Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Immunity ; 56(11): 2570-2583.e6, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37909039

ABSTRACT

Dimeric IgA (dIgA) can move through cells via the IgA/IgM polymeric immunoglobulin receptor (PIGR), which is expressed mainly on mucosal epithelia. Here, we studied the ability of dIgA to target commonly mutated cytoplasmic oncodrivers. Mutation-specific dIgA, but not IgG, neutralized KRASG12D within ovarian carcinoma cells and expelled this oncodriver from tumor cells. dIgA binding changed endosomal trafficking of KRASG12D from accumulation in recycling endosomes to aggregation in the early/late endosomes through which dIgA transcytoses. dIgA targeting of KRASG12D abrogated tumor cell proliferation in cell culture assays. In vivo, KRASG12D-specific dIgA1 limited the growth of KRASG12D-mutated ovarian and lung carcinomas in a manner dependent on CD8+ T cells. dIgA specific for IDH1R132H reduced colon cancer growth, demonstrating effective targeting of a cytoplasmic oncodriver not associated with surface receptors. dIgA targeting of KRASG12D restricted tumor growth more effectively than small-molecule KRASG12D inhibitors, supporting the potential of this approach for the treatment of human cancers.


Subject(s)
Carcinoma , Immunoglobulin A , Humans , Immunoglobulin A/metabolism , CD8-Positive T-Lymphocytes/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Cytoplasm/metabolism
2.
Immunity ; 55(1): 115-128.e9, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021053

ABSTRACT

The immune checkpoint receptor PD-1 on T follicular helper (Tfh) cells promotes Tfh:B cell interactions and appropriate positioning within tissues. Here, we examined the impact of regulation of PD-1 expression by the genomic organizer SATB1 on Tfh cell differentiation. Vaccination of CD4CreSatb1f/f mice enriched for antigen-specific Tfh cells, and TGF-ß-mediated repression of SATB1 enhanced Tfh differentiation of human T cells. Mechanistically, high Icos expression in Satb1-/- CD4+ T cells promoted Tfh cell differentiation by preventing T follicular regulatory cell skewing and resulted in increased isotype-switched B cell responses in vivo. Ovarian tumors in CD4CreSatb1f/f mice accumulated tumor antigen-specific, LIGHT+CXCL13+IL-21+ Tfh cells and tertiary lymphoid structures (TLS). TLS formation decreased tumor growth in a CD4+ T cell and CXCL13-dependent manner. The transfer of Tfh cells, but not naive CD4+ T cells, induced TLS at tumor beds and decreased tumor growth. Thus, TGF-ß-mediated silencing of Satb1 licenses Tfh cell differentiation, providing insight into the genesis of TLS within tumors.


Subject(s)
Germinal Center/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Tertiary Lymphoid Structures/immunology , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Gene Silencing , Genotype , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Transforming Growth Factor beta/genetics
3.
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
4.
Immunity ; 46(1): 51-64, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099864

ABSTRACT

Despite the importance of programmed cell death-1 (PD-1) in inhibiting T cell effector activity, the mechanisms regulating its expression remain poorly defined. We found that the chromatin organizer special AT-rich sequence-binding protein-1 (Satb1) restrains PD-1 expression induced upon T cell activation by recruiting a nucleosome remodeling deacetylase (NuRD) complex to Pdcd1 regulatory regions. Satb1 deficienct T cells exhibited a 40-fold increase in PD-1 expression. Tumor-derived transforming growth factor ß (Tgf-ß) decreased Satb1 expression through binding of Smad proteins to the Satb1 promoter. Smad proteins also competed with the Satb1-NuRD complex for binding to Pdcd1 enhancers, releasing Pdcd1 expression from Satb1-mediated repression, Satb1-deficient tumor-reactive T cells lost effector activity more rapidly than wild-type lymphocytes at tumor beds expressing PD-1 ligand (CD274), and these differences were abrogated by sustained CD274 blockade. Our findings suggest that Satb1 functions to prevent premature T cell exhaustion by regulating Pdcd1 expression upon T cell activation. Dysregulation of this pathway in tumor-infiltrating T cells results in diminished anti-tumor immunity.


Subject(s)
Epigenetic Repression/immunology , Gene Expression Regulation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Matrix Attachment Region Binding Proteins/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Animals , Enzyme-Linked Immunospot Assay , Humans , Immunoprecipitation , Lymphocyte Activation/immunology , Matrix Attachment Region Binding Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism
5.
Nature ; 562(7727): 423-428, 2018 10.
Article in English | MEDLINE | ID: mdl-30305738

ABSTRACT

Tumours evade immune control by creating hostile microenvironments that perturb T cell metabolism and effector function1-4. However, it remains unclear how intra-tumoral T cells integrate and interpret metabolic stress signals. Here we report that ovarian cancer-an aggressive malignancy that is refractory to standard treatments and current immunotherapies5-8-induces endoplasmic reticulum stress and activates the IRE1α-XBP1 arm of the unfolded protein response9,10 in T cells to control their mitochondrial respiration and anti-tumour function. In T cells isolated from specimens collected from patients with ovarian cancer, upregulation of XBP1 was associated with decreased infiltration of T cells into tumours and with reduced IFNG mRNA expression. Malignant ascites fluid obtained from patients with ovarian cancer inhibited glucose uptake and caused N-linked protein glycosylation defects in T cells, which triggered IRE1α-XBP1 activation that suppressed mitochondrial activity and IFNγ production. Mechanistically, induction of XBP1 regulated the abundance of glutamine carriers and thus limited the influx of glutamine that is necessary to sustain mitochondrial respiration in T cells under glucose-deprived conditions. Restoring N-linked protein glycosylation, abrogating IRE1α-XBP1 activation or enforcing expression of glutamine transporters enhanced mitochondrial respiration in human T cells exposed to ovarian cancer ascites. XBP1-deficient T cells in the metastatic ovarian cancer milieu exhibited global transcriptional reprogramming and improved effector capacity. Accordingly, mice that bear ovarian cancer and lack XBP1 selectively in T cells demonstrate superior anti-tumour immunity, delayed malignant progression and increased overall survival. Controlling endoplasmic reticulum stress or targeting IRE1α-XBP1 signalling may help to restore the metabolic fitness and anti-tumour capacity of T cells in cancer hosts.


Subject(s)
Endoribonucleases/metabolism , Mitochondria/metabolism , Ovarian Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , X-Box Binding Protein 1/metabolism , Amino Acid Transport Systems, Basic , Animals , Ascites/metabolism , Cell Respiration , Disease Progression , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glutamine/metabolism , Glycosylation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Signal Transduction , Survival Rate , T-Lymphocytes/metabolism , Tumor Escape/immunology , Unfolded Protein Response , X-Box Binding Protein 1/biosynthesis , X-Box Binding Protein 1/deficiency
6.
Cancer Immunol Immunother ; 72(6): 1445-1460, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36469096

ABSTRACT

Radiation therapy (RT) can prime and boost systemic anti-tumor effects via STING activation, resulting in enhanced tumor antigen presentation and antigen recognition by T cells. It is increasingly recognized that optimal anti-tumor immune responses benefit from coordinated cellular (T cell) and humoral (B cell) responses. However, the nature and functional relevance of the RT-induced immune response are controversial, beyond STING signaling, and agonistic interventions are lacking. Here, we show that B and CD4+ T cell accumulation at tumor beds in response to RT precedes the arrival of CD8+ T cells, and both cell types are absolutely required for abrogated tumor growth in non-irradiated tumors. Further, RT induces increased expression of 4-1BB (CD137) in both T and B cells; both in preclinical models and in a cohort of patients with small cell lung cancer treated with thoracic RT. Accordingly, the combination of RT and anti-41BB therapy leads to increased immune cell infiltration in the tumor microenvironment and significant abscopal effects. Thus, 4-1BB therapy enhances radiation-induced tumor-specific immune responses via coordinated B and T cell responses, thereby preventing malignant progression at unirradiated tumor sites. These findings provide a rationale for combining RT and 4-1bb therapy in future clinical trials.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Neoplasms/radiotherapy , Immunotherapy , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Lymphocyte Activation , Tumor Microenvironment
7.
Gynecol Oncol ; 173: 114-121, 2023 06.
Article in English | MEDLINE | ID: mdl-37121178

ABSTRACT

OBJECTIVE: To demonstrate that shared antibody responses in endometriosis and endometriosis-associated ovarian cancer spontaneously antagonize malignant progression and can be leveraged to develop future immunotherapies. METHODS: B cells from cyopreserved clear cell ovarian carcinoma (CCC, n = 2), endometrioid ovarian carcinoma (EC, n = 2), and endometriomas (n = 2) were isolated, activated, and EBV-immortalized. Antibodies were purified from B cell supernatants and used for screening arrays containing most of the human proteome. Targets were prioritized based on accessibility (transmembrane or secreted proteins), expression in endometriosis and cancer, and concurrent IgA and IgG responses. We focused on antibodies targeting tumor-promoting syndecan binding protein (SDCBP) to demonstrate anti-tumor activity. Immunoblots and qPCR were performed to assess SDCBP expression in ovarian cancer and endometriosis cell lines and tumor samples. Recombinant IgG4 was generated using the variable heavy and light chains of dominant B cell receptors (BCRs) reacting against the extracellular domain of SDCBP, and used in in vivo studies in human CCC- and high-grade serous ovarian carcinoma (HGSOC)-bearing immunodeficient mice. RESULTS: Nine accessible proteins detected by both IgA and IgG were identified in all samples - including SDCBP, which is expressed in ovarian carcinomas of multiple histologies. Administration of α-SDCBP IgG4 in OVCAR3 (HGSOC), TOV21G and RMG-I (CCC) tumor-bearing mice significantly decreased tumor volume compared to control irrelevant IgG4. CONCLUSIONS: Spontaneous antibody responses exert suboptimal but measurable immune pressure against malignant progression in ovarian carcinomas. Using tumor-derived antibodies for developing novel immunotherapeutics warrants further investigation.


Subject(s)
Adenocarcinoma, Clear Cell , Carcinoma, Endometrioid , Endometriosis , Ovarian Neoplasms , Humans , Female , Animals , Mice , Ovarian Neoplasms/pathology , Apoptosis , Antibody Formation , Cell Line, Tumor , Carcinoma, Ovarian Epithelial , Carcinoma, Endometrioid/pathology , Immunoglobulin A/metabolism , Adenocarcinoma, Clear Cell/pathology , Syntenins/metabolism
8.
Life (Basel) ; 14(3)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38541651

ABSTRACT

Immuno-oncology has traditionally focused on conventional MHC-restricted αß T cells. Yet, unconventional γδ T cells, which kill tumor cells in an MHC-unrestricted manner, display characteristics of effector activity and stemness without exhaustion and are nearly universally observed in human gynecologic malignancies, correlating with improved outcomes. These cells do not have a clear counterpart in mice but are also found in the healthy female reproductive tract. Interventions that modulate their in vivo activity, or cellular therapies utilizing γδ T cells as an allogeneic, "off-the-shelf" platform (e.g., for chimeric antigen receptor expression) hold significant potential against challenging tumors like ovarian cancer, which has been stubbornly resistant to the immune checkpoint inhibitors that change the landscape of other human tumors. Here, we discuss recent discoveries on the specific populations of γδ T cells that infiltrate human gynecologic cancers, their anti-tumor activity, and the prospect of redirecting their effector function against tumor cells to develop a new generation of immunotherapies that extends beyond the traditional αß T cell-centric view of the field.

9.
Blood Adv ; 7(18): 5586-5602, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37531660

ABSTRACT

The pathogenesis of cutaneous T-cell lymphoma (CTCL) remains unclear. Using single-cell RNA or T-cell receptor (TCR) sequencing of 32 619 CD3+CD4+ and CD26+/CD7+ and 29 932 CD3+CD4+ and CD26-/CD7- lymphocytes from the peripheral blood of 7 patients with CTCL, coupled to single-cell ATAC-sequencing of 26,411 CD3+CD4+ and CD26+/CD7+ and 33 841 CD3+CD4+ and CD26-/CD7- lymphocytes, we show that tumor cells in Sézary syndrome and mycosis fungoides (MF) exhibit different phenotypes and trajectories of differentiation. When compared to MF, Sézary cells exhibit narrower repertoires of TCRs and exhibit clonal enrichment. Surprisingly, we identified ≥200 mutations in hematopoietic stem cells from multiple patients with Sézary syndrome. Mutations in key oncogenes were also present in peripheral Sézary cells, which also showed the hallmarks of recent thymic egression. Together our data suggest that CTCL arises from mutated lymphocyte progenitors that acquire TCRs in the thymus, which complete their malignant transformation in the periphery.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Mycosis Fungoides , Sezary Syndrome , Skin Neoplasms , Humans , Sezary Syndrome/genetics , Sezary Syndrome/pathology , Dipeptidyl Peptidase 4 , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Mycosis Fungoides/genetics , Mycosis Fungoides/pathology , Lymphoma, T-Cell, Cutaneous/genetics , Receptors, Antigen, T-Cell
10.
Mol Cancer Ther ; 21(7): 1184-1194, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35499393

ABSTRACT

Although chimeric antigen receptor (CAR)-expressing T cells have proven success in hematologic malignancies, their effectiveness in solid tumors has been largely unsuccessful thus far. We found that some olfactory receptors are expressed in a variety of solid tumors of different histologic subtypes, with a limited pattern of expression in normal tissues. Quantification of OR2H1 expression by qRT-PCR and Western blot analysis of 17 normal tissues, 82 ovarian cancers of various histologies, eight non-small cell lung cancers (NSCLCs), and 17 breast cancers demonstrated widespread OR2H1 expression in solid epithelial tumors with expression in normal human tissues limited to the testis. CAR T cells recognizing the extracellular domain of the olfactory receptor OR2H1 were generated with a targeting motif identified through the screening of a phage display library and demonstrated OR2H1-specific cytotoxic killing in vitro and in vivo, using tumor cells with spontaneous expression of variable OR2H1 levels. Importantly, recombinant OR2H1 IgG generated with the VH/VL sequences of the CAR construct specifically detected OR2H1 protein signal in 60 human lung cancers, 40 ovarian carcinomas, and 73 cholangiocarcinomas, at positivity rates comparable with mRNA expression and without OR2H1 staining in 58 normal tissues. CRISPR/Cas9-mediated ablation of OR2H1 confirmed targeting specificity of the CAR and the tumor-promoting role of OR2H1 in glucose metabolism. Therefore, T cells redirected against OR2H1-expressing tumor cells represent a promising therapy against a broad range of epithelial cancers, likely with an admissible toxicity profile.


Subject(s)
Lung Neoplasms , Neoplasms, Glandular and Epithelial , Ovarian Neoplasms , Receptors, Odorant , Female , Humans , Cell Line, Tumor , Immunotherapy, Adoptive , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Receptors, Odorant/metabolism , T-Lymphocytes
11.
Cancer Res ; 82(5): 859-871, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34949671

ABSTRACT

Recent studies suggest that B cells could play an important role in the tumor microenvironment. However, the role of humoral responses in endometrial cancer remains insufficiently investigated. Using a cohort of 107 patients with different histological subtypes of endometrial carcinoma, we evaluated the role of coordinated humoral and cellular adaptive immune responses in endometrial cancer. Concomitant accumulation of T, B, and plasma cells at tumor beds predicted better survival. However, only B-cell markers corresponded with prolonged survival specifically in high-grade endometrioid type and serous tumors. Immune protection was associated with class-switched IgA and, to a lesser extent, IgG. Expressions of polymeric immunoglobulin receptor (pIgR) by tumor cells and its occupancy by IgA were superior predictors of outcome and correlated with defects in methyl-directed DNA mismatch repair. Mechanistically, pIgR-dependent, antigen-independent IgA occupancy drove activation of inflammatory pathways associated with IFN and TNF signaling in tumor cells, along with apoptotic and endoplasmic reticulum stress pathways, while thwarting DNA repair mechanisms. Together, these findings suggest that coordinated humoral and cellular immune responses, characterized by IgA:pIgR interactions in tumor cells, determine the progression of human endometrial cancer as well as the potential for effective immunotherapies. SIGNIFICANCE: This study provides new insights into the crucial role of humoral immunity in human endometrial cancer, providing a rationale for designing novel immunotherapies against this prevalent malignancy. See related commentary by Osorio and Zamarin, p. 766.


Subject(s)
Endometrial Neoplasms , Receptors, Polymeric Immunoglobulin , B-Lymphocytes/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Immunity, Humoral , Immunoglobulin A/metabolism , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/metabolism , Tumor Microenvironment
12.
Cancer Cell ; 40(5): 545-557.e13, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35427494

ABSTRACT

Despite repeated associations between T cell infiltration and outcome, human ovarian cancer remains poorly responsive to immunotherapy. We report that the hallmarks of tumor recognition in ovarian cancer-infiltrating T cells are primarily restricted to tissue-resident memory (TRM) cells. Single-cell RNA/TCR/ATAC sequencing of 83,454 CD3+CD8+CD103+CD69+ TRM cells and immunohistochemistry of 122 high-grade serous ovarian cancers shows that only progenitor (TCF1low) tissue-resident T cells (TRMstem cells), but not recirculating TCF1+ T cells, predict ovarian cancer outcome. TRMstem cells arise from transitional recirculating T cells, which depends on antigen affinity/persistence, resulting in oligoclonal, trogocytic, effector lymphocytes that eventually become exhausted. Therefore, ovarian cancer is indeed an immunogenic disease, but that depends on ∼13% of CD8+ tumor-infiltrating T cells (∼3% of CD8+ clonotypes), which are primed against high-affinity antigens and maintain waves of effector TRM-like cells. Our results define the signature of relevant tumor-reactive T cells in human ovarian cancer, which could be applicable to other tumors with unideal mutational burden.


Subject(s)
Immunologic Memory , Ovarian Neoplasms , CD8-Positive T-Lymphocytes , Female , Humans , Lymphocytes, Tumor-Infiltrating , Memory T Cells
13.
J Clin Invest ; 131(3)2021 02 01.
Article in English | MEDLINE | ID: mdl-33270606

ABSTRACT

Cutaneous T cell lymphoma (CTCL) has a poorly understood etiology and no known cure. Using conditional knockout mice, we found that ablation of the genomic organizer special AT-rich sequence-binding protein 1 (Satb1) caused malignant transformation of mature, skin-homing, Notch-activated CD4+ and CD8+ T cells into progressively fatal lymphoma. Mechanistically, Satb1 restrained Stat5 phosphorylation and the expression of skin-homing chemokine receptors in mature T cells. Notably, methyltransferase-dependent epigenetic repression of SATB1 was universally found in human Sézary syndrome, but not in other peripheral T cell malignancies. H3K27 and H3K9 trimethylation occluded the SATB1 promoter in Sézary cells, while inhibition of SUV39H1/2 methyltransferases (unlike EZH2 inhibition) restored protective SATB1 expression and selectively abrogated the growth of primary Sézary cells more effectively than romidepsin. Therefore, inhibition of methyltransferases that silence SATB1 could address an unmet need for patients with mycosis fungoides/Sézary syndrome, a set of incurable diseases.


Subject(s)
Enzyme Inhibitors/pharmacology , Matrix Attachment Region Binding Proteins/metabolism , Methyltransferases/antagonists & inhibitors , Neoplasm Proteins , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Humans , Methyltransferases/metabolism , Mice , Mice, Transgenic , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Sezary Syndrome/metabolism , Sezary Syndrome/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
14.
Science ; 369(6506): 942-949, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32820120

ABSTRACT

Gamma delta (γδ) T cells infiltrate most human tumors, but current immunotherapies fail to exploit their in situ major histocompatibility complex-independent tumoricidal potential. Activation of γδ T cells can be elicited by butyrophilin and butyrophilin-like molecules that are structurally similar to the immunosuppressive B7 family members, yet how they regulate and coordinate αß and γδ T cell responses remains unknown. Here, we report that the butyrophilin BTN3A1 inhibits tumor-reactive αß T cell receptor activation by preventing segregation of N-glycosylated CD45 from the immune synapse. Notably, CD277-specific antibodies elicit coordinated restoration of αß T cell effector activity and BTN2A1-dependent γδ lymphocyte cytotoxicity against BTN3A1+ cancer cells, abrogating malignant progression. Targeting BTN3A1 therefore orchestrates cooperative killing of established tumors by αß and γδ T cells and may present a treatment strategy for tumors resistant to existing immunotherapies.


Subject(s)
Antigens, CD/immunology , Butyrophilins/antagonists & inhibitors , Butyrophilins/immunology , Intraepithelial Lymphocytes/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, CD/genetics , Butyrophilins/genetics , Female , Humans , Immunotherapy/methods , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Molecules ; 14(12): 4892-914, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-20032867

ABSTRACT

Phosphatidylserine (PS), a phospholipid predominantly found in the inner leaflet of eukaryotic cellular membranes, plays important roles in many biological processes. During apoptosis, the asymmetric distribution of phospholipids of the plasma membrane gets lost and PS is translocated to the outer leaflet of the plasma membrane. There, PS acts as one major "eat me" signal that ensures efficient recognition and uptake of apoptotic cells by phagocytes. PS recognition of activated phagocytes induces the secretion of anti-inflammatory cytokines like interleukin-10 and transforming grow factor-beta. Deficiencies in the clearance of apoptotic cells result in the occurrence of secondarily necrotic cells. The latter have lost the membrane integrity and release immune activating danger signals, which may induce inflammatory responses. Accumulation of dead cells containing nuclear autoantigens in sites of immune selection may provide survival signals for autoreactive B-cells. The production of antibodies against nuclear structures determines the initiation of chronic autoimmunity in systemic lupus erythematosus. Since PS on apoptotic cells is an important modulator of the immune response, natural occurring ligands for PS like annexinA5 have profound effects on immune responses against dead and dying cells, including tumour cells. In this review we will focus on the role of PS exposure in the clearance process of dead cells and its implications in clinical situations where apoptosis plays a relevant role, like in cancer, chronic autoimmunity, and infections. Relevance of other phospholipids during the apoptosis process is also discussed.


Subject(s)
Apoptosis/physiology , Phospholipids/physiology , Humans , Ligands , Phagocytes/immunology
16.
J Leukoc Biol ; 103(5): 799-805, 2018 05.
Article in English | MEDLINE | ID: mdl-29537705

ABSTRACT

Due to their cytotoxic activities, many anticancer drugs cause extensive damage to the intestinal mucosa and have antibiotic activities. Here, we show that cisplatin induces significant changes in the repertoire of intestinal commensal bacteria that exacerbate mucosal damage. Restoration of the microbiota through fecal-pellet gavage drives healing of cisplatin-induced intestinal damage. Bacterial translocation to the blood stream is correspondingly abrogated, resulting in a significant reduction in systemic inflammation, as evidenced by decreased serum IL-6 and reduced mobilization of granulocytes. Mechanistically, reversal of dysbiosis in response to fecal gavage results in the production of protective mucins and mobilization of CD11b+ myeloid cells to the intestinal mucosa, which promotes angiogenesis. Administration of Ruminococcus gnavus, a bacterial strain selectively depleted by cisplatin treatment, could only partially restore the integrity of the intestinal mucosa and reduce systemic inflammation, without measurable increases in the accumulation of mucin proteins. Together, our results indicate that reconstitution of the full repertoire of intestinal bacteria altered by cisplatin treatment accelerates healing of the intestinal epithelium and ameliorates systemic inflammation. Therefore, fecal microbiota transplant could paradoxically prevent life-threatening bacteremia in cancer patients treated with chemotherapy.


Subject(s)
Cisplatin/pharmacology , Dysbiosis/therapy , Fecal Microbiota Transplantation , Intestines/microbiology , Ovarian Neoplasms/drug therapy , Peritoneal Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Dysbiosis/mortality , Dysbiosis/pathology , Female , Gastrointestinal Microbiome , Intestines/drug effects , Intestines/pathology , Ovarian Neoplasms/microbiology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/microbiology , Peritoneal Neoplasms/pathology , Tumor Cells, Cultured
17.
Front Immunol ; 8: 504, 2017.
Article in English | MEDLINE | ID: mdl-28496447

ABSTRACT

INTRODUCTION: Many antitumor therapies induce apoptotic cell death in order to cause tumor regression. Paradoxically, apoptotic cells are also known to promote wound healing, cell proliferation, and tumor cell repopulation in multicellular organisms. We aimed to characterize the nature of the regenerative signals concentrated in the micromilieu of dead and dying cells. METHODS: Cultures of viable melanoma B16F10 cells, mouse fibroblasts, and primary human fibroblast-like synoviocytes (FLS) in the presence of dead and dying cells, their supernatants (SNs), or purified agonists and antagonists were used to evaluate the stimulation of proliferation. Viable cell quantification was performed by either flow cytometry of harvested cells or by crystal violet staining of adherent cells. High-performance liquid chromatography and liquid chromatography coupled with mass spectrometry of cell SNs were deployed to identify the nature of growth-promoting factors. Coimplantation of living cells in the presence of SNs collected from dead and dying cells and specific agonists was used to evaluate tumor growth in vivo. RESULTS: The stimulation of proliferation of few surviving cells by bystander dead cells was confirmed for melanoma cells, mouse fibroblasts, and primary FLS. We found that small soluble molecules present in the protein-free fraction of SNs of dead and dying cells were responsible for the promotion of proliferation. The nucleoside inosine released by dead and dying cells acting via adenosine receptors was identified as putative inducer of proliferation of surviving tumor cells after irradiation and heat treatment. CONCLUSION: Inosine released by dead and dying cells mediates tumor cell proliferation via purinergic receptors. Therapeutic strategies surmounting this pathway may help to reduce the rate of recurrence after radio- and chemotherapy.

18.
PLoS Negl Trop Dis ; 10(6): e0004786, 2016 06.
Article in English | MEDLINE | ID: mdl-27347876

ABSTRACT

Transmission-blocking (TB) vaccines are considered an important tool for malaria control and elimination. Among all the antigens characterized as TB vaccines against Plasmodium vivax, the ookinete surface proteins Pvs28 and Pvs25 are leading candidates. These proteins likely originated by a gene duplication event that took place before the radiation of the known Plasmodium species to primates. We report an evolutionary genetic analysis of a worldwide sample of pvs28 and pvs25 alleles. Our results show that both genes display low levels of genetic polymorphism when compared to the merozoite surface antigens AMA-1 and MSP-1; however, both ookinete antigens can be as polymorphic as other merozoite antigens such as MSP-8 and MSP-10. We found that parasite populations in Asia and the Americas are geographically differentiated with comparable levels of genetic diversity and specific amino acid replacements found only in the Americas. Furthermore, the observed variation was mainly accumulated in the EGF2- and EGF3-like domains for P. vivax in both proteins. This pattern was shared by other closely related non-human primate parasites such as Plasmodium cynomolgi, suggesting that it could be functionally important. In addition, examination with a suite of evolutionary genetic analyses indicated that the observed patterns are consistent with positive natural selection acting on Pvs28 and Pvs25 polymorphisms. The geographic pattern of genetic differentiation and the evidence for positive selection strongly suggest that the functional consequences of the observed polymorphism should be evaluated during development of TBVs that include Pvs25 and Pvs28.


Subject(s)
Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Plasmodium vivax/genetics , Protozoan Proteins/genetics , Evolution, Molecular , Gene Expression Regulation , Haplotypes , Humans , Malaria, Vivax/parasitology , Models, Molecular , Polymorphism, Genetic , Protein Conformation , Protozoan Proteins/metabolism , Selection, Genetic
19.
Front Immunol ; 5: 560, 2014.
Article in English | MEDLINE | ID: mdl-25426116

ABSTRACT

Large amounts of dead and dying cells are produced during cancer therapy and allograft rejection. Depending on the death pathway and stimuli involved, dying cells exhibit diverse features, resulting in defined physiological consequences for the host. It is not fully understood how dying and dead cells modulate the immune response of the host. To address this problem, different death stimuli were studied in B16F10 melanoma cells by regulated inducible transgene expression of the pro-apoptotic active forms of caspase-3 (revCasp-3), Bid (tBid), and the Mycobacterium tuberculosis-necrosis inducing toxin (CpnTCTD). The immune outcome elicited for each death stimulus was assessed by evaluating the allograft rejection of melanoma tumors implanted subcutaneously in BALB/c mice immunized with dying cells. Expression of all proteins efficiently killed cells in vitro (>90%) and displayed distinctive morphological and physiological features as assessed by multiparametric flow cytometry analysis. BALB/c mice immunized with allogeneic dying melanoma cells expressing revCasp-3 or CpnTCTD showed strong rejection of the allogeneic challenge. In contrast, mice immunized with cells dying either after expression of tBid or irradiation with UVB did not, suggesting an immunologically silent cell death. Surprisingly, immunogenic cell death induced by expression of revCasp-3 or CpnTCTD correlated with elevated intracellular reactive oxygen species (ROS) levels at the time point of immunization. Conversely, early mitochondrial dysfunction induced by tBid expression or UVB irradiation accounted for the absence of intracellular ROS accumulation at the time point of immunization. Although ROS inhibition in vitro was not sufficient to abrogate the immunogenicity in our allo-immunization model, we suggest that the point of ROS generation and its intracellular accumulation may be an important factor for its role as damage associated molecular pattern in the development of allogeneic responses.

20.
Autoimmunity ; 46(5): 329-35, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23173725

ABSTRACT

Killing tumor cells is a central goal of non-surgical cancer therapy and induction of apoptosis in the malignant cells is the major strategy used to achieve it. However, this may have serious drawbacks, since apoptotic cells reportedly induce immunological tumor tolerance and activate a tumor repopulation program. The debate on which type of cell death has the most beneficial therapeutic effects in cancer treatment is intense and controversial. Stringently regulated, doxycycline-inducible transgene expression systems trigger cell death in a defined manner, which might help us find answers to these problems. A conditional suicide switch established transiently in Jurkat T-cells was used to test a set of pro-apoptotic proteins for their potency to induce cell death. The activated forms of caspase-3 (revCasp-3) and Bid (tBid) were very effective and, therefore, analyzed after stable integration into human leukemia and murine melanoma cell lines. Expression of either protein resulted in more than 95% cell death, but with strongly different kinetics. 85% cell death was observed if tBid and revCasp-3 were expressed for 4-6 hours and 18-24 hours, respectively. The human cell lines expressing these suicide switches can serve as cancer models in xeno-transplanted mice, the corresponding murine cell lines allow syngeneic and allogeneic murine cancer models to be established.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis/immunology , Amino Acid Sequence , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Cell Death/genetics , Cell Death/immunology , Clone Cells , Disease Models, Animal , Humans , Jurkat Cells , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Molecular Sequence Data , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL