Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Immunol Cell Biol ; 92(6): 475-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24732076

ABSTRACT

Antibodies targeting checkpoint inhibitors or co-stimulatory receptors on T cells have shown significant antitumor efficacy in preclinical and clinical studies. In mouse tumor models, engagement of activating Fcγ receptor (FcγR)-expressing immune cells was recently shown to be required for the tumoricidal activity of antibodies recognizing the tumor necrosis factor superfamily receptor (TNFR) GITR (CD357) and CTLA-4 (CD152). In particular, activating FcγRs facilitated the selective elimination of intratumoral T-cell populations. However, it remains unclear whether FcγRs contribute to the antitumor efficacy of other immunomodulatory antibodies. Here, we explored the mechanism of antitumor activity mediated by an agonistic antibody (clone OX86) to the co-stimulatory TNFR OX40 (CD134). OX40 was highly expressed by intratumoral T cells, particularly those of the FoxP3(+) regulatory T-cell (Treg) lineage. OX86 administration resulted in the depletion of intratumoral regulatory T cells in an activating FcγR-dependent manner, which correlated with tumor regression. Together with previous data from our group and others, these findings support a mechanism whereby antibodies targeting antigens highly expressed by intratumoral T cells can mediate their elimination by FcγR-expressing immune cells, and facilitate subsequent antitumor immunity.


Subject(s)
Neoplasms, Experimental/immunology , Receptors, IgG/immunology , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Glucocorticoid-Induced TNFR-Related Protein/genetics , Glucocorticoid-Induced TNFR-Related Protein/immunology , Lymphocyte Depletion , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Receptors, IgG/genetics , Receptors, OX40/genetics , T-Lymphocytes, Regulatory/pathology
2.
J Virol ; 87(16): 9030-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23760237

ABSTRACT

Cellular cytidine deaminases from the APOBEC3 family are potent restriction factors that are able to block the replication of retroviruses. Consequently, retroviruses have evolved a variety of different mechanisms to counteract inhibition by APOBEC3 proteins. Lentiviruses such as human immunodeficiency virus (HIV) express Vif, which interferes with APOBEC3 proteins by targeting these restriction factors for proteasomal degradation, hence blocking their ability to access the reverse transcriptase complex in the virions. Other retroviruses use less-well-characterized mechanisms to escape the APOBEC3-mediated cellular defense. Here we show that the prototype foamy virus Bet protein can protect foamy viruses and an unrelated simian immunodeficiency virus against human APOBEC3G (A3G). In our system, Bet binds to A3G and prevents its encapsidation without inducing its degradation. Bet failed to coimmunoprecipitate with A3G mutants unable to form homodimers and dramatically reduced the recovery of A3G proteins from soluble cytoplasmic cell fractions. The Bet-A3G interaction is probably a direct binding interaction and seems to be independent of RNA. Together, these data suggest a novel model whereby Bet uses two possibly complementary mechanisms to counteract A3G: (i) Bet prevents encapsidation of A3G by blocking A3G dimerization, and (ii) Bet sequesters A3G in immobile complexes, impairing its ability to interact with nascent virions.


Subject(s)
Cytidine Deaminase/antagonists & inhibitors , Host-Pathogen Interactions , Protein Multimerization , Retroviridae Proteins/metabolism , Simian foamy virus/immunology , Virulence Factors/metabolism , APOBEC-3G Deaminase , Cell Line , Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , Humans , Solubility
3.
J Hematol Oncol ; 16(1): 108, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37880715

ABSTRACT

T cell differentiation is a highly regulated, multi-step process necessary for the progressive establishment of effector functions, immunological memory, and long-term control of pathogens. In response to strong stimulation, as seen in severe or chronic infections or cancer, T cells acquire a state of hypo-responsiveness known as exhaustion, limiting their effector function. Recent advances in autologous chimeric antigen receptor (CAR)-T cell therapies have revolutionized the treatment of hematologic malignancies by taking advantage of the basic principles of T cell biology to engineer products that promote long-lasting T cell response. However, many patients' malignancies remain unresponsive to treatment or are prone to recur. Discoveries in T cell biology, including the identification of key regulators of differentiation and exhaustion, offer novel opportunities to have a durable impact on the fate of CAR-T cells after infusion. Such next-generation CAR-T cell therapies and their clinical implementation may result in the next leap forward in cancer treatment for selected patients. In this context, this review summarizes the foundational principles of T cell differentiation and exhaustion and describes how they can be utilized and targeted to further improve the design and efficacy of CAR-T cell therapies.


Subject(s)
Hematologic Neoplasms , Receptors, Chimeric Antigen , Humans , Cell Differentiation , Hematologic Neoplasms/therapy , Immunologic Memory , Immunotherapy, Adoptive , Cell- and Tissue-Based Therapy
4.
J Virol ; 85(4): 1765-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21123384

ABSTRACT

Members of the human APOBEC3 family of editing enzymes can inhibit various mobile genetic elements. APOBEC3A (A3A) can block the retrotransposon LINE-1 and the parvovirus adeno-associated virus type 2 (AAV-2) but does not inhibit retroviruses. In contrast, APOBEC3G (A3G) can block retroviruses but has only limited effects on AAV-2 or LINE-1. What dictates this differential target specificity remains largely undefined. Here, we modeled the structure of A3A based on its homology with the C-terminal domain of A3G and further compared the sequence of human A3A to those of 11 nonhuman primate orthologues. We then used these data to perform a mutational analysis of A3A, examining its ability to restrict LINE-1, AAV-2, and foreign plasmid DNA and to edit a single-stranded DNA substrate. The results revealed an essential functional role for the predicted single-stranded DNA-docking groove located around the A3A catalytic site. Within this region, amino acid differences between A3A and A3G are predicted to affect the shape of the polynucleotide-binding groove. Correspondingly, transferring some of these A3A residues to A3G endows the latter protein with the ability to block LINE-1 and AAV-2. These results suggest that the target specificity of APOBEC3 family members is partly defined by structural features influencing their interaction with polynucleotide substrates.


Subject(s)
Cytidine Deaminase/chemistry , Cytidine Deaminase/metabolism , DNA, Single-Stranded/physiology , Dependovirus/physiology , Long Interspersed Nucleotide Elements/physiology , Polynucleotides/metabolism , Proteins/chemistry , Proteins/metabolism , APOBEC-3G Deaminase , Animals , Base Sequence , Cytidine Deaminase/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Dependovirus/genetics , Dependovirus/metabolism , HEK293 Cells , Haplorhini , HeLa Cells , Humans , Long Interspersed Nucleotide Elements/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids , Polynucleotides/chemistry , Proteins/genetics , Retroelements/genetics , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Structure-Activity Relationship , Substrate Specificity
5.
J Immunother Cancer ; 9(10)2021 10.
Article in English | MEDLINE | ID: mdl-34706886

ABSTRACT

BACKGROUND: Co-stimulatory signals regulate the expansion, persistence, and function of chimeric antigen receptor (CAR) T cells. Most studies have focused on the co-stimulatory domains CD28 or 4-1BB. CAR T cell persistence is enhanced by 4-1BB co-stimulation leading to nuclear factor kappa B (NF-κB) signaling, while resistance to exhaustion is enhanced by mutations of the CD28 co-stimulatory domain. METHODS: We hypothesized that a third-generation CAR containing 4-1BB and CD28 with only PYAP signaling motif (mut06) would provide beneficial aspects of both. We designed CD19-specific CAR T cells with either 4-1BB or mut06 together with the combination of both and evaluated their immune-phenotype, cytokine secretion, real-time cytotoxic ability and polyfunctionality against CD19-expressing cells. We analyzed lymphocyte-specific protein tyrosine kinase (LCK) recruitment by the different constructs by immunoblotting. We further determined their ability to control growth of Raji cells in NOD scid gamma (NSG) mice. We also engineered bi-specific CARs against CD20/CD19 combining 4-1BB and mut06 and performed repeated in vitro antigenic stimulation experiments to evaluate their expansion, memory phenotype and phenotypic (PD1+CD39+) and functional exhaustion. Bi-specific CAR T cells were transferred into Raji or Nalm6-bearing mice to study their ability to eradicate CD20/CD19-expressing tumors. RESULTS: Co-stimulatory domains combining 4-1BB and mut06 confers CAR T cells with an increased central memory phenotype, expansion, and LCK recruitment to the CAR. This enhanced function was dependent on the positioning of the two co-stimulatory domains. A bi-specific CAR targeting CD20/CD19, incorporating 4-1BB and mut06 co-stimulation, showed enhanced antigen-dependent in vitro expansion with lower exhaustion-associated markers. Bi-specific CAR T cells exhibited improved in vivo antitumor activity with increased persistence and decreased exhaustion. CONCLUSION: These results demonstrate that co-stimulation combining 4-1BB with an optimized form of CD28 is a valid approach to optimize CAR T cell function. Cells with both mono-specific and bi-specific versions of this design showed enhanced in vitro and in vivo features such as expansion, persistence and resistance to exhaustion. Our observations validate the approach and justify clinical studies to test the efficacy and safety of this CAR in patients.


Subject(s)
CD28 Antigens/metabolism , Cell Engineering/methods , Neoplasms/genetics , Receptors, Chimeric Antigen/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Animals , Female , Humans , Male , Mice
6.
J Virol ; 83(23): 12611-21, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19776130

ABSTRACT

Retroelements are important evolutionary forces but can be deleterious if left uncontrolled. Members of the human APOBEC3 family of cytidine deaminases can inhibit a wide range of endogenous, as well as exogenous, retroelements. These enzymes are structurally organized in one or two domains comprising a zinc-coordinating motif. APOBEC3G contains two such domains, only the C terminal of which is endowed with editing activity, while its N-terminal counterpart binds RNA, promotes homo-oligomerization, and is necessary for packaging into human immunodeficiency virus type 1 (HIV-1) virions. Here, we performed a large-scale mutagenesis-based analysis of the APOBEC3G N terminus, testing mutants for (i) inhibition of vif-defective HIV-1 infection and Alu retrotransposition, (ii) RNA binding, and (iii) oligomerization. Furthermore, in the absence of structural information on this domain, we used homology modeling to examine the positions of functionally important residues and of residues found to be under positive selection by phylogenetic analyses of primate APOBEC3G genes. Our results reveal the importance of a predicted RNA binding dimerization interface both for packaging into HIV-1 virions and inhibition of both HIV-1 infection and Alu transposition. We further found that the HIV-1-blocking activity of APOBEC3G N-terminal mutants defective for packaging can be almost entirely rescued if their virion incorporation is forced by fusion with Vpr, indicating that the corresponding region of APOBEC3G plays little role in other aspects of its action against this pathogen. Interestingly, residues forming the APOBEC3G dimer interface are highly conserved, contrasting with the rapid evolution of two neighboring surface-exposed amino acid patches, one targeted by the Vif protein of primate lentiviruses and the other of yet-undefined function.


Subject(s)
Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , HIV-1/immunology , APOBEC-3G Deaminase , Amino Acid Sequence , Animals , Conserved Sequence , Cytidine Deaminase/metabolism , Dimerization , Humans , Lentiviruses, Primate , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Primates , Protein Binding , Protein Structure, Tertiary , RNA, Viral/metabolism , Sequence Homology, Amino Acid
7.
J Exp Med ; 210(9): 1685-93, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23897982

ABSTRACT

Fc γ receptor (FcγR) coengagement can facilitate antibody-mediated receptor activation in target cells. In particular, agonistic antibodies that target tumor necrosis factor receptor (TNFR) family members have shown dependence on expression of the inhibitory FcγR, FcγRIIB. It remains unclear if engagement of FcγRIIB also extends to the activities of antibodies targeting immunoregulatory TNFRs expressed by T cells. We have explored the requirement for activating and inhibitory FcγRs for the antitumor effects of antibodies targeting the TNFR glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18; CD357) expressed on activated and regulatory T cells (T reg cells). We found that although FcγRIIB was dispensable for the in vivo efficacy of anti-GITR antibodies, in contrast, activating FcγRs were essential. Surprisingly, the dependence on activating FcγRs extended to an antibody targeting the non-TNFR receptor CTLA-4 (CD152) that acts as a negative regulator of T cell immunity. We define a common mechanism that correlated with tumor efficacy, whereby antibodies that coengaged activating FcγRs expressed by tumor-associated leukocytes facilitated the selective elimination of intratumoral T cell populations, particularly T reg cells. These findings may have broad implications for antibody engineering efforts aimed at enhancing the therapeutic activity of immunomodulatory antibodies.


Subject(s)
Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Immunologic Factors/pharmacology , Neoplasms/immunology , Receptors, IgG/metabolism , Animals , CTLA-4 Antigen/metabolism , Female , Glucocorticoid-Induced TNFR-Related Protein/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Neoplasms/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
8.
Mol Microbiol ; 65(5): 1218-28, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17651388

ABSTRACT

Peptide deformylase (PDF) catalyses the removal of the formyl group from the first methionine of nascent proteins. Type 1 PDFs are found in bacteria and have orthologues in most eukaryotes. Type 2 PDFs are restricted to bacteria. Type 3 enzymes are found in Archaea and trypanosomatids and have not been studied experimentally yet. Thus, TbPDF1 and TbPDF2, the two PDF orthologues of the parasitic protozoa Trypanosoma brucei, are of type 3. An experimental analysis of these enzymes shows that both are mitochondrially localized, but that only TbPDF1 is essential for normal growth. Recombinant TbPDF1 exhibits PDF activity with a substrate specificity identical to that of bacterial enzymes. Consistent with these results, TbPDF1 is required for oxidative but not for mitochondrial substrate-level phosphorylation. Ablation of TbPDF2, in contrast, does neither affect growth on standard medium nor oxidative phosphorylation. However, a reduced level of TbPDF2 slows down growth in a medium that selects for highly efficient oxidative phosphorylation. Furthermore, combined ablation of TbPDF1 and TbPDF2 results in an earlier growth arrest than is observed by downregulation of TbPDF1 alone. These results suggest that TbPDF2 is functionally linked to TbPDF1, and that it can influence the efficiency of oxidative phosphorylation.


Subject(s)
Amidohydrolases/metabolism , Isoenzymes/metabolism , Oxidative Phosphorylation , Trypanosoma brucei brucei/enzymology , Amidohydrolases/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Isoenzymes/genetics , Molecular Sequence Data , Open Reading Frames , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity
9.
J Biol Chem ; 281(47): 35742-6, 2006 Nov 24.
Article in English | MEDLINE | ID: mdl-16997916

ABSTRACT

The KRAB transcriptional repressor domain, commonly found in zinc finger proteins, acts by inducing the formation of heterochromatin. We previously exploited this property to achieve drug-regulated transgenesis and knock down by combining doxycycline-controllable KRAB-containing fusion proteins and lentiviral vectors. Here, we asked whether KRAB-induced repression is widespread or limited to specific regions of the genome. For this, we transduced cells with a lentiviral vector expressing a target reporter and a KRAB-containing transcriptional repressor from a bicistronic mRNA. We found that approximately 1.4% of the resulting proviruses escaped repression. However, this phenotype could be reverted by expressing the KRAB-containing protein in trans. Accordingly, the irrepressible proviruses all contained, in the DNA sequence encoding the KRAB-containing effector or its upstream internal ribosomal entry site, mutations or deletions likely resulting from errors or recombination during reverse transcription. These results indicate that KRAB-induced transcriptional repression is robust and active over a variety of genomic contexts that include at least the wide range of sites targeted by lentiviral integration.


Subject(s)
DNA-Binding Proteins/physiology , Gene Silencing , Lentivirus/genetics , Repressor Proteins/physiology , Transcription, Genetic , Binding Sites , Cell Line , Cell Separation , DNA-Binding Proteins/metabolism , Doxycycline/pharmacology , Genetic Vectors , Genome, Viral , Humans , Lentivirus/metabolism , Models, Genetic , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL