Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Leukoc Biol ; 112(4): 733-744, 2022 10.
Article in English | MEDLINE | ID: mdl-35916053

ABSTRACT

Opioid use has negative effects on immune responses and may impair immune reconstitution in persons living with HIV (PLWH) infection undergoing antiretroviral treatment (ART). The effects of treatment with µ opioid receptor (MOR) agonists (e.g., methadone, MET) and antagonists (e.g., naltrexone, NTX) on immune reconstitution and immune activation in ART-suppressed PLWH have not been assessed in-depth. We studied the effects of methadone or naltrexone on measures of immune reconstitution and immune activation in a cross-sectional community cohort of 30 HIV-infected individuals receiving suppressive ART and medications for opioid use disorder (MOUD) (12 MET, 8 NTX and 10 controls). Plasma markers of inflammation and immune activation were measured using ELISA, Luminex, or Simoa. Plasma IgG glycosylation was assessed using capillary electrophoresis. Cell subsets and activation were studied using whole blood flow cytometry. Individuals in the MET group, but no in the NTX group, had higher plasma levels of inflammation and immune activation markers than controls. These markers include soluble CD14 (an independent predictor of morbidity and mortality during HIV infection), proinflammatory cytokines, and proinflammatory IgG glycans. This effect was independent of time on treatment. Our results indicate that methadone-based MOUD regimens may sustain immune activation and inflammation in ART-treated HIV-infected individuals. Our pilot study provides the foundation and rationale for future longitudinal functional studies of the impact of MOUD regimens on immune reconstitution and residual activation after ART-mediated suppression.


Subject(s)
HIV Infections , Analgesics, Opioid/therapeutic use , Cross-Sectional Studies , Cytokines , Humans , Immunoglobulin G , Inflammation/complications , Lipopolysaccharide Receptors , Methadone/therapeutic use , Naltrexone/therapeutic use , Pilot Projects , Receptors, Opioid, mu
2.
Eur J Immunol ; 51(10): 2441-2451, 2021 10.
Article in English | MEDLINE | ID: mdl-34287839

ABSTRACT

Inhibition of the BCL6 BTB domain results in killing Diffuse Large B-cell Lymphoma (DLBL) cells, reducing the T-cell dependent germinal center (GC) reaction in mice, and reversing GC hyperplasia in nonhuman primates. The available BCL6 BTB-specific inhibitors are poorly water soluble, thus, limiting their absorption in vivo and our understanding of therapeutic strategy targeting GC. We synthesized a prodrug (AP-4-287) from a potent BCL6 BTB inhibitor (FX1) with improved aqueous solubility and pharmacokinetics (PK) in mice. We also evaluated its in vivo biological activity on humoral immune responses using the sheep red blood cells (SRBC)-vaccination mouse model. AP-4-287 had a significant higher aqueous solubility and was readily converted to FX1 in vivo after intraperitoneally (i.p.) administration, but a shorter half-life in vivo. Importantly, AP-4-287 treatment led to a reversible effect on (1) the reduction in the frequency of splenic Ki67+ CD4+ T cells, Tfh cells, and GC B cells; (2) lower GC formation following vaccination; and (3) a decrease in the titers of antigen-specific IgG and IgM antibodies. Our study advances the preclinical development of drug targeting BCL6 BTB domain for the treatment of diseases that are associated with abnormal BCL6 elevation.


Subject(s)
Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Protein Interaction Domains and Motifs/drug effects , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibody Formation/drug effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Chemistry Techniques, Synthetic , Germinal Center/drug effects , Germinal Center/immunology , Germinal Center/metabolism , Immunity, Humoral/drug effects , Immunomodulation/drug effects , Indoles/administration & dosage , Indoles/chemical synthesis , Indoles/pharmacokinetics , Mice , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Proto-Oncogene Proteins c-bcl-6/chemistry , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thiazolidinediones/administration & dosage , Thiazolidinediones/chemical synthesis , Thiazolidinediones/pharmacokinetics
3.
J Immunol ; 204(4): 990-1000, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31900334

ABSTRACT

In the metastasis-targeted organs, angiogenesis is essential for the progression of dormant micrometastases to rapidly growing and clinically overt lesions. However, we observed changes suggesting angiogenic switching in the mouse lungs prior to arrival of tumor cells (i.e., in the premetastatic niche) in the models of breast carcinoma. This angiogenic switching appears to be caused by myeloid-derived suppressor cells recruited to the premetastatic lungs through complement C5a receptor 1 signaling. These myeloid cells are known to secrete several proangiogenic factors in tumors, including IL-1ß and matrix metalloproteinase-9, and we found upregulation of these genes in the premetastatic lungs. Blockade of C5a receptor 1 synergized with antiangiogenic Listeria monocytogenes-based vaccines to decrease the lung metastatic burden by reducing vascular density and improving antitumor immunity in the lungs. This was mediated even when growth of primary breast tumors was not affected by these treatments. This work provides initial evidence that angiogenesis contributes to the premetastatic niche in rapidly progressing cancers and that inhibiting this process through immunotherapy is beneficial for reducing or even preventing metastasis.


Subject(s)
Cancer Vaccines/administration & dosage , Lung Neoplasms/therapy , Mammary Neoplasms, Experimental/therapy , Myeloid-Derived Suppressor Cells/immunology , Neovascularization, Pathologic/therapy , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Cell Line, Tumor , Combined Modality Therapy/methods , Complement C5a/immunology , Complement C5a/metabolism , Female , Humans , Immunotherapy/methods , Listeria monocytogenes/immunology , Lung/blood supply , Lung/immunology , Lung/pathology , Lung Neoplasms/blood supply , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Metastasis/immunology , Neoplasm Metastasis/therapy , Neovascularization, Pathologic/immunology , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/metabolism , Tumor Microenvironment/immunology
4.
Mol Cancer Res ; 17(8): 1639-1651, 2019 08.
Article in English | MEDLINE | ID: mdl-31088907

ABSTRACT

We discovered that 90.3% of patients with angiomyolipomas, lymphangioleiomyomatosis (LAM), and tuberous sclerosis complex (TSC) carry the arginine variant of codon 72 (R72) of TP53 and that R72 increases the risk for angiomyolipoma. R72 transactivates NOTCH1 and NODAL better than the proline variant of codon 72 (P72); therefore, the expression of NOTCH1 and NODAL is increased in angiomyolipoma cells that carry R72. The loss of Tp53 and Tsc1 within nestin-expressing cells in mice resulted in the development of renal cell carcinomas (RCC) with high Notch1 and Nodal expression, suggesting that similar downstream mechanisms contribute to tumorigenesis as a result of p53 loss in mice and p53 polymorphism in humans. The loss of murine Tp53 or expression of human R72 contributes to tumorigenesis via enhancing epithelial-to-mesenchymal transition and motility of tumor cells through the Notch and Nodal pathways. IMPLICATIONS: This work revealed unexpected contributions of the p53 polymorphism to the pathogenesis of TSC and established signaling alterations caused by this polymorphism as a target for therapy. We found that the codon 72 TP53 polymorphism contributes to TSC-associated tumorigenesis via Notch and Nodal signaling.


Subject(s)
Carcinogenesis/pathology , Nodal Protein/metabolism , Polymorphism, Single Nucleotide , Receptor, Notch1/metabolism , Tuberous Sclerosis/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/physiology , Angiomyolipoma/genetics , Angiomyolipoma/metabolism , Angiomyolipoma/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mutation , Nodal Protein/genetics , Receptor, Notch1/genetics , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
5.
Carcinogenesis ; 40(2): 225-233, 2019 04 29.
Article in English | MEDLINE | ID: mdl-30364933

ABSTRACT

Identification of factors associated with human papillomavirus (HPV) cervical histopathology or recurrence/relapse following loop electrosurgical excision procedure (LEEP) would allow for better management of the disease. We investigated whether gene signatures could (i) associate with HPV cervical histopathology and (ii) identify women with post-LEEP disease recurrence/relapse. Gene array analysis was performed on paraffin-embedded cervical tissue-isolated RNA from two cross-sectional cohorts of antiretroviral therapy (ART)-suppressed HIV+HPV+ coinfected women: (i) 55 women in South Africa recruited into three groups: high risk (HR) (-) (n = 16) and HR (+) (n = 15) HPV without cervical histopathology and HR (+) HPV with cervical intraepithelial neoplasia (CIN) grade 1/2/3 (n = 24), (ii) 28 women in Botswana with CIN2/3 treated with LEEP 12-month prior to recruitment and presenting with (n = 13) and without (n = 15) lesion recurrence/relapse (tissue was analyzed at first LEEP). Three distinct gene expression signatures identified were able to segregate: (i) HR+ HPV and CIN1/2/3, (ii) HR HPV-free and cervical histopathology-free and (iii) HR+ HPV and cervical histopathology-free. Immune activation and neoplasia-associated genes (n = 272 genes; e.g. IL-1A, IL-8, TCAM1, POU4F1, MCM2, SMC1B, CXCL6, MMP12) were a feature of cancer precursor dysplasia within HR HPV infection. No difference in LEEP tissue gene expression was detected between women with or without recurrence/relapse. In conclusion, distinctive gene signatures were associated with presence of cervical histopathology in tissues from ART-suppressed HIV+/HPV+ coinfected women. Lack of detection of LEEP tissue gene signature able to segregate subsequent post-LEEP disease recurrence/relapse indicates additional factors independent of local gene expression as determinants of recurrence/relapse.


Subject(s)
Cervix Uteri/pathology , Gene Expression/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Adult , Anti-Retroviral Agents/pharmacology , Cervix Uteri/drug effects , Cervix Uteri/virology , Cross-Sectional Studies , Female , Gene Expression/drug effects , Gene Expression Profiling/methods , HIV Infections/drug therapy , HIV Infections/pathology , HIV Infections/virology , Humans , Neoplasm Recurrence, Local/etiology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/virology , Papillomaviridae/drug effects , Papillomavirus Infections/drug therapy , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/drug therapy , Uterine Cervical Dysplasia/genetics , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virology
6.
Sci Transl Med ; 10(437)2018 04 18.
Article in English | MEDLINE | ID: mdl-29669853

ABSTRACT

The persistence of HIV reservoirs, including latently infected, resting CD4+ T cells, is the major obstacle to cure HIV infection. CD32a expression was recently reported to mark CD4+ T cells harboring a replication-competent HIV reservoir during antiretroviral therapy (ART) suppression. We aimed to determine whether CD32 expression marks HIV latently or transcriptionally active infected CD4+ T cells. Using peripheral blood and lymphoid tissue of ART-treated HIV+ or SIV+ subjects, we found that most of the circulating memory CD32+ CD4+ T cells expressed markers of activation, including CD69, HLA-DR, CD25, CD38, and Ki67, and bore a TH2 phenotype as defined by CXCR3, CCR4, and CCR6. CD32 expression did not selectively enrich for HIV- or SIV-infected CD4+ T cells in peripheral blood or lymphoid tissue; isolated CD32+ resting CD4+ T cells accounted for less than 3% of the total HIV DNA in CD4+ T cells. Cell-associated HIV DNA and RNA loads in CD4+ T cells positively correlated with the frequency of CD32+ CD69+ CD4+ T cells but not with CD32 expression on resting CD4+ T cells. Using RNA fluorescence in situ hybridization, CD32 coexpression with HIV RNA or p24 was detected after in vitro HIV infection (peripheral blood mononuclear cell and tissue) and in vivo within lymph node tissue from HIV-infected individuals. Together, these results indicate that CD32 is not a marker of resting CD4+ T cells or of enriched HIV DNA-positive cells after ART; rather, CD32 is predominately expressed on a subset of activated CD4+ T cells enriched for transcriptionally active HIV after long-term ART.


Subject(s)
HIV Infections/metabolism , Receptors, IgG/metabolism , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/metabolism , HIV Infections/drug therapy , Humans , In Vitro Techniques , Lymphocytes/metabolism , Receptors, CCR4/metabolism , Receptors, CCR6/metabolism , Receptors, CXCR3/metabolism
7.
J Leukoc Biol ; 104(3): 461-471, 2018 09.
Article in English | MEDLINE | ID: mdl-29633346

ABSTRACT

Global antibody glycosylation is dynamic and plays critical roles in shaping different immunological outcomes and direct antibody functionality during HIV infection. However, the relevance of global antibody or plasma glycosylation patterns to HIV persistence after antiretroviral therapy (ART) has not been characterized. First, we compared glycomes of total plasma and isolated immunoglobulin G (IgG) from HIV+ ART-suppressed, HIV+ viremic, and HIV-negative individuals. Second, in ART-suppressed individuals, we examined the associations between glycomes and (1) levels of cell-associated HIV DNA and RNA in PBMCs and isolated CD4+ T cells, (2) CD4 count and CD4%, and (3) expression of CD4+ T-cell activation markers. HIV infection is associated with persistent alterations in the IgG glycome including decreased levels of disialylated glycans, which is associated with a lower anti-inflammatory activity, and increased levels of fucosylated glycans, which is associated with lower antibody-dependent cell-mediated cytotoxicity (ADCC). We also show that levels of certain mono- and digalactosylated nonfucosylated glycomic traits (A2G1, A2G2, and A2BG2), which have been reported to be associated with higher ADCC and higher anti-inflammatory activities, exhibit significant negative correlations with levels of cell-associated total HIV DNA and HIV RNA in ART-suppressed individuals. Finally, levels of certain circulating anti-inflammatory glycans are associated with higher levels of CD4 T cells and lower levels of T-cell activation. Our findings represent the first proof-of-concept evidence that glycomic alterations, known to be associated with differential states of inflammation and ADCC activities, are also associated with levels of HIV persistence in the setting of ART suppression.


Subject(s)
Anti-HIV Agents/therapeutic use , Galactose/metabolism , HIV Infections/drug therapy , HIV Infections/metabolism , Immunoglobulin G/metabolism , Adult , CD4-Positive T-Lymphocytes , Humans , Male , Plasma/metabolism , Viral Load/drug effects , Viremia/drug therapy , Viremia/metabolism
8.
Nat Commun ; 8(1): 1848, 2017 11 29.
Article in English | MEDLINE | ID: mdl-29184052

ABSTRACT

Differentiation abnormalities are a hallmark of tuberous sclerosis complex (TSC) manifestations; however, the genesis of these abnormalities remains unclear. Here we report on mechanisms controlling the multi-lineage, early neuronal progenitor and neural stem-like cell characteristics of lymphangioleiomyomatosis (LAM) and angiomyolipoma cells. These mechanisms include the activation of a previously unreported Rheb-Notch-Rheb regulatory loop, in which the cyclic binding of Notch1 to the Notch-responsive elements (NREs) on the Rheb promoter is a key event. This binding induces the transactivation of Rheb. The identified NRE2 and NRE3 on the Rheb promoter are important to Notch-dependent promoter activity. Notch cooperates with Rheb to block cell differentiation via similar mechanisms in mouse models of TSC. Cell-specific loss of Tsc1 within nestin-expressing cells in adult mice leads to the formation of kidney cysts, renal intraepithelial neoplasia, and invasive papillary renal carcinoma.


Subject(s)
Angiomyolipoma/pathology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/pathology , Ras Homolog Enriched in Brain Protein/metabolism , Receptor, Notch1/metabolism , Angiomyolipoma/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Female , Humans , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/metabolism , Male , Mice, SCID , Mice, Transgenic , Neural Crest/metabolism , Neural Crest/pathology , Promoter Regions, Genetic , Ras Homolog Enriched in Brain Protein/genetics , Receptor, Notch1/genetics , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
9.
J Immunol ; 198(7): 2989-2999, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28228558

ABSTRACT

Relatively little is known about factors that initiate immunosuppression in tumors and act at the interface between tumor cells and host cells. In this article, we report novel immunosuppressive properties of the ribosomal protein S19 (RPS19), which is upregulated in human breast and ovarian cancer cells and released from apoptotic tumor cells, whereupon it interacts with the complement C5a receptor 1 expressed on tumor infiltrating myeloid-derived suppressor cells. This interaction promotes tumor growth by facilitating recruitment of these cells to tumors. RPS19 also induces the production of immunosuppressive cytokines, including TGF-ß, by myeloid-derived suppressor cells in tumor-draining lymph nodes, leading to T cell responses skewed toward Th2 phenotypes. RPS19 promotes generation of regulatory T cells while reducing infiltration of CD8+ T cells into tumors. Reducing RPS19 in tumor cells or blocking the C5a receptor 1-RPS19 interaction decreases RPS19-mediated immunosuppression, impairs tumor growth, and delays the development of tumors in a transgenic model of breast cancer. This work provides initial preclinical evidence for targeting RPS19 for anticancer therapy enhancing antitumor T cell responses.


Subject(s)
Myeloid-Derived Suppressor Cells/immunology , Neoplasms, Experimental/immunology , Receptor, Anaphylatoxin C5a/immunology , Ribosomal Proteins/immunology , Animals , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , Immunoprecipitation , Mice , T-Lymphocytes/immunology
10.
J Vis Exp ; (112)2016 06 26.
Article in English | MEDLINE | ID: mdl-27403530

ABSTRACT

This paper describes the application of the syngeneic model of breast cancer (4T1) to the studies on a role of pulmonary alveolar macrophages in cancer metastasis. The 4T1 cells expressing GFP in combination with imaging and confocal microscopy are used to monitor tumor growth, track metastasizing tumor cells, and quantify the metastatic burden. These approaches are supplemented by digital histopathology that allows the automated and unbiased quantification of metastases. In this method the routinely prepared histological lung sections, which are stained with hematoxylin and eosin, are scanned and converted to the digital slides that are then analyzed by the self-trained pattern recognition software. In addition, we describe the flow cytometry approaches with the use of multiple cell surface markers to identify alveolar macrophages in the lungs. To determine impact of alveolar macrophages on metastases and antitumor immunity these cells are depleted with the clodronate-containing liposomes administrated intranasally to tumor-bearing mice. This approach leads to the specific and efficient depletion of this cell population as confirmed by flow cytometry. Tumor volumes and lung metastases are evaluated in mice depleted of alveolar macrophages, to determine the role of these cells in the metastatic progression of breast cancer.


Subject(s)
Breast Neoplasms , Macrophages, Alveolar , Animals , Liposomes , Lung , Lung Neoplasms , Mice
11.
J Immunol ; 194(11): 5529-38, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25911761

ABSTRACT

In contrast to tumor-associated macrophages, myeloid-derived suppressor cells, or inflammatory monocytes, functions of tissue resident macrophages, including alveolar macrophages (AM), in cancer were not well studied. Using a mouse model of breast cancer, we show that AM promote cancer metastasis to the lungs by suppressing antitumor T cells in this organ. AM accumulated in the premetastatic lungs through complement C5a receptor-mediated proliferation but not through recruitment from the circulation. AM preconditioned by breast tumors inhibited Th1 and favored generation of Th2 cells that had lower tumoricidal activity than Th1 cells. In addition, AM reduced the number and maturation of lung dendritic cells by regulating TGF-ß in the lung environment. Depletion of AM reversed immunosuppression imposed by these cells and strengthened local Th1 responses, which significantly reduced lung metastatic burden. C5a receptor deficiency, which also lessens myeloid-derived suppressor cells in the premetastatic niche, synergized with the depletion of AM in preventing metastasis, leading to protection of mice from lung metastases. This study identifies AM as a new component of the premetastatic niche, which is harnessed by tumors to impose immunosuppression, and as a new target for cancer immunotherapies to eliminate or reduce metastasis. Because the lungs are the most common target for hematogenous metastasis, this research offers a plausible explanation for susceptibility of the lungs to cancer metastasis.


Subject(s)
Lung Neoplasms/immunology , Lung Neoplasms/secondary , Macrophages, Alveolar/immunology , Mammary Neoplasms, Experimental/pathology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Line , Cell Proliferation , Dendritic Cells/immunology , Female , Humans , Lung/immunology , Macrophages, Alveolar/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptor, Anaphylatoxin C5a/immunology , Transforming Growth Factor beta/biosynthesis , Tumor Microenvironment/immunology
12.
Cancer Res ; 74(13): 3454-65, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24786787

ABSTRACT

The impact of complement on cancer metastasis has not been well studied. In this report, we demonstrate in a preclinical mouse model of breast cancer that the complement anaphylatoxin C5a receptor (C5aR) facilitates metastasis by suppressing effector CD8(+) and CD4(+) T-cell responses in the lungs. Mechanisms of this suppression involve recruitment of immature myeloid cells to the lungs and regulation of TGFß and IL10 production in these cells. TGFß and IL10 favored generation of T regulatory cells (Treg) and Th2-oriented responses that rendered CD8(+) T cells dysfunctional. Importantly, pharmacologic blockade of C5aR or its genetic ablation in C5aR-deficient mice were sufficient to reduce lung metastases. Depletion of CD8(+) T cells abolished this beneficial effect, suggesting that CD8(+) T cells were responsible for the effects of C5aR inhibition. In contrast to previous findings, we observed that C5aR signaling promoted Treg generation and suppressed T-cell responses in organs where metastases arose. Overall, our findings indicated that the immunomodulatory functions of C5aR are highly context dependent. Furthermore, they offered proof-of-concept for complement-based immunotherapies to prevent or reduce cancer metastasis.


Subject(s)
Mammary Neoplasms, Experimental/immunology , Receptor, Anaphylatoxin C5a/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Complement C5a/immunology , Female , Immunosuppression Therapy , Interleukin-10/biosynthesis , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lung/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Metastasis , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Transforming Growth Factor beta/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...