Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Immunity ; 54(12): 2772-2783.e5, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34788602

ABSTRACT

Humoral immunity is essential for protection against pathogens, emphasized by the prevention of 2-3 million deaths worldwide annually by childhood immunizations. Long-term protective immunity is dependent on the continual production of neutralizing antibodies by the subset of long-lived plasma cells (LLPCs). LLPCs are not intrinsically long-lived, but require interaction with LLPC niche stromal cells for survival. However, it remains unclear which and how these interactions sustain LLPC survival and long-term humoral immunity. We now have found that the immunosuppressive enzyme indoleamine 2,3- dioxygenase 1 (IDO1) is required to sustain antibody responses and LLPC survival. Activation of IDO1 occurs upon the engagement of CD80/CD86 on the niche dendritic cells by CD28 on LLPC. Kynurenine, the product of IDO1 catabolism, activates the aryl hydrocarbon receptor in LLPC, reinforcing CD28 expression and survival signaling. These findings expand the immune function of IDO1 and uncover a novel pathway for sustaining LLPC survival and humoral immunity.


Subject(s)
Dendritic Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Plasma Cells/immunology , Animals , Antibodies, Neutralizing/metabolism , B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Cell Self Renewal , Cell Survival , Cells, Cultured , Female , Immunity, Humoral , Immunologic Memory , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mice , Mice, Knockout
2.
J Immunol ; 212(3): 475-486, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38117752

ABSTRACT

Macrophages represent the most abundant immune component of the tumor microenvironment and often exhibit protumorigenic (M2-like) phenotypes that contribute to disease progression. Despite their generally accepted protumorigenic role, macrophages can also display tumoricidal (or M1-like) behavior, revealing that macrophages can be functionally reprogrammed, depending on the cues received within the tumor microenvironment. Moreover, such plasticity may be achieved by pharmacologic or biologic interventions. To that end, we previously demonstrated that a novel immunomodulator termed the "very small size particle" (VSSP) facilitates maturation of dendritic cells and differentiation of myeloid-derived suppressor cells to APCs with reduced suppressive activity in cancer models. VSSP was further shown to act in the bone marrow to drive the differentiation of progenitors toward monocytes, macrophages, and dendritic cells during emergency myelopoiesis. However, the underlying mechanisms for VSSP-driven alterations in myeloid differentiation and function remained unclear. In this study, in mouse models, we focused on macrophages and tested the hypothesis that VSSP drives macrophages toward M1-like functional states via IRF8- and PU.1-dependent mechanisms. We further hypothesized that such VSSP-mediated actions would be accompanied by enhanced antitumor responses. Overall, we showed that (1) VSSP drives naive or M2-derived macrophages to M1-like states, (2) the M1-like state induced by VSSP occurs via IRF8- and PU.1-dependent mechanisms, and (3) single-agent VSSP induces an antitumor response that is accompanied by alterations in the intratumoral myeloid compartment. These results provide a deeper mechanistic underpinning of VSSP and strengthen its use to drive M1-like responses in host defense, including cancer.


Subject(s)
Nanoparticles , Neoplasms , Mice , Animals , Gangliosides , Macrophages , Neoplasms/pathology , Phenotype , Interferon Regulatory Factors , Tumor Microenvironment
3.
Proc Natl Acad Sci U S A ; 117(38): 23730-23741, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32879009

ABSTRACT

Although plasmacytoid dendritic cells (pDCs) have been shown to play a critical role in generating viral immunity and promoting tolerance to suppress antitumor immunity, whether and how pDCs cross-prime CD8 T cells in vivo remain controversial. Using a pDC-targeted vaccine model to deliver antigens specifically to pDCs, we have demonstrated that pDC-targeted vaccination led to strong cross-priming and durable CD8 T cell immunity. Surprisingly, cross-presenting pDCs required conventional DCs (cDCs) to achieve cross-priming in vivo by transferring antigens to cDCs. Taking advantage of an in vitro system where only pDCs had access to antigens, we further demonstrated that cross-presenting pDCs were unable to efficiently prime CD8 T cells by themselves, but conferred antigen-naive cDCs the capability of cross-priming CD8 T cells by transferring antigens to cDCs. Although both cDC1s and cDC2s exhibited similar efficiency in acquiring antigens from pDCs, cDC1s but not cDC2s were required for cross-priming upon pDC-targeted vaccination, suggesting that cDC1s played a critical role in pDC-mediated cross-priming independent of their function in antigen presentation. Antigen transfer from pDCs to cDCs was mediated by previously unreported pDC-derived exosomes (pDCexos), that were also produced by pDCs under various conditions. Importantly, all these pDCexos primed naive antigen-specific CD8 T cells only in the presence of bystander cDCs, similarly to cross-presenting pDCs, thus identifying pDCexo-mediated antigen transfer to cDCs as a mechanism for pDCs to achieve cross-priming. In summary, our data suggest that pDCs employ a unique mechanism of pDCexo-mediated antigen transfer to cDCs for cross-priming.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cross-Priming/immunology , Dendritic Cells/metabolism , Exosomes/metabolism , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Dendritic Cells/immunology , Exosomes/immunology , Humans , Mice , Mice, Inbred C57BL
4.
J Immunol ; 199(10): 3700-3710, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29046346

ABSTRACT

The CD27-CD70 pathway is known to provide a costimulatory signal, with CD70 expressed on APCs and CD27 functions on T cells. Although CD70 is also expressed on activated T cells, it remains unclear how T cell-derived CD70 affects T cell function. Therefore, we have assessed the role of T cell-derived CD70 using adoptive-transfer models, including autoimmune inflammatory bowel disease and allogeneic graft-versus-host disease. Surprisingly, compared with wild-type T cells, CD70-/- T cells caused more severe inflammatory bowel disease and graft-versus-host disease and produced higher levels of inflammatory cytokines. Mechanistic analyses reveal that IFN-ƎĀ³ induces CD70 expression in T cells, and CD70 limits T cell expansion via a regulatory T cell-independent mechanism that involves caspase-dependent T cell apoptosis and upregulation of inhibitory immune checkpoint molecules. Notably, T cell-intrinsic CD70 signaling contributes, as least in part, to the inhibitory checkpoint function. Overall, our findings demonstrate for the first time, to our knowledge, that T cell-derived CD70 plays a novel immune checkpoint role in inhibiting inflammatory T cell responses. This study suggests that T cell-derived CD70 performs a critical negative feedback function to downregulate inflammatory T cell responses.


Subject(s)
CD27 Ligand/metabolism , Graft vs Host Disease/immunology , Inflammatory Bowel Diseases/immunology , Interferon-gamma/metabolism , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Apoptosis , CD27 Ligand/genetics , Caspases/metabolism , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Lymphocytes/transplantation , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
5.
Proc Natl Acad Sci U S A ; 112(9): 2823-8, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25730849

ABSTRACT

Recent studies have demonstrated that Ɵ-catenin in DCs serves as a key mediator in promoting both CD4(+) and CD8(+) T-cell tolerance, although how Ɵ-catenin exerts its functions remains incompletely understood. Here we report that activation of Ɵ-catenin in DCs inhibits cross-priming of CD8(+) T cells by up-regulating mTOR-dependent IL-10, suggesting blocking Ɵ-catenin/mTOR/IL-10 signaling as a viable approach to augment CD8(+) T-cell immunity. However, vaccination of DC-Ɵ-catenin(-/-) (CD11c-specific deletion of Ɵ-catenin) mice surprisingly failed to protect them against tumor challenge. Further studies revealed that DC-Ɵ-catenin(-/-) mice were deficient in generating CD8(+) T-cell immunity despite normal clonal expansion, likely due to impaired IL-10 production by Ɵ-catenin(-/-) DCs. Deletion of Ɵ-catenin in DCs or blocking IL-10 after clonal expansion similarly led to reduced CD8(+) T cells, suggesting that Ɵ-catenin in DCs plays a positive role in CD8(+) T-cell maintenance postclonal expansion through IL-10. Thus, our study has not only identified mTOR/IL-10 as a previously unidentified mechanism for Ɵ-catenin-dependent inhibition of cross-priming, but also uncovered an unexpected positive role that Ɵ-catenin plays in maintenance of CD8(+) T cells. Despite Ɵ-catenin's opposite functions in regulating CD8(+) T-cell responses, selectively blocking Ɵ-catenin with a pharmacological inhibitor during priming phase augmented DC vaccine-induced CD8(+) T-cell immunity and improved antitumor efficacy, suggesting manipulating Ɵ-catenin signaling as a feasible therapeutic strategy to improve DC vaccine efficacy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunity, Cellular , Interleukin-10/immunology , beta Catenin/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Dendritic Cells/pathology , Interleukin-10/genetics , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , beta Catenin/genetics
6.
J Immunol ; 194(10): 4717-28, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25833397

ABSTRACT

In health, long-lived plasma cells (LLPC) are essential for durable protective humoral immunity, and, conversely, in disease are a major source of pathogenic Abs in autoimmunity, graft rejection, and allergy. However, the molecular basis for their longevity is largely unknown. We have recently found that CD28 signaling in plasma cells (PC) is essential for sustaining Ab titers, by supporting the survival of LLPC, but not short-lived PC (SLPC). We now find that, unlike SLPC, CD28 activation in LLPC induces prosurvival downstream Vav signaling. Knockin mice with CD28 cytoplasmic tail mutations that abrogate Vav signaling (CD28-AYAA) had significantly fewer LLPC but unaffected SLPC numbers, whereas mice with mutations that abrogate PI3K signaling (CD28-Y170F) were indistinguishable from wild-type controls. This was consistent with the loss of CD28's prosurvival effect in LLPC from CD28-AYAA, but not CD28-Y170F, mice. Furthermore, the CD28 Vav motif in the B lineage was essential for the long-term maintenance of Ag-specific LLPC populations and Ab titers in vivo. Signaling downstream of the CD28 Vav motif induced previously undescribed transcriptional regulation of B lymphocyte-induced maturation protein-1, a key mediator of PC differentiation and maintenance. These findings suggest CD28 signaling in LLPC modulates the central B lymphocyte-induced maturation protein-1 transcriptional nexus involved in long-term survival and function.


Subject(s)
CD28 Antigens/metabolism , Plasma Cells/cytology , Plasma Cells/immunology , Signal Transduction/immunology , Transcription Factors/biosynthesis , Amino Acid Motifs , Animals , Antibody Formation/immunology , Blotting, Western , CD28 Antigens/immunology , Cell Differentiation/immunology , Cell Survival/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoprecipitation , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Proline , Real-Time Polymerase Chain Reaction , Transcription Factors/immunology , Up-Regulation
7.
Cancer Immunol Immunother ; 65(11): 1339-1352, 2016 11.
Article in English | MEDLINE | ID: mdl-27576783

ABSTRACT

Survivin is an anti-apoptotic protein that is highly expressed in many cancers, including malignant gliomas. Preclinical studies established that the conjugated survivin peptide mimic SurVaxM (SVN53-67/M57-KLH) could stimulate an anti-tumor immune response against murine glioma in vivo, as well as human glioma cells ex vivo. The current clinical study was conducted to test safety, immunogenicity and clinical effects of the vaccine. Recurrent malignant glioma patients whose tumors were survivin-positive, and who had either HLA-A*02 or HLA-A*03 MHC class I allele-positivity, were given subcutaneous injections of SurVaxM (500Ā Āµg) in Montanide ISA 51 with sargramostim (100Ā Āµg) at 2-week intervals. SurVaxM was well tolerated with mostly grade one adverse events (AE) and no serious adverse events (SAE) attributable to the study drug. Six patients experienced local injection site reactions; three patients reported fatigue (grades 1 and 2), and 2 patients experienced myalgia (grade 1). Six of eight immunologically evaluable patients developed both cellular and humoral immune responses to vaccine. The vaccine also stimulated HLA-A*02, HLA-A*03 and HLA-A*24 restricted T cell responses. Three patients maintained a partial clinical response or stable disease for more than 6Ā months. Median progression-free survival was 17.6Ā weeks, and median overall survival was 86.6Ā weeks from study entry with seven of nine patients surviving more than 12Ā months.


Subject(s)
Brain Neoplasms/therapy , Cancer Vaccines/immunology , Glioma/therapy , Immunotherapy, Active/methods , Inhibitor of Apoptosis Proteins/immunology , Peptides/immunology , T-Lymphocytes/immunology , Adult , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Female , Glioma/immunology , Glioma/mortality , HLA-A2 Antigen/metabolism , HLA-A3 Antigen/metabolism , Humans , Immunity, Humoral , Inhibitor of Apoptosis Proteins/genetics , Interferon-gamma/metabolism , Male , Middle Aged , Peptides/genetics , Recurrence , Survival Analysis , Survivin , Treatment Outcome , Vaccines, Subunit
8.
Blood ; 124(12): 1873-9, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25097176

ABSTRACT

Multiple myeloma is a plasma cell malignancy in which significant advances have been observed during the last 15 years. Our understanding of the disease has been advanced through its molecular characterization. We have also seen improvements in patient care with the development of 2 new classes of active agents, proteasome inhibitors and immunomodulatory drugs (IMiDs), resulting in a significant improvement in overall survival of myeloma patients such that it can now be debated as to whether some subsets of myeloma patients can be cured. However, the advances in our understanding of myeloma biology occurred in parallel with advances in treatment as opposed to being directly informed by the research. Moreover, the molecular characterization of malignant plasma cells would not have predicted the effectiveness of these novel therapies.We hypothesize that proteasome inhibitors and IMiDs are highly active because malignant plasma cells are constrained by many of the characteristics of their normal counterparts and these novel therapies target both normal plasma cell biology and the cancer biology of myeloma. Thus, a better understanding of normal plasma cell biology will likely yield as many actionable targets as mapping the genomic landscape of this disease.


Subject(s)
Multiple Myeloma/etiology , Multiple Myeloma/therapy , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Immunologic Factors/therapeutic use , Multiple Myeloma/genetics , Mutation , Plasma Cells/immunology , Plasma Cells/pathology , Plasma Cells/physiology , Proteasome Inhibitors/therapeutic use
9.
Blood ; 123(24): 3770-9, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24782505

ABSTRACT

Chemotherapeutic resistance remains a significant hurdle in the treatment of multiple myeloma (MM) and is significantly mediated by interactions between MM cells and stromal cells of the bone marrow microenvironment. Despite the importance of these interactions, the specific molecules and downstream signaling components involved remain incompletely understood. We have previously shown that the prototypic T-cell costimulatory receptor CD28, which is also expressed on MM cells, is a key mediator of MM survival and apoptotic resistance. Crosslinking CD28 by agonistic antibodies or myeloid dendritic cells (DC; these express the CD28 ligands CD80/CD86) prevents apoptosis caused by chemotherapy or serum withdrawal. We now report that CD28 pro-survival signaling is dependent upon downstream activation of phosphatidyl-inositol 3-kinase/Akt, inactivation of the transcription factor FoxO3a, and decreased expression of the pro-apoptotic molecule Bim. Conversely, blocking the CD28-CD80/CD86 interaction between MM cells and DC in vitro abrogates the DC's ability to protect MM cells against chemotherapy-induced death. Consistent with these observations, in vivo blockade of CD28-CD80/CD86 in the Vk*MYC murine myeloma model sensitizes MM cells to chemotherapy and significantly reduces tumor burden. Taken together, our findings suggest that CD28 is an important mediator of MM survival during stress and can be targeted to overcome chemotherapy resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , CD28 Antigens/physiology , Drug Resistance, Neoplasm/genetics , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Animals , Antibodies/pharmacology , CD28 Antigens/immunology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Dendritic Cells/physiology , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Mice, Transgenic , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/genetics
10.
J Biol Chem ; 289(11): 7747-62, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24415757

ABSTRACT

Dendritic cells (DC) play a critical role in modulating antigen-specific immune responses elicited by T cells via engagement of the prototypic T cell costimulatory receptor CD28 by the cognate ligands CD80/CD86, expressed on DC. Although CD28 signaling in T cell activation has been well characterized, it has only recently been shown that CD80/CD86, which have no demonstrated binding domains for signaling proteins in their cytoplasmic tails, nonetheless also transduce signals to the DC. Functionally, CD80/CD86 engagement results in DC production of the pro-inflammatory cytokine IL-6, which is necessary for full T cell activation. However, ligation of CD80/CD86 by CTLA4 also induces DC production of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO), which depletes local pools of the essential amino acid tryptophan, resulting in blockade of T cell activation. Despite the significant role of CD80/CD86 in immunological processes and the seemingly opposing roles they play by producing IL-6 and IDO upon their activation, how CD80/CD86 signal remains poorly understood. We have now found that cross-linking CD80/CD86 in human DC activates the PI3K/AKT pathway. This results in phosphorylation/inactivation of its downstream target, FOXO3A, and alleviates FOXO3A-mediated suppression of IL-6 expression. A second event downstream of AKT phosphorylation is activation of the canonical NF-κB pathway, which induces IL-6 expression. In addition to these downstream pathways, we unexpectedly found that CD80/CD86-induced PI3K signaling is regulated by previously unrecognized cross-talk with NOTCH1 signaling. This cross-talk is facilitated by NOTCH-mediated up-regulation of the expression of prolyl isomerase PIN1, which in turn increases enzyme activity of casein kinase II. Subsequently, phosphatase and tensin homolog (which suppresses PI3K activity) is inactivated via phosphorylation by casein kinase II. This results in full activation of PI3K signaling upon cross-linking CD80/CD86. Similar to IL-6, we have found that CD80/CD86-induced IDO production by DC at late time points is also dependent upon the PI3K Ć¢Ā†Ā’ AKT Ć¢Ā†Ā’ NF-κB pathway and requires cross-talk with NOTCH signaling. These data further suggest that the same signaling pathways downstream of DC CD80/CD86 cross-linking induce early IL-6 production to enhance T cell activation, followed by later IDO production to self-limit this activation. In addition to characterizing the pathways downstream of CD80/CD86 in IL-6 and IDO production, identification of a novel cross-talk between NOTCH1 and PI3K signaling may provide new insights in other biological processes where PI3K signaling plays a major role.


Subject(s)
Dendritic Cells/cytology , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Interleukin-6/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Casein Kinase II/metabolism , Cell Proliferation , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Regulation, Enzymologic , Humans , Interferon-gamma/metabolism , Interleukin-23/metabolism , Jurkat Cells , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , RNA, Small Interfering/metabolism , T-Lymphocytes/metabolism
11.
Biochim Biophys Acta ; 1840(3): 1004-13, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24211252

ABSTRACT

BACKGROUND: CD7 expression is found on ~30% of acute myeloblastic leukemias (AML). The leukemic progenitor cell line KG1a (CD7+) constitutively expresses GM-CSF while the parental KG1 (CD7-) cell line does not. This study focuses on the molecular basis of CD7 mediated GM-CSF regulation. METHODS: KG1a cells were treated with recombinant SECTM1-Fc protein, the PI3K kinase inhibitors wortmannin, LY292004, or PI4K activator spermine. Stable KG1-CD7+, KG1a-shCD7, KG1a-shETS1 as well as KG1a-GFP, KG1a-PKCƟII-GFP cell lines were generated and the levels of CD7, GM-CSF and ETS-1 mRNA and protein were compared by real-time-PCR, western blotting, flow cytometry and ELISA. RESULTS: SECTM1 is expressed in Human Bone Marrow Endothelial Cells (HBMEC) and its expression can be upregulated by both IFN-ƎĀ³. KG1a cells demonstrated high expression levels of CD7 and ETS-1 allowing a constitutative signaling through the PI3K/Atk pathway to promote GM-CSF expression, while KG1 cells with low expression of CD7 and ETS-1 showed low GM-CSF expression. On KG1a cells GM-CSF expression could be negatively regulated by PI3K inhibitors or by recombinant SECTM1-Fc. Overexpression of CD7 in KG1 cells was insufficient to promote GM-CSF expression, while silencing of CD7 or ETS-1 resulted in reduced GM-CSF expression levels. Differentiation capable KG1a cells overexpressing PKCƟII illustrated complete loss of CD7, but maintained normal levels of both ETS-1 and GM-CSF expression. CONCLUSION: These findings add an additional layer to the previously described autocrine/paracrine signaling between leukemic progenitor cells and the bone marrow microenvironment and highlight a role for SECTM1 in both normal and malignant hematopoiesis. GENERAL SIGNIFICANCE: This work shows that SECTM1 secreted from bone marrow stromal cells may interact with CD7 to influence GM-CSF expression in leukemic cells.


Subject(s)
Antigens, CD7/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Leukemia, Myeloid, Acute/metabolism , Membrane Proteins/physiology , Neoplastic Stem Cells/metabolism , Proto-Oncogene Protein c-ets-1/physiology , Cell Line, Tumor , Humans , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Transcription, Genetic
12.
Blood ; 119(6): 1450-8, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22144178

ABSTRACT

Bortezomib, a therapeutic agent for multiple myeloma (MM) and mantle cell lymphoma, suppresses proteosomal degradation leading to substantial changes in cellular transcriptional programs and ultimately resulting in apoptosis. Transcriptional regulators required for bortezomib-induced apoptosis in MM cells are largely unknown. Using gene expression profiling, we identified 36 transcription factors that displayed altered expression in MM cells treated with bortezomib. Analysis of a publically available database identified Kruppel-like family factor 9 (KLF9) as the only transcription factor with significantly higher basal expression in MM cells from patients who responded to bortezomib compared with nonresponders. We demonstrated that KLF9 in cultured MM cells was up-regulated by bortezomib; however, it was not through the induction of endoplasmic reticulum stress. Instead, KLF9 levels correlated with bortezomib-dependent inhibition of histone deacetylases (HDAC) and were increased by the HDAC inhibitor LBH589 (panobinostat). Furthermore, bortezomib induced binding of endogenous KLF9 to the promoter of the proapoptotic gene NOXA. Importantly, KLF9 knockdown impaired NOXA up-regulation and apoptosis caused by bortezomib, LBH589, or a combination of theses drugs, whereas KLF9 overexpression induced apoptosis that was partially NOXA-dependent. Our data identify KLF9 as a novel and potentially clinically relevant transcriptional regulator of drug-induced apoptosis in MM cells.


Subject(s)
Apoptosis/drug effects , Boronic Acids/pharmacology , Hydroxamic Acids/pharmacology , Kruppel-Like Transcription Factors/genetics , Multiple Myeloma/genetics , Pyrazines/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Bortezomib , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles , Kruppel-Like Transcription Factors/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Oligonucleotide Array Sequence Analysis , Panobinostat , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Cell Rep ; 43(7): 114445, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38968073

ABSTRACT

Pro-survival metabolic adaptations to stress in tumorigenesis remain less well defined. We find that multiple myeloma (MM) is unexpectedly dependent on beta-oxidation of long-chain fatty acids (FAs) for survival under both basal and stress conditions. However, under stress conditions, a second pro-survival signal is required to sustain FA oxidation (FAO). We previously found that CD28 is expressed on MM cells and transduces a significant pro-survival/chemotherapy resistance signal. We now find that CD28 signaling regulates autophagy/lipophagy that involves activation of the Ca2+Ć¢Ā†Ā’AMPKĆ¢Ā†Ā’ULK1 axis and regulates the translation of ATG5 through HuR, resulting in sustained lipophagy, increased FAO, and enhanced MM survival. Conversely, blocking autophagy/lipophagy sensitizes MM to chemotherapy inĀ vivo. Our findings link a pro-survival signal to FA availability needed to sustain the FAO required for cancer cell survival under stress conditions and identify lipophagy as a therapeutic target to overcome treatment resistance in MM.


Subject(s)
Autophagy , Cell Survival , Multiple Myeloma , Signal Transduction , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Humans , Autophagy/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Stress, Physiological/drug effects , Mice , Fatty Acids/metabolism , Drug Resistance, Neoplasm , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/genetics
14.
bioRxiv ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38562904

ABSTRACT

Recent FDA approvals of chimeric antigen receptor (CAR) T cell therapy for multiple myeloma (MM) have reshaped the therapeutic landscape for this incurable cancer. In pivotal clinical trials B cell maturation antigen (BCMA) targeted, 4-1BB co-stimulated (BBƎĀ¶) CAR T cells dramatically outperformed standard-of-care chemotherapy, yet most patients experienced MM relapse within two years of therapy, underscoring the need to improve CAR T cell efficacy in MM. We set out to determine if inhibition of MM bone marrow microenvironment (BME) survival signaling could increase sensitivity to CAR T cells. In contrast to expectations, blocking the CD28 MM survival signal with abatacept (CTLA4-Ig) accelerated disease relapse following CAR T therapy in preclinical models, potentially due to blocking CD28 signaling in CAR T cells. Knockout studies confirmed that endogenous CD28 expressed on BBƎĀ¶ CAR T cells drove in vivo anti-MM activity. Mechanistically, CD28 reprogrammed mitochondrial metabolism to maintain redox balance and CAR T cell proliferation in the MM BME. Transient CD28 inhibition with abatacept restrained rapid BBƎĀ¶ CAR T cell expansion and limited inflammatory cytokines in the MM BME without significantly affecting long-term survival of treated mice. Overall, data directly demonstrate a need for CD28 signaling for sustained in vivo function of CAR T cells and indicate that transient CD28 blockade could reduce cytokine release and associated toxicities.

15.
Nat Genet ; 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294496

ABSTRACT

Although the spatial, cellular and molecular landscapes of resected pancreatic ductal adenocarcinoma (PDAC) are well documented, the characteristics of its metastatic ecology remain elusive. By applying spatially resolved transcriptomics to matched primary and metastatic PDAC samples, we discovered a conserved continuum of fibrotic, metabolic and immunosuppressive spatial ecotypes across anatomical regions. We observed spatial tumor microenvironment heterogeneity spanning beyond that previously appreciated in PDAC. Through comparative analysis, we show that the spatial ecotypes exhibit distinct enrichment between primary and metastatic sites, implying adaptability to the local environment for survival and progression. The invasive border ecotype exhibits both pro-tumorigenic and anti-tumorigenic cell-type enrichment, suggesting a potential immunotherapy target. The ecotype heterogeneity across patients emphasizes the need to map individual patient landscapes to develop personalized treatment strategies. Collectively, our findings provide critical insights into metastatic PDAC biology and serve as a valuable resource for future therapeutic exploration and molecular investigations.

16.
Cell Mol Immunol ; 21(3): 260-274, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38233562

ABSTRACT

Metabolic flexibility has emerged as a critical determinant of CD8+ T-cell antitumor activity, yet the mechanisms driving the metabolic flexibility of T cells have not been determined. In this study, we investigated the influence of the nuclear cap-binding complex (CBC) adaptor protein ARS2 on mature T cells. In doing so, we discovered a novel signaling axis that endows activated CD8+ T cells with flexibility of glucose catabolism. ARS2 upregulation driven by CD28 signaling reinforced splicing factor recruitment to pre-mRNAs and affected approximately one-third of T-cell activation-induced alternative splicing events. Among these effects, the CD28-ARS2 axis suppressed the expression of the M1 isoform of pyruvate kinase in favor of PKM2, a key determinant of CD8+ T-cell glucose utilization, interferon gamma production, and antitumor effector function. Importantly, PKM alternative splicing occurred independently of CD28-driven PI3K pathway activation, revealing a novel means by which costimulation reprograms glucose metabolism in CD8+ T cells.


Subject(s)
Alternative Splicing , CD28 Antigens , CD28 Antigens/metabolism , Alternative Splicing/genetics , Phosphatidylinositol 3-Kinases/metabolism , CD8-Positive T-Lymphocytes , Glucose/metabolism
17.
Breast Cancer Res Treat ; 139(2): 477-88, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624818

ABSTRACT

Immune signatures in breast tumors differ by estrogen receptor (ER) status. The purpose of this study was to assess associations between ER phenotypes and circulating levels of cytokines that co-ordinate cell-mediated [T-helper type 1 (Th1)] and humoral [T-helper type 2 (Th2)] immunity. We conducted a case-case comparison of 523 women with newly diagnosed breast cancer to evaluate associations between 27 circulating cytokines, measured using Luminex XMap technology, and breast cancer phenotypes [ER(-) vs. ER(+); triple negative breast cancer (TNBC) vs. luminal A (LumA)]. Ratios of Th1 to Th2 cytokines were also evaluated. Levels of interleukin (IL)-5, a Th-2 cytokine, were higher in ER(-) than in ER(+) tumors. The highest tertile of IL-5 was more strongly associated with ER(-) (OR = 2.33, 95 % CI 1.40-3.90) and TNBCs (OR = 2.78, 95 % CI 1.53-5.06) compared to ER(+) and LumA cancers, respectively, particularly among premenopausal women (OR = 4.17, 95 % CI 1.86-9.34, ER(-) vs. ER(+); OR = 5.60, 95 % CI 2.09-15.01, TNBC vs. LumA). Elevated Th1 cytokines were also detected in women with ER(-) and TNBCs, with women in the highest tertile of interferon α2 (OR = 2.39, 95 % CI 1.31-4.35) or tumor necrosis factor-α (OR = 2.27, 95 % CI 1.21-4.26) being twice as likely to have TNBC versus LumA cancer. When cytokine ratios were examined, women with the highest ratios of Th1 cytokines to IL-5 levels were least likely to have ER(-) or TNBCs compared to ER(+) or LumA cancers, respectively. The strongest associations were in premenopausal women, who were up to 80 % less likely to have TNBC than LumA cancers (IL-12p40/IL-5, OR = 0.19, 95 % CI 0.07-0.56). These findings indicate that immune function is associated with ER(-) and TNBC and may be most relevant among younger women, who are likely to be diagnosed with these aggressive phenotypes.


Subject(s)
Breast Neoplasms/metabolism , Cytokines/blood , Triple Negative Breast Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Humans , Middle Aged , Neoplasm Grading , Neoplasm Staging , Receptors, Estrogen , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Triple Negative Breast Neoplasms/blood , Triple Negative Breast Neoplasms/immunology
18.
Blood ; 118(5): 1329-39, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21659544

ABSTRACT

Dependence on Bcl-2 proteins is a common feature of cancer cells and provides a therapeutic opportunity. ABT-737 is an antagonist of antiapoptotic Bcl-2 proteins and therefore is a good predictor of Bcl-x(L)/Bcl-2 dependence. Surprisingly, analysis of Mcl-1-dependent multiple myeloma cell lines revealed codependence on Bcl-2/Bcl-x(L) in half the cells tested. Codependence is not predicted by the expression level of antiapoptotic proteins, rather through interactions with Bim. Consistent with these findings, acquired resistance to ABT-737 results in loss of codependence through redistribution of Bim to Mcl-1. Overall, these results suggest that complex interactions, and not simply expression patterns of Bcl-2 proteins, need to be investigated to understand Bcl-2 dependence and how to better use agents, such as ABT-737.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Membrane Proteins/physiology , Multiple Myeloma/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins/physiology , bcl-X Protein/physiology , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Proteins/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/pharmacology , Sulfonamides/pharmacology , Tissue Distribution , bcl-X Protein/genetics , bcl-X Protein/metabolism
19.
J Immunol ; 187(3): 1243-53, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21715687

ABSTRACT

Interactions between the malignant plasma cells of multiple myeloma and stromal cells within the bone marrow microenvironment are essential for myeloma cell survival, mirroring the same dependence of normal bone marrow-resident long-lived plasma cells on specific marrow niches. These interactions directly transduce prosurvival signals to the myeloma cells and also induce niche production of supportive soluble factors. However, despite their central importance, the specific molecular and cellular components involved remain poorly characterized. We now report that the prototypic T cell costimulatory receptor CD28 is overexpressed on myeloma cells during disease progression and in the poor-prognosis subgroups and plays a previously unrecognized role as a two-way molecular bridge to support myeloid stromal cells in the microenvironment. Engagement by CD28 to its ligand CD80/CD86 on stromal dendritic cell directly transduces a prosurvival signal to myeloma cell, protecting it against chemotherapy and growth factor withdrawal-induced death. Simultaneously, CD28-mediated ligation of CD80/CD86 induces the stromal dendritic cell to produce the prosurvival cytokine IL-6 (involving novel cross-talk with the Notch pathway) and the immunosuppressive enzyme IDO. These findings identify CD28 and CD80/CD86 as important molecular components of the interaction between myeloma cells and the bone marrow microenvironment, point to similar interaction for normal plasma cells, and suggest novel therapeutic strategies to target malignant and pathogenic (e.g., in allergy and autoimmunity) plasma cells.


Subject(s)
Antigens, Neoplasm/physiology , CD28 Antigens/genetics , Immune Tolerance , Multiple Myeloma/immunology , Plasma Cells/immunology , Tumor Microenvironment/immunology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , CD28 Antigens/metabolism , CD28 Antigens/physiology , Cell Survival/genetics , Cell Survival/immunology , Coculture Techniques , Disease Progression , Humans , Immune Tolerance/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Plasma Cells/metabolism , Plasma Cells/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Cells, Cultured , Tumor Microenvironment/genetics
20.
Br J Haematol ; 157(1): 59-66, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22171982

ABSTRACT

Over-expression of anti-apoptotic BCL2 has been reported in chronic lymphocytic leukaemia (CLL), but targeting BCL2 alone did not yield appreciable clinical results. However, it was demonstrated that BCL2 inhibitors enhanced the clinical efficacy of chemo and immunotherapeutics. Lenalidomide, an immunomodulator, is clinically effective in CLL and can enhance the anti-CLL effects of CD20 targeting monoclonal antibody, rituximab. Here, we investigated the mechanism of immune-directed killing of lenalidomide in CLL and evaluated if concurrent targeting of CD20 and BCL2 can enhance this effect. In vitro treatment with lenalidomide enhanced the antibody-mediated cellular cytotoxicity (ADCC) directed by rituximab in autologous leukaemic cells. Furthermore, peripheral blood mononuclear cells obtained from patients after treatment with lenalidomide and rituximab showed increased ADCC in vitro versus control (pre-treatment sample). This effect was further enhanced with pre-treatment of tumour cells with AT-101 (a BH3 mimetic that functions as BCL2 antagonist). Our data suggest that AT-101 in combination with lenalidomide can potentially be an effective therapeutic regimen for CLL.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Gossypol/analogs & derivatives , Immunologic Factors/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell , Proto-Oncogene Proteins c-bcl-2 , Thalidomide/analogs & derivatives , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Down-Regulation/drug effects , Down-Regulation/immunology , Female , Gene Expression Regulation, Leukemic/immunology , Gossypol/pharmacology , Gossypol/therapeutic use , Humans , Immunologic Factors/therapeutic use , Lenalidomide , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/immunology , Rituximab , Thalidomide/pharmacology , Thalidomide/therapeutic use , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL