Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Immunity ; 54(6): 1154-1167.e7, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33979578

ABSTRACT

Blockade of the inhibitory receptor TIM-3 shows efficacy in cancer immunotherapy clinical trials. TIM-3 inhibits production of the chemokine CXCL9 by XCR1+ classical dendritic cells (cDC1), thereby limiting antitumor immunity in mammary carcinomas. We found that increased CXCL9 expression by splenic cDC1s upon TIM-3 blockade required type I interferons and extracellular DNA. Chemokine expression as well as combinatorial efficacy of TIM-3 blockade and paclitaxel chemotherapy were impaired by deletion of Cgas and Sting. TIM-3 blockade increased uptake of extracellular DNA by cDC1 through an endocytic process that resulted in cytoplasmic localization. DNA uptake and efficacy of TIM-3 blockade required DNA binding by HMGB1, while galectin-9-induced cell surface clustering of TIM-3 was necessary for its suppressive function. Human peripheral blood cDC1s also took up extracellular DNA upon TIM-3 blockade. Thus, TIM-3 regulates endocytosis of extracellular DNA and activation of the cytoplasmic DNA sensing cGAS-STING pathway in cDC1s, with implications for understanding the mechanisms underlying TIM-3 immunotherapy.


Subject(s)
DNA/metabolism , Dendritic Cells/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction/physiology , Animals , Biological Transport/physiology , Cell Line , Cell Line, Tumor , Chemokines/metabolism , Cytoplasm/metabolism , Endocytosis/physiology , Female , HEK293 Cells , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL
2.
Nature ; 591(7850): 464-470, 2021 03.
Article in English | MEDLINE | ID: mdl-33536615

ABSTRACT

Most ovarian cancers are infiltrated by prognostically relevant activated T cells1-3, yet exhibit low response rates to immune checkpoint inhibitors4. Memory B cell and plasma cell infiltrates have previously been associated with better outcomes in ovarian cancer5,6, but the nature and functional relevance of these responses are controversial. Here, using 3 independent cohorts that in total comprise 534 patients with high-grade serous ovarian cancer, we show that robust, protective humoral responses are dominated by the production of polyclonal IgA, which binds to polymeric IgA receptors that are universally expressed on ovarian cancer cells. Notably, tumour B-cell-derived IgA redirects myeloid cells against extracellular oncogenic drivers, which causes tumour cell death. In addition, IgA transcytosis through malignant epithelial cells elicits transcriptional changes that antagonize the RAS pathway and sensitize tumour cells to cytolytic killing by T cells, which also contributes to hindering malignant progression. Thus, tumour-antigen-specific and -antigen-independent IgA responses antagonize the growth of ovarian cancer by governing coordinated tumour cell, T cell and B cell responses. These findings provide a platform for identifying targets that are spontaneously recognized by intratumoural B-cell-derived antibodies, and suggest that immunotherapies that augment B cell responses may be more effective than approaches that focus on T cells, particularly for malignancies that are resistant to checkpoint inhibitors.


Subject(s)
Antigens, Neoplasm/immunology , Immunoglobulin A/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Transcytosis , Antibody Specificity , Antigens, CD/immunology , Cell Line , Disease Progression , Female , Humans , Ovarian Neoplasms/prevention & control , Receptors, Fc/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , Transcytosis/immunology , Tumor Microenvironment/immunology
3.
Immunity ; 52(4): 668-682.e7, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294407

ABSTRACT

The primary mechanisms supporting immunoregulatory polarization of myeloid cells upon infiltration into tumors remain largely unexplored. Elucidation of these signals could enable better strategies to restore protective anti-tumor immunity. Here, we investigated the role of the intrinsic activation of the PKR-like endoplasmic reticulum (ER) kinase (PERK) in the immunoinhibitory actions of tumor-associated myeloid-derived suppressor cells (tumor-MDSCs). PERK signaling increased in tumor-MDSCs, and its deletion transformed MDSCs into myeloid cells that activated CD8+ T cell-mediated immunity against cancer. Tumor-MDSCs lacking PERK exhibited disrupted NRF2-driven antioxidant capacity and impaired mitochondrial respiratory homeostasis. Moreover, reduced NRF2 signaling in PERK-deficient MDSCs elicited cytosolic mitochondrial DNA elevation and, consequently, STING-dependent expression of anti-tumor type I interferon. Reactivation of NRF2 signaling, conditional deletion of STING, or blockade of type I interferon receptor I restored the immunoinhibitory potential of PERK-ablated MDSCs. Our findings demonstrate the pivotal role of PERK in tumor-MDSC functionality and unveil strategies to reprogram immunosuppressive myelopoiesis in tumors to boost cancer immunotherapy.


Subject(s)
Carcinoma, Lewis Lung/immunology , Carcinoma, Ovarian Epithelial/immunology , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/immunology , Membrane Proteins/immunology , Skin Neoplasms/immunology , eIF-2 Kinase/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Female , Humans , Immunosuppression Therapy , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Unfolded Protein Response/immunology , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
4.
Cell Death Discov ; 5: 134, 2019.
Article in English | MEDLINE | ID: mdl-31531232

ABSTRACT

Polyphenols elicit antitumor activities, in part, through the induction of anti- or pro-oxidant effects in cancer cells which promote priming of protective anti-tumor immunity. We recently characterized a polyphenol-rich extract from Caesalpinia spinosa (P2Et) that stimulates in vivo antitumor responses against breast and melanoma tumor models via the promotion of immunogenic cancer cell death (ICD). However, the primary mediators whereby P2Et promotes ICD remained unknown. Here, we sought to elucidate the role that severe endoplasmic reticulum (ER) stress plays in mediating P2Et-induced apoptosis and ICD in murine melanoma cells. Our findings demonstrate a substantial selective induction of specific ER-stress mediators in B16-F10 melanoma cells treated with P2Et. While knockout of the ER stress-associated PKR-like ER kinase (PERK) prevented induction of apoptosis and expression of ICD markers in P2Et-treated cells, deletion of X-box binding protein 1 (Xbp1) did not. P2Et-driven activation of PERK in melanoma cells was found to promote ER-calcium release, disrupt mitochondrial membrane potential, and trigger upregulation of ICD drivers, surface calreticulin expression, and extracellular release of ATP and HMGB1. Notably, calcium release inhibition, but not targeting of PERK-driven integrated stress responses, prevented P2Et-induced apoptosis. Collectively, these results underline the central role of PERK-directed calcium release in mediating the antitumor and immunogenic actions of P2Et in melanoma cells.

6.
Cancer Res ; 79(19): 5034-5047, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31409640

ABSTRACT

Myeloid-derived suppressor cells (MDSC) represent a primary mechanism of immune evasion in tumors and have emerged as a major obstacle for cancer immunotherapy. The immunoinhibitory activity of MDSC is tightly regulated by the tumor microenvironment and occurs through mechanistic mediators that remain unclear. Here, we elucidated the intrinsic interaction between the expression of AMP-activated protein kinase alpha (AMPKα) and the immunoregulatory activity of MDSC in tumors. AMPKα signaling was increased in tumor-MDSC from tumor-bearing mice and patients with ovarian cancer. Transcription of the Ampkα1-coding gene, Prkaa1, in tumor-MDSC was induced by cancer cell-derived granulocyte-monocyte colony-stimulating factor (GM-CSF) and occurred in a Stat5-dependent manner. Conditional deletion of Prkaa1 in myeloid cells, or therapeutic inhibition of Ampkα in tumor-bearing mice, delayed tumor growth, inhibited the immunosuppressive potential of MDSC, triggered antitumor CD8+ T-cell immunity, and boosted the efficacy of T-cell immunotherapy. Complementarily, therapeutic stimulation of AMPKα signaling intrinsically promoted MDSC immunoregulatory activity. In addition, Prkaa1 deletion antagonized the differentiation of monocytic-MDSC (M-MDSC) to macrophages and re-routed M-MDSC, but not granulocytic-MDSC (PMN-MDSC), into cells that elicited direct antitumor cytotoxic effects through nitric oxide synthase 2-mediated actions. Thus, our results demonstrate the primary role of AMPKα1 in the immunosuppressive effects induced by tumor-MDSC and support the therapeutic use of AMPK inhibitors to overcome MDSC-induced T-cell dysfunction in cancer. SIGNIFICANCE: AMPKα1 regulates the immunosuppressive activity and differentiation of tumor-MDSC, suggesting AMPK inhibition as a potential therapeutic strategy to restore protective myelopoiesis in cancer.


Subject(s)
AMP-Activated Protein Kinases/immunology , Carcinoma, Ovarian Epithelial/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Tumor Microenvironment/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Carcinoma, Ovarian Epithelial/metabolism , Cell Differentiation/immunology , Female , Humans , Mice , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms, Experimental/metabolism , Tumor Escape/immunology
7.
Nat Commun ; 10(1): 1280, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894532

ABSTRACT

Understanding the intrinsic mediators that render CD8+ T cells dysfunctional in the tumor microenvironment is a requirement to develop more effective cancer immunotherapies. Here, we report that C/EBP homologous protein (Chop), a downstream sensor of severe endoplasmic reticulum (ER) stress, is a major negative regulator of the effector function of tumor-reactive CD8+ T cells. Chop expression is increased in tumor-infiltrating CD8+ T cells, which correlates with poor clinical outcome in ovarian cancer patients. Deletion of Chop in T cells improves spontaneous antitumor CD8+ T cell immunity and boosts the efficacy of T cell-based immunotherapy. Mechanistically, Chop in CD8+ T cells is elevated primarily through the ER stress-associated kinase Perk and a subsequent induction of Atf4; and directly represses the expression of T-bet, a master regulator of effector T cell function. These findings demonstrate the primary role of Chop in tumor-induced CD8+ T cell dysfunction and the therapeutic potential of blocking Chop or ER stress to unleash T cell-mediated antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Ovarian Epithelial/genetics , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/immunology , Ovarian Neoplasms/genetics , T-Box Domain Proteins/genetics , Transcription Factor CHOP/genetics , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/therapy , Cell Line, Tumor , Female , Humans , Immunity, Cellular , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Knockout , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Survival Analysis , T-Box Domain Proteins/immunology , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
8.
BMC Infect Dis ; 18(1): 463, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-30219037

ABSTRACT

BACKGROUND: Helicobacter pylori is a major aetiologic agent associated with gastritis. H. pylori infections increase the expression of the Toll-like receptor (TLR), which in turn modulates the expression of microRNA (miRNA)-146a and miRNA-155. The objective of this study was to compare the expression of miRNA-146a and miRNA-155 in gastric lesions of paediatric and adult patients with different pathologies and in Mongolian gerbils (Meriones unguiculatus) infected with H. pylori 26,695. METHODS: Quantification of miRNA expression was performed by quantitative real-time polymerase chain reaction (qRT-PCR) of paraffin-embedded gastric lesions of children with or without an infection (n = 25), adults with follicular gastritis and metaplasia (n = 32) and eight-week-old M. unguiculatus males (Hsd:MON) infected with H. pylori 26,695 for 0, 3, 6, 12 and 18 months (n = 25). The genes RNU48 and RNU6 were used as endogenous controls for data normalization. Statistical analyses were performed using Kruskal-Wallis, Mann-Whitney, ANOVA and Student's t-test. RESULTS: The expression of miRNA-146a and miRNA-155 in infected children increased by 247.6- and 79.4-fold (on average), respectively, compared to that observed in the control group. However, these results were not significant (p = 0.12 and p = 0.07 respectively). In some children a gradual increase in expression was observed, while in others, expression was very high. Additionally, the expression levels of miRNA-146a and miRNA-155 increased by an average of 21.7- and 62-fold, respectively, in adult patients with follicular gastritis when compared to those of the controls. In M. unguiculatus infected with H. pylori 26,695, the expression of both miRNAs increased as the infection progressed. CONCLUSION: This is the first report to show differences in the expression of miRNA-146a and miRNA-155 in paediatric and adult patients with gastritis who were infected with H. pylori. In addition, in M. unguiculatus infected with H. pylori, miRNA expression was associated with the progression of infection and the ability of the bacteria to adapt to the host.


Subject(s)
Gastritis/genetics , Helicobacter Infections/genetics , Helicobacter pylori/physiology , MicroRNAs/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Disease Models, Animal , Disease Progression , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/microbiology , Gene Expression Profiling , Gerbillinae , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Humans , Male , Middle Aged , Young Adult
9.
Mol Cell Biol ; 38(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29483302

ABSTRACT

Insulin receptor substrate 1 (IRS-1) is a common cytosolic adaptor molecule involved in signal transduction from insulin and insulin-like growth factor I (IGF-I) receptors. IRS-1 can also be found in the nucleus. We report here a new finding of unique IRS-1 nuclear structures, which we observed initially in glioblastoma biopsy specimens and glioblastoma xenografts. These nuclear structures can be reproduced in vitro by the ectopic expression of IRS-1 cDNA cloned in frame with the nuclear localization signal (NLS-IRS-1). In these structures, IRS-1 localizes at the periphery, while the center harbors a key autophagy protein, LC3. These new nuclear structures are highly dynamic, rapidly exchange IRS-1 molecules with the surrounding nucleoplasm, disassemble during mitosis, and require a growth stimulus for their reassembly and maintenance. In tumor cells engineered to express NLS-IRS-1, the IRS-1/LC3 nuclear structures repress autophagy induced by either amino acid starvation or rapamycin treatment. In this process, IRS-1 nuclear structures sequester LC3 inside the nucleus, possibly preventing its cytosolic translocation and the formation of new autophagosomes. This novel mechanism provides a quick and reversible way of inhibiting autophagy, which could counteract autophagy-induced cancer cell death under severe stress, including anticancer therapies.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , Insulin Receptor Substrate Proteins/physiology , Adaptor Proteins, Signal Transducing , Autophagy/physiology , Cell Nucleus/physiology , Cell Survival/genetics , Glioblastoma/metabolism , HeLa Cells , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/ultrastructure , Insulin-Like Growth Factor I/physiology , Microtubule-Associated Proteins/physiology , Neoplasms , Phosphoproteins , Receptor, IGF Type 1/physiology , Signal Transduction
10.
Oncotarget ; 8(41): 70317-70331, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-29050282

ABSTRACT

Immunotherapy has shown a tremendous success in treating cancer. Unfortunately, this success is frequently associated with severe autoimmune pathology. In this study, we used the transgenic RIP-gp mouse model to assess the antitumor therapeutic benefit of peptide vaccination while evaluating the possible associated autoimmune pathology. We report that palmitoylated gp33-41 peptide and poly-IC adjuvant vaccine (BiVax) generated ∼ 5-10 % of antigen specific T cell responses in wild type and supposedly immune tolerant RIP-gp mice. Boosting with BiVax in combination with αCD40 antibody (TriVax) or BiVax in combination with IL-2/αIL-2 antibody complexes (IL2Cx) significantly increased the immune responses (∼30-50%). Interestingly, although both boosts were equally effective in generating vast T cell responses, BiVax/IL2Cx showed better control of tumor growth than TriVax. However, this effect was associated with high incidence of diabetes in an antigen and CD8 dependent fashion. T cell responses generated by BiVax/IL2Cx, but not those generated by TriVax were highly resistant to PD-1/PD-L1 inhibitory signals. Nevertheless, PD-1 blockade enhanced the ability of TriVax to control tumor growth but increased the incidence of diabetes. Finally, we show that severe autoimmunity by BiVax/IL2Cx was prevented while preserving outstanding antitumor responses by utilizing a tumor antigen not expressed in the pancreas. Our data provides a clear evidence that peptide based vaccines can expand vast endogenous T cell responses which effectively control tumor growth but with high potential of autoimmune pathology.

11.
Cancer Res ; 77(20): 5628-5638, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28904063

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a major obstacle to promising forms of cancer immunotherapy, but tools to broadly limit their immunoregulatory effects remain lacking. In this study, we assessed the therapeutic effect of the humanized anti-Jagged1/2-blocking antibody CTX014 on MDSC-mediated T-cell suppression in tumor-bearing mice. CTX014 decreased tumor growth, affected the accumulation and tolerogenic activity of MDSCs in tumors, and inhibited the expression of immunosuppressive factors arginase I and iNOS. Consequently, anti-Jagged therapy overcame tumor-induced T-cell tolerance, increased the infiltration of reactive CD8+ T cells into tumors, and enhanced the efficacy of T-cell-based immunotherapy. Depletion of MDSC-like cells restored tumor growth in mice treated with anti-Jagged, whereas coinjection of MDSC-like cells from anti-Jagged-treated mice with cancer cells delayed tumor growth. Jagged1/2 was induced in MDSCs by tumor-derived factors via NFkB-p65 signaling, and conditional deletion of NFkB-p65 blocked MDSC function. Collectively, our results offer a preclinical proof of concept for the use of anti-Jagged1/2 to reprogram MDSC-mediated T-cell suppression in tumors, with implications to broadly improve the efficacy of cancer therapy. Cancer Res; 77(20); 5628-38. ©2017 AACR.


Subject(s)
Immunotherapy/methods , Jagged-1 Protein/antagonists & inhibitors , Jagged-1 Protein/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Animals , Antibodies/immunology , Antibodies/pharmacology , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/therapy , Female , Humans , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes/immunology
12.
Cancer Prev Res (Phila) ; 10(7): 377-388, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28468928

ABSTRACT

Intestinal cyclic guanosine monophosphate (cGMP) signaling regulates epithelial homeostasis and has been implicated in the suppression of colitis and colon cancer. In this study, we investigated the cGMP-elevating ability of the phosphodiesterase-5 (PDE5) inhibitor sildenafil to prevent disease in the azoxymethane/dextran sulfate sodium (AOM/DSS) inflammation-driven colorectal cancer model. Treatment of mice with sildenafil activated cGMP signaling in the colon mucosa and protected against dextran-sulfate sodium (DSS)-induced barrier dysfunction. In mice treated with AOM/DSS, oral administration of sildenafil throughout the disease course reduced polyp multiplicity by 50% compared with untreated controls. Polyps that did form in sildenafil treated mice were less proliferative and more differentiated compared with polyps from untreated mice, but apoptosis was unaffected. Polyps in sildenafil treated mice were also less inflamed; they exhibited reduced myeloid-cell infiltration and reduced expression of iNOS, IFNγ, and IL6 compared with untreated controls. Most of the protection conferred by sildenafil was during the initiation stage of carcinogenesis (38% reduction in multiplicity). Administration of sildenafil during the later promotion stages did not affect multiplicity but had a similar effect on the polyp phenotype, including increased mucus production, and reduced proliferation and inflammation. In summary, the results demonstrate that oral administration of sildenafil suppresses polyp formation and inflammation in mice treated with AOM/DSS. This validation of PDE5 as a target highlights the potential therapeutic value of PDE5 inhibitors for the prevention of colitis-driven colon cancer in humans. Cancer Prev Res; 10(7); 377-88. ©2017 AACRSee related editorial by Piazza, p. 373.


Subject(s)
Carcinogenesis/drug effects , Colitis/drug therapy , Colon/drug effects , Colorectal Neoplasms/prevention & control , Intestinal Mucosa/drug effects , Phosphodiesterase 5 Inhibitors/therapeutic use , Administration, Oral , Animals , Apoptosis/drug effects , Azoxymethane/toxicity , Colitis/chemically induced , Colitis/complications , Colon/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Dextran Sulfate/toxicity , Humans , Immunohistochemistry , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/etiology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/prevention & control , Polyps/etiology , Polyps/pathology , Polyps/prevention & control , Signal Transduction/drug effects , Sildenafil Citrate/therapeutic use
13.
Cancer Lett ; 371(1): 90-8, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26639196

ABSTRACT

Infection with Helicobacter pylori (H. pylori) leads to inflammatory events that can promote gastric cancer development. Immune cells transition from the circulation into the infected mucosa through the interaction of their receptors and ligands in the endothelial compartment. CD44 expression is increased in advanced gastric lesions. However, the association of this molecule with the progression of these lesions over time has not been investigated. In addition, there is a lack of understanding of the CD44-dependent cellular processes that lead to gastritis, and possibly to gastric cancer. Here we studied H. pylori-positive subjects with gastric lesions that ranged from multifocal atrophic gastritis to dysplasia to determine gene expression changes associated with disease progression over a period of 6 years. We report that CD44 expression is significantly increased in individuals whose gastric lesions progressed along the gastric precancerous cascade. We also show that CD44-/- mice develop less severe and less extensive H. pylori-induced metaplasia, and show fewer infiltrating Gr1+ cells compared to wild type mice. We present data suggesting that CD44 is associated with disease progression. Mechanisms associated with these effects include induction of interferon gamma responses.


Subject(s)
Gastric Mucosa/metabolism , Gastritis, Atrophic/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/pathogenicity , Hyaluronan Receptors/metabolism , Precancerous Conditions/metabolism , Stomach Neoplasms/metabolism , Animals , Antigens, Ly/metabolism , Cells, Cultured , Chemotaxis, Leukocyte , Disease Models, Animal , Disease Progression , Female , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis, Atrophic/diagnosis , Gastritis, Atrophic/genetics , Gastritis, Atrophic/immunology , Gastritis, Atrophic/microbiology , Helicobacter Infections/diagnosis , Helicobacter Infections/genetics , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Interferon-gamma/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice, Knockout , Neutrophil Infiltration , Neutrophils/immunology , Neutrophils/metabolism , Precancerous Conditions/diagnosis , Precancerous Conditions/genetics , Precancerous Conditions/immunology , Precancerous Conditions/microbiology , Signal Transduction , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology , Time Factors
14.
Oncotarget ; 7(4): 3806-18, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26675551

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of several human cancers, including Kaposi's sarcoma (KS), which preferentially arise in immunocompromised patients and lack effective therapeutic options. We have previously shown that KSHV or viral protein LANA up-regulates the glycoprotein CD147, thereby inducing primary endothelial cell invasiveness. In the current study, we identify the global network controlled by CD147 in KSHV-infected endothelial cells using Illumina microarray analysis. Among downstream genes, two specific metalloproteases, ADAMTS1 and 9, are strongly expressed in AIDS-KS tissues and contribute to KSHV-infected endothelial cell invasiveness through up-regulation of IL-6 and VEGF. By using a KS-like nude mouse model, we found that targeting CD147 and downstream ADAMTSs significantly suppressed KSHV-induced tumorigenesis in vivo. Taken together, targeting CD147 and associated proteins may represent a promising therapeutic strategy against these KSHV-related malignancies.


Subject(s)
ADAM Proteins/metabolism , Basigin/metabolism , Cell Transformation, Viral , Endothelium, Vascular/pathology , Herpesvirus 8, Human/pathogenicity , Sarcoma, Kaposi/pathology , ADAM Proteins/genetics , ADAMTS1 Protein , Animals , Apoptosis , Basigin/genetics , Blotting, Western , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/virology , Gene Expression Profiling , Humans , Mice , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/virology , Xenograft Model Antitumor Assays
15.
Blood ; 126(26): 2821-31, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26531163

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) is a principal causative agent of primary effusion lymphoma (PEL) with a poor prognosis in immunocompromised patients. However, it still lacks effective treatment which urgently requires the identification of novel therapeutic targets for PEL. Here, we report that the hepatocyte growth factor (HGF)/c-MET pathway is highly activated by KSHV in vitro and in vivo. The selective c-MET inhibitor, PF-2341066, can induce PEL apoptosis through cell cycle arrest and DNA damage, and suppress tumor progression in a xenograft murine model. By using microarray analysis, we identify many novel genes that are potentially controlled by HGF/c-MET within PEL cells. One of the downstream candidates, ribonucleoside-diphosphate reductase subunit M2 (RRM2), also displays the promising therapeutic value for PEL treatment. Our findings provide the framework for development of HGF/c-MET-focused therapy and implementation of clinical trials for PEL patients.


Subject(s)
Hepatocyte Growth Factor/antagonists & inhibitors , Lymphoma, Primary Effusion/pathology , Piperidines/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Signal Transduction/drug effects , Adult , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Comet Assay , Crizotinib , DNA Damage/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Herpesviridae Infections/complications , Herpesvirus 8, Human , Humans , Immunoblotting , Immunocompromised Host , Lymphoma, Primary Effusion/immunology , Male , Mice , Mice, SCID , Middle Aged , Oligonucleotide Array Sequence Analysis , Pyrazoles , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays , Young Adult
16.
Oncotarget ; 6(27): 24246-60, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26327294

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent for several human cancers including primary effusion lymphoma (PEL), a rapidly progressive malignancy arising preferentially in immunocompromised patients. With conventional chemotherapy, PEL continues to portend high mortality, dictating the development of novel therapeutic strategies. Sphingosine kinase 2 (SphK2) represents a key gatekeeper for sphingolipid metabolism, responsible for conversion of ceramides to sphingosine-1-phosphate (S1P). We have previously demonstrated that targeting SphK2 using a novel selective inhibitor, ABC294640, leads to intracellular accumulation of ceramides and induces apoptosis for KSHV-infected PEL cells, while suppressing tumor progression in vivo. In the current study, we sought to determine whether specific ceramide/dh-ceramide species and related ceramide synthases (CerS) impact viability for KSHV-infected PEL cells during targeting of SphK2. We found that several specific ceramide and dihydro(dh)-ceramide species and their associated CerS reduce PEL survival and tumor expansion in vitro and in vivo. Moreover, we found that dhC16-Cer induces PEL apoptosis in part through activation of KSHV lytic gene expression. These data further implicate bioactive sphingolipids in regulation of PEL survival, and provide justification for future studies evaluating clinically relevant ceramide analogs or mimetics for their potential as therapeutic agents for PEL.


Subject(s)
Apoptosis , Ceramides/metabolism , Gene Expression Regulation, Viral , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Oxidoreductases/metabolism , Adamantane/analogs & derivatives , Adamantane/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pyridines/chemistry , RNA Interference , Sphingolipids/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Up-Regulation
17.
Cancer Immunol Res ; 3(11): 1236-47, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26025381

ABSTRACT

Myeloid-derived suppressor cells (MDSC) promote tumor growth by inhibiting T-cell immunity and promoting malignant cell proliferation and migration. The therapeutic potential of blocking MDSC in tumors has been limited by their heterogeneity, plasticity, and resistance to various chemotherapy agents. Recent studies have highlighted the role of energy metabolic pathways in the differentiation and function of immune cells; however, the metabolic characteristics regulating MDSC remain unclear. We aimed to determine the energy metabolic pathway(s) used by MDSC, establish its impact on their immunosuppressive function, and test whether its inhibition blocks MDSC and enhances antitumor therapies. Using several murine tumor models, we found that tumor-infiltrating MDSC (T-MDSC) increased fatty acid uptake and activated fatty acid oxidation (FAO). This was accompanied by an increased mitochondrial mass, upregulation of key FAO enzymes, and increased oxygen consumption rate. Pharmacologic inhibition of FAO blocked immune inhibitory pathways and functions in T-MDSC and decreased their production of inhibitory cytokines. FAO inhibition alone significantly delayed tumor growth in a T-cell-dependent manner and enhanced the antitumor effect of adoptive T-cell therapy. Furthermore, FAO inhibition combined with low-dose chemotherapy completely inhibited T-MDSC immunosuppressive effects and induced a significant antitumor effect. Interestingly, a similar increase in fatty acid uptake and expression of FAO-related enzymes was found in human MDSC in peripheral blood and tumors. These results support the possibility of testing FAO inhibition as a novel approach to block MDSC and enhance various cancer therapies.


Subject(s)
Colonic Neoplasms/therapy , Fatty Acids/metabolism , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid Cells/immunology , Animals , Antineoplastic Agents/therapeutic use , Antioxidants/therapeutic use , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Combined Modality Therapy , Drug Evaluation, Preclinical/methods , Energy Metabolism/immunology , Epoxy Compounds/therapeutic use , Female , Humans , Immune Tolerance/immunology , Immunotherapy, Adoptive/methods , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Tumor Microenvironment/immunology
18.
PLoS One ; 9(9): e106257, 2014.
Article in English | MEDLINE | ID: mdl-25229241

ABSTRACT

During the last decade, mounting evidence has implicated the human neurotropic virus JC virus in the pathology of colon cancer. However, the mechanisms of JC virus-mediated oncogenesis are still not fully determined. One candidate to mediate these effects is the viral early transcriptional product T-Antigen, which has the ability to inactivate cell cycle regulatory proteins such as p53. In medulloblastomas, T-Antigen has been shown to bind the Wnt signaling pathway protein ß-catenin; however, the effects of this interaction on downstream cell cycle regulatory proteins remain unknown. In light of these observations, we investigated the association of T-Antigen and nuclear ß-catenin in colon cancer cases and the effects of this complex in the activation of the transcription and cell cycle regulators c-Myc and Cyclin D1 in vitro. Gene amplification demonstrated the presence of viral sequences in 82.4% of cases and we detected expression of T-Antigen in 64.6% of cases by immunohistochemistry. Further, we found that T-Antigen and ß-catenin co-localized in the nuclei of tumor cells and we confirmed the physical binding between these two proteins in vitro. The nuclear presence of T-Antigen and ß-catenin resulted in the significant enhancement of TCF-dependent promoter activity and activation of the ß-catenin downstream targets, c-Myc and Cyclin D1. These observations provide further evidence for a role of JCV T-Antigen in the dysregulation of the Wnt signaling pathway and in the pathogenesis of colon cancer.


Subject(s)
Antigens, Viral, Tumor/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Cyclin D1/metabolism , JC Virus/immunology , JC Virus/metabolism , Proto-Oncogene Proteins c-myc/metabolism , beta Catenin/metabolism , Colonic Neoplasms/genetics , Cyclin D1/genetics , HCT116 Cells , Humans , In Vitro Techniques , beta Catenin/genetics
19.
Immunity ; 41(3): 389-401, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25238096

ABSTRACT

Adaptation of malignant cells to the hostile milieu present in tumors is an important determinant of their survival and growth. However, the interaction between tumor-linked stress and antitumor immunity remains poorly characterized. Here, we show the critical role of the cellular stress sensor C/EBP-homologous protein (Chop) in the accumulation and immune inhibitory activity of tumor-infiltrating myeloid-derived suppressor cells (MDSCs). MDSCs lacking Chop had decreased immune-regulatory functions and showed the ability to prime T cell function and induce antitumor responses. Chop expression in MDSCs was induced by tumor-linked reactive oxygen and nitrogen species and regulated by the activating-transcription factor-4. Chop-deficient MDSCs displayed reduced signaling through CCAAT/enhancer-binding protein-ß, leading to a decreased production of interleukin-6 (IL-6) and low expression of phospho-STAT3. IL-6 overexpression restored immune-suppressive activity of Chop-deficient MDSCs. These findings suggest the role of Chop in tumor-induced tolerance and the therapeutic potential of targeting Chop in MDSCs for cancer immunotherapy.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , T-Lymphocytes/immunology , Transcription Factor CHOP/genetics , Tumor Escape/immunology , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Line, Tumor , Cell Proliferation , Endothelial Cells/metabolism , Female , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Neoplasms , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/immunology , Reactive Nitrogen Species/immunology , Reactive Oxygen Species/immunology , STAT3 Transcription Factor/biosynthesis , Transcription Factor CHOP/biosynthesis
20.
Cancer Immunol Res ; 2(8): 800-11, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24830414

ABSTRACT

An impaired antitumor immunity is found in patients with cancer and represents a major obstacle in the successful development of different forms of immunotherapy. Signaling through Notch receptors regulates the differentiation and function of many cell types, including immune cells. However, the effect of Notch in CD8(+) T-cell responses in tumors remains unclear. Thus, we aimed to determine the role of Notch signaling in CD8(+) T cells in the induction of tumor-induced suppression. Our results using conditional knockout mice show that Notch-1 and Notch-2 were critical for the proliferation and IFNγ production of activated CD8(+) T cells and were significantly decreased in tumor-infiltrating T cells. Conditional transgenic expression of Notch-1 intracellular domain (N1IC) in antigen-specific CD8(+) T cells did not affect activation or proliferation of CD8(+) T cells, but induced a central memory phenotype and increased cytotoxicity effects and granzyme B levels. Consequently, a higher antitumor response and resistance to tumor-induced tolerance were found after adoptive transfer of N1IC-transgenic CD8(+) T cells into tumor-bearing mice. Additional results showed that myeloid-derived suppressor cells (MDSC) blocked the expression of Notch-1 and Notch-2 in T cells through nitric oxide-dependent mechanisms. Interestingly, N1IC overexpression rendered CD8(+) T cells resistant to the tolerogenic effect induced by MDSC in vivo. Together, the results suggest the key role of Notch in the suppression of CD8(+) T-cell responses in tumors and the therapeutic potential of N1IC in antigen-specific CD8(+) T cells to reverse T-cell suppression and increase the efficacy of T cell-based immunotherapies in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive , Neoplasms/therapy , Receptor, Notch1/immunology , Animals , Antigens/immunology , Cell Line, Tumor , Immune Tolerance , Mice, Transgenic , Neoplasms/immunology , Neoplasms/pathology , Ovalbumin/immunology , Receptor, Notch2/immunology , Signal Transduction , Tumor Burden
SELECTION OF CITATIONS
SEARCH DETAIL
...