validated the method using GFP-PKCζ, a marker for cell polarity, and LAT-GFP, a marker for T-cell receptor signaling, and have related the asymmetric distribution of these proteins to the direction and speed of cell migration. These approaches could be readily adapted to other fluorescent fusion proteins, tissues and biological questions.


Subject(s)
Green Fluorescent Proteins/metabolism , Intracellular Space/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Cell Movement/physiology , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Protein Transport , Recombinant Fusion Proteins/genetics , Thymus Gland/metabolism
18.
Front Immunol ; 12: 661737, 2021.
Article in English | MEDLINE | ID: mdl-34025662

ABSTRACT

The recent advent of targeted and immune-based therapies has revolutionized the treatment of melanoma and transformed outcomes for patients with metastatic disease. The majority of patients develop resistance to the current standard-of-care targeted therapy, dual BRAF and MEK inhibition, prompting evaluation of a new combination incorporating a CDK4/6 inhibitor. Based on promising preclinical data, combined BRAF, MEK and CDK4/6 inhibition has recently entered clinical trials for the treatment of BRAFV600 melanoma. Interestingly, while BRAF- and MEK-targeted therapy was initially developed on the basis of potent tumor-intrinsic effects, it was later discovered to have significant immune-potentiating activity. Recent studies have also identified immune-related impacts of CDK4/6 inhibition, though these are less well defined and can be both immune-potentiating and immune-inhibitory. BRAFV600 melanoma patients are also eligible to receive immunotherapy, specifically checkpoint inhibitors against PD-1 and CTLA-4. The immunomodulatory activity of BRAF/MEK-targeted therapies has prompted interest in combination therapies incorporating these with immune checkpoint inhibitors, however recent clinical trials investigating this approach have produced variable results. Here, we summarize the immunomodulatory effects of BRAF, MEK and CDK4/6 inhibitors, shedding light on the prospective utility of this combination alone and in conjunction with immune checkpoint blockade. Understanding the mechanisms that underpin the clinical efficacy of these available therapies is a critical step forward in optimizing novel combination and scheduling approaches to combat melanoma and improve patient outcomes.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunomodulation , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Clinical Trials as Topic , Humans , Melanoma/immunology , Molecular Targeted Therapy , Skin Neoplasms/drug therapy
19.
Sci Adv ; 7(8)2021 02.
Article in English | MEDLINE | ID: mdl-33608275

ABSTRACT

Multimodal single-cell RNA sequencing enables the precise mapping of transcriptional and phenotypic features of cellular differentiation states but does not allow for simultaneous integration of critical posttranslational modification data. Here, we describe SUrface-protein Glycan And RNA-seq (SUGAR-seq), a method that enables detection and analysis of N-linked glycosylation, extracellular epitopes, and the transcriptome at the single-cell level. Integrated SUGAR-seq and glycoproteome analysis identified tumor-infiltrating T cells with unique surface glycan properties that report their epigenetic and functional state.

20.
Cancer Immunol Res ; 9(2): 136-146, 2021 02.
Article in English | MEDLINE | ID: mdl-33303574

ABSTRACT

Combined inhibition of BRAF, MEK, and CDK4/6 is currently under evaluation in clinical trials for patients with melanoma harboring a BRAFV600 mutation. While this triple therapy has potent tumor-intrinsic effects, the impact of this combination on antitumor immunity remains unexplored. Here, using a syngeneic BrafV600ECdkn2a-/-Pten-/- melanoma model, we demonstrated that triple therapy promoted durable tumor control through tumor-intrinsic mechanisms and promoted immunogenic cell death and T-cell infiltration. Despite this, tumors treated with triple therapy were unresponsive to immune checkpoint blockade (ICB). Flow cytometric and single-cell RNA sequencing analyses of tumor-infiltrating immune populations revealed that triple therapy markedly depleted proinflammatory macrophages and cross-priming CD103+ dendritic cells, the absence of which correlated with poor overall survival and clinical responses to ICB in patients with melanoma. Indeed, immune populations isolated from tumors of mice treated with triple therapy failed to stimulate T-cell responses ex vivo While combined BRAF, MEK, and CDK4/6 inhibition demonstrates favorable tumor-intrinsic activity, these data suggest that collateral effects on tumor-infiltrating myeloid populations may impact antitumor immunity. These findings have important implications for the design of combination strategies and clinical trials that incorporate BRAF, MEK, and CDK4/6 inhibition with immunotherapy for the treatment of patients with melanoma.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Immunotherapy/methods , Melanoma/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cyclin-Dependent Kinase 4/immunology , Male , Melanoma/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/immunology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL