Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Commun Biol ; 6(1): 447, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37185301

RESUMO

Efficacy of immune checkpoint inhibitors in cancers can be limited by CD8 T cell dysfunction or HLA-I down-regulation. Tumor control mechanisms independent of CD8/HLA-I axis would overcome these limitations. Here, we report potent CD4 T cell-mediated tumor regression and memory responses in humanized immune system (HIS) mice implanted with HT-29 colorectal tumors. The regressing tumors showed increased CD4 cytotoxic T lymphocyte (CTL) infiltration and enhanced tumor HLA-II expression compared to progressing tumors. The intratumoral CD4 T cell subset associated with tumor regression expressed multiple cytotoxic markers and exhibited clonal expansion. Notably, tumor control was abrogated by depletion of CD4 but not CD8 T cells. CD4 T cells derived from tumor-regressing mice exhibited HLA-II-dependent and tumor-specific killing ex vivo. Taken together, our study demonstrates a critical role of human CD4 CTLs in mediating tumor clearance independent of CD8 T cells and provides a platform to study human anti-tumor immunity in vivo.


Assuntos
Neoplasias , Linfócitos T Citotóxicos , Humanos , Camundongos , Animais , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Neoplasias/metabolismo
2.
Sci Immunol ; 6(66): eabj4026, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34919442

RESUMO

Despite the enormous promise of T cell therapies, the isolation and study of human T cell receptors (TCRs) of dedicated specificity remains a major challenge. To overcome this limitation, we generated mice with a genetically humanized system of T cell immunity. We used VelociGene technology to replace the murine TCRαß variable regions, along with regions encoding the extracellular domains of co-receptors CD4 and CD8, and major histocompatibility complex (MHC) class I and II, with corresponding human sequences. The resulting "VelociT" mice have normal myeloid and lymphoid immune cell populations, including thymic and peripheral αß T cell subsets comparable with wild-type mice. VelociT mice expressed a diverse TCR repertoire, mounted functional T cell responses to lymphocytic choriomeningitis virus infection, and could develop experimental autoimmune encephalomyelitis. Immunization of VelociT mice with human tumor-associated peptide antigens generated robust, antigen-specific responses and led to identification of a TCR against tumor antigen New York esophageal squamous cell carcinoma-1 with potent antitumor activity. These studies demonstrate that VelociT mice mount clinically relevant T cell responses to both MHC-I­ and MHC-II­restricted antigens, providing a powerful new model for analyzing T cell function in human disease. Moreover, VelociT mice are a new platform for de novo discovery of therapeutic human TCRs.


Assuntos
Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T/imunologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T alfa-beta/genética
3.
Sci Immunol ; 5(54)2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33443027

RESUMO

Although T cell checkpoint inhibitors have transformed the treatment of cancer, the molecular determinants of tumor cell sensitivity to T cell-mediated killing need further elucidation. Here, we describe a mouse genome-scale CRISPR knockout screen that identifies tumor cell TNFα signaling as an important component of T cell-induced apoptosis, with NF-κB signaling and autophagy as major protective mechanisms. Knockout of individual autophagy genes sensitized tumor cells to killing by T cells that were activated via specific TCR or by a CD3 bispecific antibody. Conversely, inhibition of mTOR signaling, which results in increased autophagic activity, protected tumor cells from T cell killing. Autophagy functions at a relatively early step in the TNFα signaling pathway, limiting FADD-dependent caspase-8 activation. Genetic inactivation of tumor cell autophagy enhanced the efficacy of immune checkpoint blockade in mouse tumor models. Thus, targeting the protective autophagy pathway might sensitize tumors to T cell-engaging immunotherapies in the clinic.


Assuntos
Apoptose , Autofagia , Citotoxicidade Imunológica , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Caspase 8/metabolismo , Linhagem Celular Tumoral , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Edição de Genes , Técnicas de Silenciamento de Genes , Camundongos , Transdução de Sinais , Fator de Necrose Tumoral alfa/farmacologia
4.
Sci Rep ; 9(1): 20068, 2019 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-31882897

RESUMO

Bispecific antibodies (bsAb) that bridge tumor cells and CD3-positive effector T cells are being developed against many tumor cell targets. While tumor cell factors other than target expression level appear to play a role in determining the efficacy of CD3 bsAb, the identity of such factors remains largely unknown. Using a co-culture system of primary human T cells and B lymphoma cell lines, we demonstrate a range of sensitivities to CD20xCD3 bsAb that is independent of CD20 surface expression. To identify genes that modulate tumor cell sensitivity to CD3 bsAb, we employed a genome-scale CRISPR activation screen in a CD20xCD3-sensitive human B lymphoma cell line. Among the most highly enriched sgRNAs were those targeting genes with predicted effects on cell-cell adhesion, including sialophorin (SPN). Increased expression of SPN impeded tumor cell clustering with T cells, thereby limiting CD3 bsAb-mediated tumor cell lysis. This inhibitory effect of SPN appeared to be dependent on sialylated core 2 O-glycosylation of the protein. While SPN is not endogenously expressed in the majority of B cell lymphomas, it is highly expressed in acute myeloid leukemia. CRISPR-mediated SPN knockout in AML cell lines facilitated T cell-tumor cell clustering and enhanced CD3 bsAb-mediated AML cell lysis. In sum, our data establish that the cell cross-linking mechanism of CD3 bsAb is susceptible to subversion by anti-adhesive molecules expressed on the tumor cell surface. Further evaluation of anti-adhesive pathways may provide novel biomarkers of clinical response and enable the development of effective combination regimens for this promising therapeutic class.


Assuntos
Anticorpos Biespecíficos/imunologia , Complexo CD3/imunologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Genoma Humano , Neoplasias/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias/imunologia
5.
J Autoimmun ; 100: 62-74, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30879886

RESUMO

Cytokine storm syndrome (CSS) is a life-threatening condition characterized by excessive activation of T cells and uncontrolled inflammation, mostly described in patients with familial hemophagocytic lymphohistiocytosis and certain systemic auto-inflammatory diseases, such as systemic juvenile idiopathic arthritis (sJIA). Defects in T cell cytotoxicity as a mechanism for uncontrolled inflammation following viral infections fail to represent the whole spectrum of CSS. Evidence implicates dysregulated innate immune responses, especially activation of monocytes and macrophages, in patients with CSS. However, the direct contribution of monocytes/macrophages to CSS development and the signaling pathways involved in their activation have not been formally investigated. We find that depletion of monocytes/macrophages during early stages of CSS development, by clodronate-liposomes or neutralizing anti-CSF1 antibody, reduces mortality and inflammatory cytokine levels in two CSS mouse models, one dependent on T cells and the second induced by repeated TLR9 stimulation. We further demonstrate that activation of Plcγ2 in myeloid cells controls CSS development by driving macrophage pro-inflammatory responses. Intriguingly, the Plcγ2 downstream effector Tmem178, a negative modulator of calcium levels, acts in a negative feedback loop to restrain inflammatory cytokine production. Genetic deletion of Tmem178 leads to pro-inflammatory macrophage polarization in vitro and more severe CSS in vivo. Importantly, Tmem178 levels are reduced in macrophages from mice with CSS and after exposure to plasma from sJIA patients with active disease. Our data identify a novel Plcγ2/Tmem178 axis as a modulator of inflammatory cytokine production by monocytes/macrophages. We also find that loss of Tmem178 accentuates the pro-inflammatory responses in CSS.


Assuntos
Síndrome de Ativação Macrofágica/imunologia , Macrófagos/imunologia , Proteínas de Membrana/imunologia , Monócitos/imunologia , Fosfolipase C gama/imunologia , Transdução de Sinais/imunologia , Animais , Humanos , Síndrome de Ativação Macrofágica/genética , Síndrome de Ativação Macrofágica/patologia , Macrófagos/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Monócitos/patologia , Fosfolipase C gama/genética , Transdução de Sinais/genética
6.
J Bone Miner Res ; 30(10): 1852-63, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25891971

RESUMO

Increased diacylglycerol (DAG) levels are observed in numerous pathologies, including conditions associated with bone loss. However, the effects of DAG accumulation on the skeleton have never been directly examined. Because DAG is strictly controlled by tissue-specific diacylglycerol kinases (DGKs), we sought to examine the biological consequences of DAG accumulation on bone homeostasis by genetic deletion of DGKζ, a highly expressed DGK isoform in osteoclasts (OCs). Strikingly, DGKζ(-/-) mice are osteoporotic because of a marked increase in OC numbers. In vitro, DGKζ(-/-) bone marrow macrophages (BMMs) form more numerous, larger, and highly resorptive OCs. Surprisingly, although increased DAG levels do not alter receptor activator of NF-κB (RANK)/RANK ligand (RANKL) osteoclastogenic pathway, DGKζ deficiency increases responsiveness to the proliferative and pro-survival cytokine macrophage colony-stimulating factor (M-CSF). We find that M-CSF is responsible for increased DGKζ(-/-) OC differentiation by promoting higher expression of the transcription factor c-Fos, and c-Fos knockdown in DGKζ(-/-) cultures dose-dependently reduces OC differentiation. Using a c-Fos luciferase reporter assay lacking the TRE responsive element, we also demonstrate that M-CSF induces optimal c-Fos expression through DAG production. Finally, to demonstrate the importance of the M-CSF/DGKζ/DAG axis on regulation of c-Fos during osteoclastogenesis, we turned to PLCγ2(+/-) BMMs, which have reduced DAG levels and form fewer OCs because of impaired expression of the master regulator of osteoclastogenesis NFATc1 and c-Fos. Strikingly, genetic deletion of DGKζ in PLCγ2(+/-) mice rescues OC formation and normalizes c-Fos levels without altering NFATc1 expression. To our knowledge, this is the first report implicating M-CSF/DGKζ/DAG axis as a critical regulator of bone homeostasis via its actions on OC differentiation and c-Fos expression.


Assuntos
Diacilglicerol Quinase/metabolismo , Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas c-fos/biossíntese , Animais , Diacilglicerol Quinase/genética , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Fosfolipase C gama/genética , Fosfolipase C gama/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Ligante RANK/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Elementos de Resposta
7.
J Cell Biochem ; 116(11): 2476-83, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25864714

RESUMO

The interaction between Receptor Activator of NF-κB Ligand (RANKL) and its receptor RANK is essential for the differentiation and bone resorbing capacity of the osteoclast. Osteoprotegerin (OPG), a soluble homodimer, acts as a decoy receptor for RANKL and thus inhibits osteoclastogenesis. An imbalance in the RANKL/RANK/OPG axis, with decreased OPG and/or increased RANKL, is associated with diseases that favor bone loss, including osteoporosis. Recently, we established a yeast surface display system and screened libraries of randomly mutated RANKL proteins to identify mutations that abolish binding to OPG while preserving recognition of RANK. These efforts yielded several RANKL variants possessing substantially higher affinity for RANK compared to their wild-type (WT) counterpart. Using recombinant RANKL mutant proteins, we find those with increased affinity for RANK produce more robust signaling in osteoclast lineage cells and have greater osteoclastogenic potential. Our results are the first to document gain of function RANKL mutations. They indicate that the physiological RANKL/RANK interaction is not optimized for maximal signaling and function, perhaps reflecting the need to maintain receptor specificity within the tumor necrosis factor superfamily (TNFSF). Instead, we find, a biphasic relationship exists between RANKL/RANK affinity and osteoclastogenic capacity. In our panel of RANKL variants, this relationship is driven entirely by manipulation of the kinetic off-rate. Our structure-based and yeast surface display-derived insights into manipulating this critical signaling axis may aid in the design of novel anti-resorptive therapies as well as provide a paradigm for design of other receptor-specific TNF superfamily ligand variants.


Assuntos
Macrófagos/citologia , Osteoclastos/citologia , Ligante RANK/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Cinética , Macrófagos/metabolismo , Camundongos , Mutação , Osteoclastos/metabolismo , Osteoprotegerina/metabolismo , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
8.
Sci Signal ; 7(339): ra80, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25140055

RESUMO

Signaling by receptor activator of nuclear factor κB (RANK) in response to its ligand RANKL, which is a member of the tumor necrosis factor (TNF) superfamily of cytokines, stimulates osteoclast formation and bone resorption. Thus, this ligand-receptor pair is a therapeutic target for various disorders, such as osteoporosis and metastasis of cancer to bone. RANKL exists as a physiological homotrimer, with each monomer recognizing a single molecule of RANK or the decoy receptor osteoprotegerin (OPG), which inhibits osteoclastogenesis. We engineered a RANKL protein in which all three monomers of RANKL were encoded as a single polypeptide chain, which enabled us to independently control receptor binding at each binding interface. To generate an effective RANK inhibitor, we used an unbiased forward genetic approach to identify mutations in RANKL that had a 500-fold increased affinity for RANK but had decreased affinity for the decoy receptor OPG. Incorporating mutations that blocked receptor binding into this high-affinity RANKL variant generated a mutant RANKL that completely inhibited wild-type RANKL-induced osteoclastogenesis in vitro and bone resorption in mice. Our approach may be generalized to enable the inhibition of other TNF receptor signaling systems, which are implicated in a wide range of pathological conditions.


Assuntos
Substituição de Aminoácidos , Mutação de Sentido Incorreto , Osteoclastos/metabolismo , Multimerização Proteica/genética , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Animais , Reabsorção Óssea/genética , Humanos , Camundongos , Osteoprotegerina/antagonistas & inibidores , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligante RANK/antagonistas & inibidores , Ligante RANK/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/genética
9.
J Biol Chem ; 288(47): 33634-33641, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24081142

RESUMO

Phospholipase C γ2 (PLCγ2) is a critical regulator of innate immune cells and osteoclasts (OCs) during inflammatory arthritis. Both the catalytic domain and the adaptor motifs of PLCγ2 are required for OC formation and function. Due to the high homology between the catalytic domains of PLCγ2 and the ubiquitously expressed PLCγ1, molecules encompassing the adaptor motifs of PLCγ2 were designed to test the hypothesis that uncoupling the adaptor and catalytic functions of PLCγ2 could specifically inhibit osteoclastogenesis and bone erosion. Wild-type (WT) bone marrow macrophages (BMM) that overexpress the tandem Src homology 2 (SH2) domains of PLCγ2 (SH2(N+C)) failed to form mature OCs and resorb bone in vitro. Activation of the receptor activator of NF-κB (RANK) signaling pathway, which is critical for OC development, was impaired in cells expressing SH2(N+C). Arrest in OC differentiation was evidenced by a reduction of p38 and Iκ-Bα phosphorylation as well as decreased NFATc1 and c-Fos/c-Jun levels. Consistent with our hypothesis, SH2(N+C) abrogated formation of the RANK-Gab2 complex, which mediates NF-κB and AP-1 activation following RANK ligand (RANKL) stimulation. Furthermore, the ability of SH2(N+C) to prevent inflammatory osteolysis was examined in vivo following RANKL or LPS injections over the calvaria. Both models induced osteolysis in the control group, whereas the SH2(N+C)-treated cohort was largely protected from bone erosion. Collectively, these data indicate that inflammatory osteolysis can be abrogated by treatment with a molecule composed of the tandem SH2 domains of PLCγ2.


Assuntos
Células da Medula Óssea/enzimologia , Osteoclastos/enzimologia , Osteólise/enzimologia , Fosfolipase C gama/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Células da Medula Óssea/patologia , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Camundongos , Inibidor de NF-kappaB alfa , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/patologia , Osteólise/tratamento farmacológico , Osteólise/genética , Osteólise/patologia , Fosfolipase C gama/antagonistas & inibidores , Fosfolipase C gama/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Domínios de Homologia de src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA