Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jorge Schettini is active.

Publication


Featured researches published by Jorge Schettini.


Journal of Immunology | 2008

MUC1 Enhances Tumor Progression and Contributes Toward Immunosuppression in a Mouse Model of Spontaneous Pancreatic Adenocarcinoma

Teresa L. Tinder; Durai B. Subramani; Gargi D. Basu; Judy M. Bradley; Jorge Schettini; Arefayene Million; Todd C. Skaar

MUC1, a membrane tethered mucin glycoprotein, is overexpressed and aberrantly glycosylated in >80% of human ductal pancreatic adenocarcinoma. However, the role of MUC1 in pancreatic cancer has been elusive, partly due to the lack of an appropriate model. We report the characterization of a novel mouse model that expresses human MUC1 as a self molecule (PDA.MUC1 mice). Pancreatic tumors arise in an appropriate MUC1-tolerant background within an immune-competent host. Significant enhancement in the development of pancreatic intraepithelial preneoplastic lesions and progression to adenocarcinoma is observed in PDA.MUC1 mice, possibly due to increased proliferation. Tumors from PDA.MUC1 mice express higher levels of cyclooxygenase-2 and IDO compared with PDA mice lacking MUC1, especially during early stages of tumor development. The increased proinflammatory milieu correlates with an increased percentage of regulatory T cells and myeloid suppressor cells in the pancreatic tumor and tumor draining lymph nodes. Data shows that during pancreatic cancer progression, MUC1-mediated mechanisms enhance the onset and progression of the disease, which in turn regulate the immune responses. Thus, the mouse model is ideally suited for testing novel chemopreventive and therapeutic strategies against pancreatic cancer.


Cancer Research | 2011

Pancreatic Ductal Adenocarcinoma Mice Lacking Mucin 1 Have a Profound Defect in Tumor Growth and Metastasis

Dahlia M. Besmer; Jennifer M. Curry; Lopamudra Das Roy; Teresa L. Tinder; Mahnaz Sahraei; Jorge Schettini; Sun-Il Hwang; Yong Y Lee; Sandra J. Gendler

MUC1 is overexpressed and aberrantly glycosylated in more than 60% of pancreatic ductal adenocarcinomas. The functional role of MUC1 in pancreatic cancer has yet to be fully elucidated due to a dearth of appropriate models. In this study, we have generated mouse models that spontaneously develop pancreatic ductal adenocarcinoma (KC), which are either Muc1-null (KCKO) or express human MUC1 (KCM). We show that KCKO mice have significantly slower tumor progression and rates of secondary metastasis, compared with both KC and KCM. Cell lines derived from KCKO tumors have significantly less tumorigenic capacity compared with cells from KCM tumors. Therefore, mice with KCKO tumors had a significant survival benefit compared with mice with KCM tumors. In vitro, KCKO cells have reduced proliferation and invasion and failed to respond to epidermal growth factor, platelet-derived growth factor, or matrix metalloproteinase 9. Further, significantly less KCKO cells entered the G(2)-M phase of the cell cycle compared with the KCM cells. Proteomics and Western blotting analysis revealed a complete loss of cdc-25c expression, phosphorylation of mitogen-activated protein kinase (MAPK), as well as a significant decrease in nestin and tubulin-α2 chain expression in KCKO cells. Treatment with a MEK1/2 inhibitor, U0126, abrogated the enhanced proliferation of the KCM cells but had minimal effect on KCKO cells, suggesting that MUC1 is necessary for MAPK activity and oncogenic signaling. This is the first study to utilize a Muc1-null PDA mouse to fully elucidate the oncogenic role of MUC1, both in vivo and in vitro.


Breast Cancer Research | 2009

Breast cancer-associated metastasis is significantly increased in a model of autoimmune arthritis

Lopamudra Das Roy; Latha B. Pathangey; Teresa L. Tinder; Jorge Schettini; Helen E. Gruber

IntroductionSites of chronic inflammation are often associated with the establishment and growth of various malignancies including breast cancer. A common inflammatory condition in humans is autoimmune arthritis (AA) that causes inflammation and deformity of the joints. Other systemic effects associated with arthritis include increased cellular infiltration and inflammation of the lungs. Several studies have reported statistically significant risk ratios between AA and breast cancer. Despite this knowledge, available for a decade, it has never been questioned if the site of chronic inflammation linked to AA creates a milieu that attracts tumor cells to home and grow in the inflamed bones and lungs which are frequent sites of breast cancer metastasis.MethodsTo determine if chronic inflammation induced by autoimmune arthritis contributes to increased breast cancer-associated metastasis, we generated mammary gland tumors in SKG mice that were genetically prone to develop AA. Two breast cancer cell lines, one highly metastatic (4T1) and the other non-metastatic (TUBO) were used to generate the tumors in the mammary fat pad. Lung and bone metastasis and the associated inflammatory milieu were evaluated in the arthritic versus the non-arthritic mice.ResultsWe report a three-fold increase in lung metastasis and a significant increase in the incidence of bone metastasis in the pro-arthritic and arthritic mice compared to non-arthritic control mice. We also report that the metastatic breast cancer cells augment the severity of arthritis resulting in a vicious cycle that increases both bone destruction and metastasis. Enhanced neutrophilic and granulocytic infiltration in lungs and bone of the pro-arthritic and arthritic mice and subsequent increase in circulating levels of proinflammatory cytokines, such as macrophage colony stimulating factor (M-CSF), interleukin-17 (IL-17), interleukin-6 (IL-6), vascular endothelial growth factor (VEGF), and tumor necrosis factor-alpha (TNF-alpha) may contribute to the increased metastasis. Treatment with anti-IL17 + celecoxib, an anti-inflammatory drug completely abrogated the development of metastasis and significantly reduced the primary tumor burden.ConclusionsThe data clearly has important clinical implications for patients diagnosed with metastatic breast cancer, especially with regards to the prognosis and treatment options.


Clinical & Developmental Immunology | 2008

Physiological Role of Plasmacytoid Dendritic Cells and Their Potential Use in Cancer Immunity

Jorge Schettini

Dendritic cells (DCs) play a pivotal role in the control of innate and adaptive immune responses. They are a heterogeneous cell population, where plasmacytoid dendritic cells (pDCs) are a unique subset capable of secreting high levels of type I IFNs. It has been demonstrated that pDCs can coordinate events during the course of viral infection, atopy, autoimmune diseases, and cancer. Therefore, pDC, as a main source of type I IFN, is an attractive target for therapeutic manipulations of the immune system to elicit a powerful immune response against tumor antigens in combination with other therapies. The therapeutic vaccination with antigen-pulsed DCs has shown a limited efficacy to generate an effective long-lasting immune response against tumor cells. A rational manipulation and design of vaccines which could include DC subsets outside “Langerhans cell paradigm” might allow us to improve the therapeutic approaches for cancer patients.


BMC Cancer | 2014

Systemic neutralization of IL-17A significantly reduces breast cancer associated metastasis in arthritic mice by reducing CXCL12/SDF-1 expression in the metastatic niches

Lopamudra Das Roy; Mahnaz Sahraei; Jorge Schettini; Helen E. Gruber; Dahlia M. Besmer

BackgroundIL-17A is a pro-inflammatory cytokine that is normally associated with autoimmune arthritis and other pro-inflammatory conditions. Recently, IL-17A has emerged as a critical factor in enhancing breast cancer (BC)-associated metastases. We generated immune competent arthritic mouse models that develop spontaneous BC-associated bone and lung metastasis. Using these models, we have previously shown that neutralization of IL-17A resulted in significant reduction in metastasis. However, the underlying mechanism/s remains unknown.MethodsWe have utilized two previously published mouse models for this study: 1) the pro-arthritic mouse model (designated SKG) injected with metastatic BC cell line (4T1) in the mammary fat pad, and 2) the PyV MT mice that develop spontaneous mammary gland tumors injected with type II collagen to induce autoimmune arthritis. Mice were treated with anti-IL-17A neutralizing antibody and monitored for metastasis and assessed for pro-inflammatory cytokines and chemokines associated with BC-associated metastasis.ResultsWe first corroborate our previous finding that in vivo neutralization of IL-17A significantly reduced metastasis to the bones and lungs in both models. Next, we report that treatment with anti-IL17A antibody significantly reduced the expression of a key chemokine, CXCL12 (also known as stromal derived factor-1 (SDF - 1)) in the bones and lungs of treated mice. CXCL12 is a ligand for CXCR4 (expressed on BC cells) and their interaction is known to be critical for metastasis. Interestingly, levels of CXCR4 in the tumor remained unchanged with treatment. Consequently, protein lysates derived from the bones and lungs of treated mice were significantly less chemotactic for the BC cells than lysates from untreated mice; and addition of exogenous SDF-1 to the lysates from treated mice completely restored BC cell migration. In addition, cytokines such as IL-6 and M-CSF were significantly reduced in the lung and bone lysates following treatment. The data presented suggests that systemic neutralization of IL-17A can block the CXCR4/SDF-1 signaling pathway by reducing the expression of SDF-1 in the metastatic niches and significantly reducing metastasis in both mouse models.ConclusionIn our model, neutralization of IL-17A regulates SDF-1 expression in the metastatic niches either directly or indirectly via reducing levels of IL-6 and M-CSF.


Cancer Immunology, Immunotherapy | 2012

Intratumoral delivery of CpG-conjugated anti-MUC1 antibody enhances NK cell anti-tumor activity.

Jorge Schettini; Amritha Kidiyoor; Dahlia M. Besmer; Teresa L. Tinder; Lopamudra Das Roy; Joseph Lustgarten; Sandra J. Gendler

Monoclonal antibodies (mAbs) against tumor-associated antigens are useful anticancer agents. Antibody-dependent cellular cytotoxicity (ADCC) is one of the major mechanisms responsible for initiating natural killer cell (NK)-mediated killing of tumors. However, the regulation of ADCC via NK cells is poorly understood. We have investigated the cytolytic activity of NK cells against pancreatic cancer cells that were coated with an antibody directed against the human tumor antigen, Mucin-1 designated HMFG-2, either alone or conjugated to CpG oligodeoxynucleotide (CpG ODN). Conjugated antibodies were tested for their ability to elicit ADCC in vitro and in vivo against pancreatic cancer cells. NK cells cultured in the presence of immobilized CpG ODN, HMFG-2 Ab, or CpG ODN-conjugated HMFG-2 Ab were able to up-regulate perforin similarly. Interestingly, a significant higher ADCC was observed when CpG ODN-conjugated HMFG-2-coated tumor cells were co-cultured with NK cells compared to unconjugated HMFG-2 Ab or CpG ODN alone. Moreover, MyD88-deficient NK cells can perform ADCC in vitro. Furthermore, intratumoral injections of CpG ODN-conjugated HMFG-2 induced a significant reduction in tumor burden in vivo in an established model of pancreatic tumor in nude mice compared to CpG ODN or the HMFG-2 alone. Depletion of macrophages or NK cells before treatment confirmed that both cells were required for the anti-tumor response in vivo. Results also suggest that CpG ODN and HMFG-2 Ab could be sensed by NK cells on the mAb-coated tumor cells triggering enhanced ADCC in vitro and in vivo.


Frontiers in Immunology | 2014

Pancreatic Cancer Cells Isolated from Muc1-Null Tumors Favor the Generation of a Mature Less Suppressive MDSC Population

Amritha Kidiyoor; Jorge Schettini; Dahlia M. Besmer; Stephen L. Rego; Sritama Nath; Jennifer M. Curry; Lopamudra Das Roy; Didier Dréau

Mucin 1 (MUC1) is a transmembrane mucin glycoprotein that is over-expressed and aberrantly glycosylated in >80% of human pancreatic ductal adenocarcinoma (PDA) and is associated with poor prognosis. To understand the role of MUC1 in PDA, we have recently developed two mouse models of spontaneous PDA, one that expresses full-length human MUC1 transgene (KCM mice) and one that is null for MUC1 (KCKO mice). We have previously reported that KCM mice express high levels of myeloid derived suppressor cells (MDSCs) in their tumors and develop highly aggressive PDA. To further understand the underlying mechanism for high MDSC levels in KCM-tumors, we generated primary cell lines from KCM and KCKO-tumors. In this study, we report that MDSCs derived using KCM cells express significantly higher levels of arginase 1 and inducible nitric oxide synthase (markers associated with immune suppression) and lower levels of CD115 (a marker associated with maturation of myeloid cells) as compared to KCKO-derived MDSCs. Functionally, KCM-derived MDSCs secrete significantly higher levels of urea and nitric oxide (NO) when co-cultured with normal splenic cells as compared to KCKO-derived MDSCs. Data indicates that KCM-derived MDSCs remain immature and are more suppressive as compared to KCKO-derived MDSCs. This was further corroborated in vivo where MDSCs isolated from KCM-tumor-bearing mice retained their immature state and were highly suppressive as compared to MDSCs derived from KCKO-tumor-bearing mice. Finally, we show that KCM cells secrete significantly higher levels of prostaglandin E2 (PGE2), a COX-2 metabolite and a known driver of suppressive MDSCs as compared to KCKO cells. Thus, inhibiting PGE2 with a specific COX-2 inhibitor reverses the immunosuppressive and immature phenotype of KCM-derived MDSCs. This is the first report that clearly suggests a functional role of pancreatic tumor-associated MUC1 in the development of functional MDSCs.


Cancer Research | 2011

Abstract 1070: Pancreatic Ductal Adenocarcinoma (PDA) mice lacking Mucin 1 has a profound defect in tumor growth and metastasis

Dahlia M. Besmer; Lopamudra Das Roy; Jennifer M. Curry; Teresa L. Tinder; Mahnaz Manouchehrabadi; Jorge Schettini; Sun-Il Hwang; Yong-Yook Lee; Sandra J. Gendler

MUC1 is over expressed and aberrantly glycosolated in >60% of pancreatic ductal adenocarcinomas. Over expression of MUC1 in pancreatic cancer is known, however the functional role has yet to be fully elucidated due to a dearth of appropriate models. In the present study, we have generated mouse models that spontaneously develop pancreatic ductal adenocarcinoma (KC), which are either Muc1 null (KCKO) or express human MUC1 as a self antigen (KCM). We show that PDA mice lacking Muc1 have significantly slower tumor progression, with tumor weight reaching up to a third of the KCM tumor and half of KC tumor. One out of 10 KCKO mice developed secondary metastasis versus 6 out of 10 and 3 out of 10 KCM and KC respectively. Cell lines derived from KCKO tumors have significantly lower tumorigenic capacity compared to cells derived from KCM tumors. Therefore, mice with KCKO tumors had a significant survival benefit compared to mice bearing the KCM tumors. In vitro, the KCKO cells have reduced proliferation and invasion compared to KCM cells and failed to respond to exogenous epidermal growth factor (EGF), platelet-derived growth factor (PDGF), or matrix metalloproteinase-9 (MMP9) stimuli. Further, significantly fewer KCKO cells entered the G 2 M phase of the cell cycle compared to the KCM cells, consequently more KCKO cells were observed at the S-phase of the cell cycle. Proteomics and western blotting analysis revealed a complete loss of cdc-25c expression, phosphorylation of MAPK, as well as a significant decrease in Nestin and Tubulin α-2 chain expression in KCKO cells. Treatment of the KCM cells with a MEK1/2 inhibitor, U0126, completely abrogated the enhanced proliferation of the KCM cells but had minimal effect on KCKO cells, strongly suggesting that MUC1 is necessary for MAPK activity and downstream oncogenic signaling. This is the first study to utilize a Muc1-null PDA mouse in order to fully elucidate the oncogenic role of MUC1, both in vivo and in vitro. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1070. doi:10.1158/1538-7445.AM2011-1070


Cancer Research | 2011

Abstract 3644: Pancreatic tumor cells that develop within Muc1 knock-out mice generate less immunosuppressive MDSCs in vitro

Amritha Kidiyoor; Jorge Schettini; Lopamudra Das Roy; Dahlia M. Besmer; Teresa L. Tinder

Epithelial Mucin 1 (MUC1) is a well characterized tumor associated antigen found to be over expressed and aberrantly glycosylated in human pancreatic adenocarcinomas. In recent years, Myeloid- derived suppressor cells (MDSCs), major regulators of the immune response, have been implicated in the progression of cancer. We have recently shown that Muc1 null mice have a higher propensity of MDSCs and are unable to reject allogeneic tumors as effectively as their wild-type counterparts. Here, we studied the effects of tumor derived soluble factors (TDSFs) from pancreatic cancer cell lines that over expressed human MUC1 (KCM) or were null for Muc1 (KCKO) on the generation of MDSCs from bone marrow (BM) cells in vitro. The two cell lines were derived from a spontaneous mouse model of pancreatic ductal adenocarcinoma (PDA) that was bred to the Muc1-null (KCKO) or the human MUC1 transgenic (KCM) mice. BM cells were cultured in complete DMEM supplemented with 10ng/ml GM-CSF and 10ng/ml IL-4 + 30% v/v conditioned media (TCCM) from KCKO or KCM cell lines for 5 days. On day 3, floating cells were removed and fresh media with cytokines and TCCM was added. On day 5, cells were stained for MDSCs and analyzed by flow cytometry. Statistical analysis was performed using GraphPad software and p values were calculated using One-way ANOVA. We observed a significant increase in the monocytic (CD11b+Ly6C+Ly6G-) and the granulocytic (CD11b+Ly6C+LyG+) MDSC subpopulations when naive BM cells were cultured in the presence of TCCM from KCKO versus KCM cells (p Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3644. doi:10.1158/1538-7445.AM2011-3644


Cancer Research | 2010

Abstract 4765: Treatment with anti - IL 17 A coupled with COX-2 inhibitor significantly decreases breast cancer associated secondary metastasis in a model of autoimmune arthritis

Lopamudra Das Roy; Jorge Schettini; Helen E. Gruber; Mahnaz Sahraei

Introduction: Inflammation is a critical hallmark of autoimmune arthritis (AA) and cancer. We have previously shown that the site of chronic inflammation linked to AA creates a milieu that attracts tumor cells to home and grow in the inflamed bones and lungs which are frequent sites of breast cancer metastasis. We have identified IL-17, a critical pro-inflammatory cytokine involved in osteoclastic bone resorption in AA as the underlying mechanism for increased metastasis. In addition, high levels of cyclooxygenase 2 (COX-2) is linked to both AA and breast cancer metastasis. We report that blocking the IL-17 and COX-2 pathways simultaneously significantly reduces the development of breast cancer associated metastasis in a spontaneous model of AA. Methods: 4T1 mammary gland tumors were generated in mice genetically prone to develop AA (designated SKG mice). When tumors reached > 0.2 gms, anti-IL17 antibody treatment was injected intraperitoneally once a week for three weeks. Celecoxib, a specific COX-2 inhibitor was orally administered starting at the same time as the anti-IL-17 antibody but was given daily until sacrifice. One week after the last injection, mice were sacrificed. Secondary metastasis and the associated inflammatory milieu were evaluated. Summary of Results: Data demonstrated that compared to the untreated mice or mice treated with isotype control antibody, survival was significantly enhanced and primary tumor burden was significantly decreased in the anti-IL-17+celecoxib treated mice. Importantly, we observed significant decrease in lung and bone metastasis as compared to the control group. The contributing factor was recognized to be the low neutrophilic and granulocytic infiltration and therefore low inflammation in the lungs as well as reduced severity of arthritis with low cellular infiltration and low osteoclast formation in the bones of mice treated with anti-IL-17+celecoxib. Conclusion: We report that treatment with anti-IL 17 and celecoxib significantly reduced primary tumor burden, reduced metastasis to the lungs and bones, and increased survival in SKG mice. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4765.

Collaboration


Dive into the Jorge Schettini's collaboration.

Top Co-Authors

Avatar

Lopamudra Das Roy

University of North Carolina at Charlotte

View shared research outputs
Top Co-Authors

Avatar

Dahlia M. Besmer

University of North Carolina at Charlotte

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amritha Kidiyoor

University of North Carolina at Charlotte

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennifer M. Curry

University of North Carolina at Charlotte

View shared research outputs
Top Co-Authors

Avatar

Mahnaz Sahraei

University of North Carolina at Charlotte

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sun-Il Hwang

Carolinas Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge