Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Occup Med (Lond) ; 73(2): 80-84, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36719096

ABSTRACT

BACKGROUND: Attrition from combat service carries significant organizational and personal ramifications, but predicting factors associated with attrition remains challenging. AIMS: To evaluate medical and psychosocial factors associated with attrition from basic combat training (BCT) in the Israel Defense Forces (IDF). In addition, we identify subsets of the recruit population which exhibit certain trends in terms of medical corresponding with a high risk of attrition. METHODS: A cross-sectional study of IDF combat trainees undergoing infantry BCT between 2012 and 2017. Data were collected from the soldiers' electronic medical and administrative records. We used multivariable logistic regression and the SAS® decision-tree tool to analyse key predictive factors for attrition. RESULTS: A total of 46 472 soldiers enlisted to BCT during the research period. The mean body mass index (BMI) was 21.8 (SD 3.54). The overall attrition rate was 10%. The following factors were associated with attrition from BCT: ethnicity (P < 0.01), BMI (P < 0.01), pre-enlisting motivation score (P < 0.01) and the number of mental health officer visits (P < 0.01). Using a decision-tree model, we found a high attrition rate among soldiers who had >5.2 to primary care physician visits (11% attrition rate versus 3%) or more than 11 sick leave days (59% versus 19%). CONCLUSIONS: This study sheds light on unique measures relating to attrition. Attrition is associated with several demographic and psychosocial factors. Early prediction of motivation and monitoring of healthcare utilization may enable early identification and focused interventions targeting soldiers at high risk for attrition. These findings need to be further translated into actionable directives and further investigations.


Subject(s)
Military Personnel , Humans , Military Personnel/psychology , Israel/epidemiology , Cross-Sectional Studies , Mental Health
2.
Oncogene ; 31(47): 4935-45, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-22266848

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) has one of the worst prognoses of all cancers. Mucin 1 (MUC1), a transmembrane mucin glycoprotein, is a key modulator of several signaling pathways that affect oncogenesis, motility and metastasis. Its expression is known to be associated with poor prognosis in patients. However, the precise mechanism remains elusive. We report a novel association of MUC1 with platelet-derived growth factor-A (PDGFA). PDGFA is one of the many drivers of tumor growth, angiogenesis and metastasis in PDA. Using mouse PDA models as well as human samples, we show clear evidence that MUC1 regulates the expression and secretion of PDGFA. This, in turn, influences proliferation and invasion of pancreatic cancer cells leading to higher tumor burden in vivo. In addition, we reveal that MUC1 overexpressing cells are heavily dependent on PDGFA both for proliferation and invasion, whereas MUC1-null cells are not. Moreover, PDGFA and MUC1 are critical for translocation of ß catenin to the nucleus for oncogenesis to ensue. Finally, we elucidate the underlying mechanism by which MUC1 regulates PDGFA expression and secretion in pancreatic cancer cells. We show that MUC1 associates with Hif1-α, a known transcription factor involved in controlling PDGFA expression. Furthermore, MUC1 facilitates Hif1-α translocation to the nucleus. In summary, we have demonstrated that MUC1-induced invasion and proliferation occurs via increased exogenous production of PDGFA. Thus, impeding MUC1 regulation of PDGFA signaling may be therapeutically beneficial for patients with PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Gene Expression Regulation, Neoplastic , Mucin-1/physiology , Pancreatic Neoplasms/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Progression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Platelet-Derived Growth Factor/genetics , Protein Binding , Protein Transport , Tumor Burden , beta Catenin/metabolism
3.
Am J Physiol Gastrointest Liver Physiol ; 300(2): G327-33, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21109593

ABSTRACT

The colonic mucus layer serves as an important barrier and prevents colonic bacteria from invading the mucosa and cause inflammation. The regulation of colonic mucus secretion is poorly understood. The aim of this study was to investigate the role of the mucus barrier in induction of colitis. Furthermore, regulation of mucus secretion by luminal bacterial products was studied. The colon of anesthetized Muc2(-/-), Muc1(-/-), wild-type (wt), and germ-free mice was exteriorized, the mucosal surface was visualized, and mucus thickness was measured with micropipettes. Colitis was induced by DSS (dextran sodium sulfate, 3%, in drinking water), and disease activity index (DAI) was assessed daily. The colonic mucosa of germ-free and conventionally housed mice was exposed to the bacterial products LPS (lipopolysaccharide) and PGN (peptidoglycan). After DSS induction of colitis, the thickness of the firmly adherent mucus layer was significantly thinner after 5 days and onward, which paralleled the increment of DAI. Muc2(-/-) mice, which lacked firmly adherent mucus, were predisposed to colitis, whereas Muc1(-/-) mice were protected with significantly lower DAI by DSS compared with wt mice. The mucus barrier increased in Muc1(-/-) mice in response to DSS, whereas significantly fewer T cells were recruited to the inflamed colon. Mice housed under germ-free conditions had an extremely thin adherent colonic mucus layer, but when exposed to bacterial products (PGN or LPS) the thickness of the adherent mucus layer was quickly restored to levels observed in conventionally housed mice. This study demonstrates a correlation between decreasing mucus barrier and increasing clinical symptoms during onset of colitis. Mice lacking colonic mucus (Muc2(-/-)) were hypersensitive to DSS-induced colitis, whereas Muc1(-/-) were protected, probably through the ability to increase the mucus barrier but also by decreased T cell recruitment to the afflicted site. Furthermore, the ability of bacteria to regulate the thickness of the colonic mucus was demonstrated.


Subject(s)
Colitis/physiopathology , Colon/metabolism , Intestinal Mucosa/metabolism , Mucin-1/metabolism , Mucin-2/metabolism , Animals , Bacterial Translocation , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Colon/drug effects , Dextran Sulfate , Disease Susceptibility , Down-Regulation , Germ-Free Life , Intestinal Mucosa/drug effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-1/genetics , Mucin-2/genetics , Peptidoglycan/pharmacology , Severity of Illness Index , T-Lymphocytes/pathology
4.
Oncogene ; 30(12): 1449-59, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21102519

ABSTRACT

Increased motility and invasiveness of pancreatic cancer cells are associated with epithelial to mesenchymal transition (EMT). Snai1 and Slug are zinc-finger transcription factors that trigger this process by repressing E-cadherin and enhancing vimentin and N-cadherin protein expression. However, the mechanisms that regulate this activation in pancreatic tumors remain elusive. MUC1, a transmembrane mucin glycoprotein, is associated with the most invasive forms of pancreatic ductal adenocarcinomas (PDA). In this study, we show that over expression of MUC1 in pancreatic cancer cells triggers the molecular process of EMT, which translates to increased invasiveness and metastasis. EMT was significantly reduced when MUC1 was genetically deleted in a mouse model of PDA or when all seven tyrosines in the cytoplasmic tail of MUC1 were mutated to phenylalanine (mutated MUC1 CT). Using proteomics, RT-PCR and western blotting, we revealed a significant increase in vimentin, Slug and Snail expression with repression of E-Cadherin in MUC1-expressing cells compared with cells expressing the mutated MUC1 CT. In the cells that carried the mutated MUC1 CT, MUC1 failed to co-immunoprecipitate with ß-catenin and translocate to the nucleus, thereby blocking transcription of the genes associated with EMT and metastasis. Thus, functional tyrosines are critical in stimulating the interactions between MUC1 and ß-catenin and their nuclear translocation to initiate the process of EMT. This study signifies the oncogenic role of MUC1 CT and is the first to identify a direct role of the MUC1 in initiating EMT during pancreatic cancer. The data may have implications in future design of MUC1-targeted therapies for pancreatic cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Mucin-1/metabolism , Pancreatic Neoplasms/pathology , Amino Acid Sequence , Animals , Cadherins/metabolism , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mucin-1/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Tyrosine/genetics , Tyrosine/metabolism
5.
Oncogene ; 28(48): 4225-36, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-19734944

ABSTRACT

Mice transgenic for MUC1 (mucin 1) and polyomavirus middle T (PyMT) develop mammary carcinomas within 15 weeks with 100% penetrance. PyMT-induced mammary tumorigenesis is closely correlated with robust telomerase expression and activity. To assess the role of telomerase activation and telomere maintenance in mammary carcinoma tumorigenesis, we generated mice expressing MUC1 and PyMT (MMT mice) but deficient in the telomerase RNA component, mTerc, on the C57BL/6 background. Successive generational intercrosses of mTerc(-/-)MMT mice produced cohorts with progressively shorter telomeres that were audited for mammary tumor formation. Relative to MMT (N=14) and G0 mTerc(+/-) female controls (G0=14), mTerc(-/-)MMT females (G1=11, G2=15, G3=15 and G4=5) showed decreased tumor volumes and increased tumor latency-MMT=95.6 days; G0 mTerc(+/-)MMT=98.6 days versus G1, G2, G3 and G4 mTerc(-/-)MMT mice with latencies of 122.6, 138.9, 140.7 and 220.9 days, respectively (controls versus G1-G4, P<0.005). The progressive impairment of lung metastasis was also observed with each successive mTerc(-/-)MMT generation. The impairment of tumorigenesis was associated with decreased proliferation of mammary epithelial and tumor cells and increased apoptosis of tumor cells. Together, these results indicate that, in the setting of viral oncoprotein mammary tumorigenesis, telomerase-dependent telomere maintenance facilitates the formation and metastatic progression of mammary tumors.


Subject(s)
Mammary Neoplasms, Experimental/genetics , Oncogenes/genetics , Telomerase/deficiency , Telomere/pathology , Animals , Cell Cycle/genetics , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Female , Humans , Mammary Neoplasms, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic/metabolism , Mucin-1/genetics , Oncogenes/physiology , RNA
6.
Acta Gastroenterol Belg ; 71(3): 342-4, 2008.
Article in English | MEDLINE | ID: mdl-19198584

ABSTRACT

Intramural hemorrhage as a cause for small bowel obstruction is extremely rare. We presented an unusual case report of small bowel obstruction caused by intramural jejunal hemorrhage secondary to anticoagulant therapy. An 85-year-old male patient with atrial fibrillation on long-term warfarin presented with nausea and vomiting for 2 days, accompanied with no bowel movement since the onset. Physical exam was unremarkable except soft abdomen with distension but no tenderness, hyperactive bowel sounds and positive fecal occult blood test. Investigations showed anemia with hemoglobin/ hematocrit of 10 (g/dl) / 30%, prothrombin time with an International Normalized Ratio (INR) of 9.58. Abdominal x-ray showed air fluid levels suggestive of small bowel obstruction. Contrast-enhanced abdominal computerized tomography showed circumferential wall thickening, luminal narrowing and partial small bowel obstruction secondary to intramural jejunal hemorrhage. Patient recovered completely 48 hours after medical treatment (nothing per oral, intravenous fluids, nasal gastric tube, Vitamin K, frozen fresh plasma and packed red blood cell transfusion). Spontaneous intramural small-bowel hematoma is rare and occurs in patients who receive excessive anticoagulation with warfarin or who have some other risk factors for bleeding. Intramural hematoma most commonly involves the jejunum, followed by the ileum and the duodenum. The spectrum of presentation is wide, from abdominal pain, emesis to gastrointestinal tract hemorrhage. Abdominal CT is the key for diagnosis, with characteristics including circumferential wall thickening, intramural hyperdensity, luminal narrowing, and intestinal obstruction. Early diagnosis is crucial because most patients are treated nonoperatively with a good outcome.


Subject(s)
Anticoagulants/adverse effects , Gastrointestinal Hemorrhage/chemically induced , Intestinal Obstruction/etiology , Intestine, Small , Jejunal Diseases/chemically induced , Warfarin/adverse effects , Aged, 80 and over , Atrial Fibrillation/drug therapy , Humans , Male
7.
Vaccine ; 25(9): 1607-18, 2007 Feb 19.
Article in English | MEDLINE | ID: mdl-17166639

ABSTRACT

A MUC1-based vaccine was used in a preclinical model of colon cancer. The trial was conducted in a MUC1-tolerant immune competent host injected with MC38 colon cancer cells expressing MUC1. The vaccine included: MHC class I-restricted MUC1 peptides, MHC class II-restricted pan-helper-peptide, unmethylated CpG oligodeoxynucleotide, and granulocyte macrophage-colony stimulating factor. Immunization was successful in breaking MUC1 self-tolerance, and in eliciting a robust anti-tumor response. The vaccine stimulated IFN-gamma-producing CD4(+) helper and CD8(+) cytotoxic T cells against MUC1 and other undefined MC38 tumor antigens. In the prophylactic setting, immunization caused complete rejection of tumor cells, while in the therapeutic regimen, tumor burden was significantly reduced.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Disease Models, Animal , Immunotherapy/methods , Mucin-1/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Cell Line, Tumor , Colonic Neoplasms/physiopathology , Colonic Neoplasms/prevention & control , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunization , Interferon-gamma/biosynthesis , Lymphocyte Activation , Mice , Mucin-1/administration & dosage , Mucin-1/metabolism , Mucins , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology
8.
Histol Histopathol ; 21(8): 849-55, 2006 08.
Article in English | MEDLINE | ID: mdl-16691537

ABSTRACT

UNLABELLED: The immunohistochemical detection (IHC) of MUC1-CT employing a polyclonal antibody (CT33) in relation to CT2 monoclonal antibody (MAb) was analyzed. Western blot (WB) was used to determine the molecular mass of CT. MATERIALS AND METHODS: We studied 163 breast and 89 colorectal cancer specimens, 10 breast and 14 colorectal benign conditions, and 12 breast and 20 colorectal normal samples. From each tumor sample, subcellular fractions were obtained and analyzed by SDS-PAGE and WB. A nonparametric statistical analysis was employed; data were standardized and a Kendall-Tau correlation was applied. RESULTS: By IHC, 146/163 (90%) and 151/163 (93%) of breast cancer were positive with CT33 and CT2, respectively; a statistically significant correlation was obtained (t=0.5199). Seven out of ten (70%) benign breast specimens were positive with CT33 while all samples stained with CT2; in normal breast sample tissues, all were positive with both Abs. In colorectal cancer samples, both antibodies stained 47/89 (53%) samples; CT2 reacted in 13/14 (93%) of benign samples while CT33 showed a positive reaction in 9/14 (64%) of benign specimens. In normal samples, CT2 showed staining in 17/20 (85%) of samples and CT33 was reactive in 12/20 (60%). By WB, in breast and colorectal cancer samples, similar results were obtained with both antibodies: a main band at about 30kDa which represents the smaller subunit. CONCLUSION: CT33 polyclonal antibody has demonstrated its efficacy to detect MUC1 in breast and colorectal cancer tissues with similar reactivity to CT2. It is worthwhile to affirm that CT33 is a good indicator of MUC1 expression.


Subject(s)
Antibodies, Monoclonal/metabolism , Breast Neoplasms/metabolism , Colorectal Neoplasms/metabolism , Mucin-1/metabolism , Organic Cation Transport Proteins/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/metabolism , Biomarkers, Tumor , Breast/anatomy & histology , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cell Fractionation , Colon/anatomy & histology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/pathology , Humans , Immunoenzyme Techniques , Mucin-1/immunology , Organic Cation Transport Proteins/immunology , Rectum/anatomy & histology , Rectum/metabolism , Rectum/pathology
9.
Gene Ther ; 13(1): 60-6, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16121204

ABSTRACT

Ovarian cancer represents the fifth leading cause of cancer death among women in the United States, with >16 000 deaths expected this year. This study was carried out to investigate the potential of sodium iodide symporter (NIS)-mediated radioiodide therapy as a novel approach for ovarian cancer treatment. Radioiodide is routinely and effectively used for the treatment of benign and malignant thyroid disease as a result of native thyroidal expression of NIS, which mediates iodide uptake. In vitro gene transfer studies in ovarian cancer cells revealed a 12- and five-fold increase in iodide uptake when transduced with Ad/CMV/NIS or Ad/MUC1/NIS, respectively. Western blot/immunohistochemistry confirmed NIS protein expression. In vivo ovarian tumor xenografts were infected with the adenoviral constructs. (123)I imaging revealed a clear image of the CMV/NIS-transduced tumor, with a less intense image apparent following infection with MUC1/NIS. Therapeutic doses of (131)I following CMV/NIS infection caused a mean 53% reduction in tumor volume (P<0.0001). MUC1/NIS-transduced tumors did not regress, although at 8 weeks following therapy, tumor volume was significantly less that of control animals (166 versus 332%, respectively, P<0.05). This study represents a promising first step investigating the potential for NIS-mediated radioiodide imaging and therapy of ovarian tumors.


Subject(s)
Genetic Therapy/methods , Iodine Radioisotopes , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/therapy , Symporters/genetics , Adenoviridae/genetics , Animals , Blotting, Western/methods , Cell Line, Tumor , Female , Genetic Vectors/administration & dosage , Humans , Immunohistochemistry/methods , Iodine Radioisotopes/therapeutic use , Mice , Mucin-1/genetics , Neoplasm Transplantation , Promoter Regions, Genetic , Radionuclide Imaging , Symporters/metabolism , Transduction, Genetic/methods , Transplantation, Heterologous
10.
J Leukoc Biol ; 77(1): 90-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15513966

ABSTRACT

MUC1 (CD227) is a large transmembrane epithelial mucin glycoprotein, which is aberrantly overexpressed in most adenocarcinomas and is a target for immune therapy for epithelial tumors. Recently, MUC1 has been detected in a variety of hematopoietic cell malignancies including T and B cell lymphomas and myelomas; however, its function in these cells is not clearly defined. Using the Jurkat T cell lymphoma cell line and normal human T cells, we demonstrate that MUC1 is not only expressed in these cells but is also phosphorylated upon T cell receptor (TCR) ligation and associates with the Src-related T cell tyrosine kinase, p56lck. Upon TCR-mediated activation of Jurkat cells, MUC1 is found in the low-density membrane fractions, where linker of T cell activation is contained. Abrogation of MUC1 expression in Jurkat cells by MUC1-specific small interfering RNA resulted in defects in TCR-mediated downstream signaling events associated with T cell activation. These include reduction in Ca2+ influx and extracellular signal-regulated kinase 1/2 phosphorylation, leading to a decrease in CD69 expression, proliferation, and interleukin-2 production. These results suggest a regulatory role of MUC1 in modulating proximal signal transduction events through its interaction with proteins of the activation complex.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Lymphoma/metabolism , Mucin-1/metabolism , T-Lymphocytes/metabolism , Adult , Amino Acid Sequence , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Proliferation , Humans , Interleukin-2/metabolism , Jurkat Cells , Lectins, C-Type , Lymphocyte Activation , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Mucin-1/genetics , Phosphorylation , RNA, Small Interfering/pharmacology , Receptors, Antigen, T-Cell/metabolism , Sequence Homology, Amino Acid
11.
Br J Cancer ; 90(3): 657-64, 2004 Feb 09.
Article in English | MEDLINE | ID: mdl-14760381

ABSTRACT

MUC4 is highly expressed in human pancreatic tumours and pancreatic tumour cell lines, but is minimally or not expressed in normal pancreas or chronic pancreatitis. Here, we investigated the aberrant regulation of MUC4 expression in vivo using clonal human pancreatic tumour cells (CD18/HPAF) grown either orthotopically in the pancreas (OT) or ectopically in subcutaneous tissue (SC) in the nude mice. Histological examination of the OT and SC tumours showed moderately differentiated and anaplastic morphology, respectively. The OT tumour cells showed metastases to distant lymph nodes and faster tumour growth (P<0.01) compared to the SC tumours. The MUC4 transcripts in OT tumours were very high compared to the undetectable levels in SC tumours. The SC tumour cells regained their ability to express MUC4 transcripts after in vitro culture. Immunohistochemical analysis using MUC4-specific polyclonal antiserum confirmed the results obtained by Northern blot analysis. Interestingly, the OT tumours showed expression of TGFbeta2 compared to no expression in SC, suggesting a possible link between MUC4 and TGFbeta2. The MUC4 expression, morphology, and metastasis of human pancreatic tumour cells are regulated by a local host microenvironment. TGFbeta2 may serve as an interim regulator of this function.


Subject(s)
Biomarkers, Tumor/analysis , Gene Expression Regulation, Neoplastic , Immunosuppressive Agents/pharmacology , Mucins/biosynthesis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology , Transforming Growth Factor beta/pharmacology , Animals , Blotting, Northern , Choristoma , Humans , Immunohistochemistry , Mice , Mice, Nude , Mucin-4 , Transforming Growth Factor beta2 , Transplantation, Heterologous , Tumor Cells, Cultured
12.
J Leukoc Biol ; 72(4): 692-701, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12377938

ABSTRACT

The MUC1 mucin (CD227) is a cell surface mucin originally thought to be restricted to epithelial tissues. We report that CD227 is expressed on human blood dendritic cells (DC) and monocyte-derived DC following in vitro activation. Freshly isolated murine splenic DC had very low levels of CD227; however, all DC expressed CD227 following in vitro culture. In the mouse spleen, CD227 was seen on clusters within the red pulp and surrounding the marginal zone in the white pulp. Additionally, we confirm CD227 expression by activated human T cells and show for the first time that the CD227 cytoplasmic domain is tyrosine-phosphorylated in activated T cells and DC and is associated with other phosphoproteins, indicating a role in signaling. The function of CD227 on DC and T cells requires further elucidation.


Subject(s)
Dendritic Cells/immunology , Mucin-1/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CD11c Antigen/immunology , Cells, Cultured , Cytoplasm , Dendritic Cells/cytology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Phosphorylation , Spleen/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
13.
Clin Immunol ; 101(2): 192-200, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11683578

ABSTRACT

Fusions of autologous tumor cells with allogeneic dendritic cells (DC) represent an approach for the induction of antitumor immunity. In the present studies, we investigated the antitumor effects of vaccinating MUC1-transgenic (MUC1.Tg) mice with MC38/MUC1 carcinoma cells fused to allogeneic DC from BALB/c mice (allo-DC, H-2(d)) or syngeneic DC from C57BL/6 mice (syn-DC, H-2(b)). Both allo and syn fusion cells (FC/MUC1) expressed MHC class II, costimulatory molecules, and the MUC1 antigen. Allo-FC/MUC1 exhibited dual expression of MHC class I haplotypes (H-2(d)/H-2(b))and MUC1 antigen. By contrast, only H-2(b) and MUC1 antigen were expressed by syn-FC/MUC1. CTLs from MUC1.Tg mice immunized with allo- or syn-FC/MUC1 fusion cells lysed MC38/MUC1 targets. Moreover, immunization with allo- or syn-FC/MUC1 was effective in eliminating established MUC1-positive pulmonary metastases in MUC1.Tg mice. These results indicate that immunization of MUC1.Tg mice with syn- or allo-FC/MUC1 is effective in reversing immunologic unresponsiveness to MUC1 antigen and inducing immunity against MUC1-positive tumors. The findings in the present study have broader clinical implications for fusion cell vaccines.


Subject(s)
Cancer Vaccines/immunology , Cell Fusion , Dendritic Cells/immunology , Mucin-1/immunology , Animals , Female , Histocompatibility Antigens Class I/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mucin-1/analysis , Mucin-1/genetics , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Vaccination
14.
Trends Mol Med ; 7(10): 471-5, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11597523

ABSTRACT

Recent discoveries regarding the identification of tumor-associated antigens and antigen presentation have made successful immunotherapy strategies possible with little, if any, toxicity. Here, we describe transgenic mammary, pancreas, prostate, stomach and lung adenocarcinoma animal models that can be used to study various immunotherapeutic strategies. The challenge in developing a tumor vaccine is effective antigen presentation that elicits anti-tumor immune responses without precipitating autoimmunity. Clinical trials must be preceded by appropriate animal studies to demonstrate that the concepts can be translated into efficacious therapy for cancer. Although many xenograph or transplantable tumor models have been used, the most effective studies are in spontaneous tumor models. These models are clinically relevant, as tumors arise in an appropriate tissue background and in a host conditioned by the physiological events of neoplastic progression and tumorigenesis and in the context of a viable immune system.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/therapy , Disease Models, Animal , Immunotherapy/methods , Adenocarcinoma/pathology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Female , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/therapy , Mice , Mice, Transgenic , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy
15.
J Mammary Gland Biol Neoplasia ; 6(3): 339-53, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11547902

ABSTRACT

MUC1 is a large, heavily glycosylated mucin expressed on the apical surfaces of most simple, secretory epithelia including the mammary gland, gastrointestinal, respiratory, urinary and reproductive tracts. Although MUC1 was thought to be an epithelial-specific protein, it is now known to be expressed on a variety of hematopoietic cells as well. Mucins function in protection and lubrication of epithelial surfaces. Transmembrane mucins, which contain cytoplasmic tail domains, appear to have additional functions through their abilities to interact with many proteins involved in signal transduction and cell adhesion. The goal of this review is to highlight recent discoveries that suggest that MUC1 may be a multifunctional protein, located on the surfaces of cells as a sensor of the environment, poised to signal to the interior when things go awry.


Subject(s)
Breast Neoplasms/metabolism , Mucin-1/physiology , Animals , Female , Humans , Mammary Neoplasms, Experimental/metabolism
16.
Clin Cancer Res ; 7(3 Suppl): 848s-855s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300482

ABSTRACT

We have reported previously that MUC1 transgenic mice with spontaneous tumors of the pancreas (designated MET) naturally develop MHC class I-restricted, MUC1-specific CTLs as tumors progress (P. Mukherjee et al., J. Immunol., 165: 3451-3460, 2000). From these MET mice, we have isolated, expanded, and cloned naturally occurring MUC1-specific CTLs in vitro. In this report, we show that the CTL line is predominantly CD8+ T cells and expresses T-cell receptor Vbeta chains 5.1/5.2, 11, 13, and 2 and Valpha chains 2, 8.3, 3.2, and 11.1/11.2. These CTLs recognize several epitopes on the MUC1 tandem repeat with highest affinity to APGSTAPPA. The CTL clone, on the other hand, is 100% CD8+ cells and expresses a single Vbeta chain of 5.1/5.2 and Valpha2. It recognizes only the H-2Db class I-restricted epitope of MUC1, APGSTAPPA. When adoptively transferred, the CTLs were effective in eradicating MUC1-expressing injected tumor cells including mammary gland cells (C57mg) and B16 melanomas. These results suggest that MUC1-specific CTLs are capable of possibly preventing, or at least substantially delaying, MUC1-expressing tumor formation. To our knowledge, this is the first evidence that demonstrates that the naturally occurring MUC1-specific CTLs isolated from one tumor model has antitumor effects on other MUC1-expressing tumors in vivo. Therefore, our data confirm that MUC1 is an important tumor rejection antigen and can serve as a target for immunotherapy.


Subject(s)
Mucin-1/chemistry , T-Lymphocytes, Cytotoxic/metabolism , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cell Separation , Epitopes , Flow Cytometry , Humans , Interferon-gamma/metabolism , Melanoma, Experimental , Mice , Mice, Transgenic , Peptides/chemistry , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/metabolism , Time Factors
17.
J Biol Chem ; 276(16): 13057-64, 2001 Apr 20.
Article in English | MEDLINE | ID: mdl-11278868

ABSTRACT

MUC1 is a large (>400 kDa), heavily glycosylated transmembrane protein that is aberrantly expressed on greater than 90% of human breast carcinomas and subsequent metastases. The precise function of MUC1 overexpression in tumorigenesis is unknown, although various domains of MUC1 have been implicated in cell adhesion, cell signaling, and immunoregulation. Stimulation of the MDA-MB-468 breast cancer line as well as mouse mammary glands with epidermal growth factor results in the co-immunoprecipitation of MUC1 with a tyrosine-phosphorylated protein of approximately 180 kDa. We have generated transgenic lines overexpressing full-length (MMF), cytoplasmic tail deleted (DeltaCT), or tandem repeat deleted (DeltaTR)-human MUC1 under the control of the mouse mammary tumor virus promoter to further examine the role of MUC1 in signaling and tumorigenesis. Immunoprecipitation experiments revealed that full-length transgenic MUC1 physically associates with all four erbB receptors, and co-localizes with erbB1 in the lactating gland. Furthermore, we detected a sharp increase in ERK1/2 activation in MUC1 transgenic mammary glands compared with Muc1 null and wild-type animals. These results point to a novel function of increased MUC1 expression, potentiation of erbB signaling through the activation of mitogenic MAP kinase pathways.


Subject(s)
ErbB Receptors/metabolism , Mammary Glands, Animal/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mucin-1/metabolism , Animals , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Mammary Glands, Animal/cytology , Mice , Mice, Knockout , Mice, Transgenic , Mucin-1/genetics , Phosphoproteins/metabolism , Phosphotyrosine , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Receptor, ErbB-4
18.
Int Immunol ; 13(2): 233-40, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11157856

ABSTRACT

We investigated the influence of organ-specific parameters on tolerance and immunity to human MUC1. C57Bl/6 mice (wild-type) and C57Bl/6 transgenic for MUC1 (MUC1.Tg) were challenged in the pancreas with Panc02-MUC1, a C57Bl/6-syngeneic pancreatic cancer cell line expressing human MUC1. Wild-type mice produced immune responses to MUC1 when presented on tumor cells growing in the pancreas; however, the responses to tumors in the pancreas were less effective than responses produced by tumor challenge at the s.c. site. Tumor immunity specific for MUC1 was produced in wild-type mice by two different procedures: (i) s.c. immunization of wild-type mice with a low dose of Panc02-MUC1 or (ii) adoptive transfer of spleen and lymph node cells harvested from wild-type mice previously immunized s.c. with Panc02-MUC1. This demonstrates that immune responses to MUC1 presented at the s.c. site can be detected and adoptively transferred. MUC1.Tg mice were immunologically tolerant to MUC1; however, some immunological protection against orthotopic challenge with Panc02-MUC1 was conferred by adoptive transfer of CD4+ and CD8+ T cells from wild-type mice. These results show that it is more difficult to produce immune responses to tumors growing at the pancreatic site than the s.c. site. Panc02-MUC1 cells growing in the pancreas were accessible to the immune system, and immune responses evoked by s.c. presentation of this molecule in wild-type mice were effective in rejecting tumor cells in the pancreas of both wild-type and MUC1.Tg mice. No effective anti-tumor immune responses against MUC1 were produced in MUC1.Tg mice.


Subject(s)
Graft Rejection/immunology , Mucin-1/immunology , Pancreas/immunology , Pancreatic Neoplasms/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Graft Rejection/pathology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation/immunology , Organ Specificity/genetics , Organ Specificity/immunology , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Survival Analysis , Tumor Cells, Cultured/transplantation
20.
Glycoconj J ; 18(11-12): 931-42, 2001.
Article in English | MEDLINE | ID: mdl-12820727

ABSTRACT

Pancreatic cancer is a highly aggressive, treatment refractory disease and is the fourth leading cause of death in the United States. In humans, 90% of pancreatic adenocarcinomas over-express altered forms of a tumor-associated antigen, MUC1 (an epithelial mucin glycoprotein), which is a target for immunotherapy. Using a clinically relevant mouse model of pancreas cancer that demonstrates peripheral and central tolerance to human MUC1 and develops spontaneous tumors of the pancreas, we have previously reported the presence of functionally active, low affinity, MUC1-specific precursor cytotoxic T cells (pCTLs). Hypothesis for this study is that MUC1-based immunization may enhance the low level MUC1-specific immunity that may lead to an effective anti-tumor response. Data demonstrate that MUC1 peptide-based immunization elicits mature MUC1-specific CTLs in the peripheral lymphoid organs. The mature CTLs secrete IFN-gamma and are cytolytic against MUC1-expressing tumor cells in vitro. However, active CTLs that infiltrate the pancreas tumor microenvironment become cytolytically anergic and are tolerized to MUC1 antigen, allowing the tumor to grow. We demonstrate that the CTL tolerance could be reversed at least in vitro with the use of anti-CD40 co-stimulation. The pancreas tumor cells secrete immunosuppressive cytokines, including IL-10 and TGF-beta that are partly responsible for the down-regulation of CTL activity. In addition, they down-regulate their MHC class I molecules to avoid immune recognition. CD4+ CD25+ T regulatory cells, which secrete IL-10, were also found in the tumor environment. Together these data indicate the use of several immune evasion mechanisms by tumor cells to evade CTL killing. Thus altering the tumor microenvironment to make it more conducive to CTL killing may be key in developing a successful anti-cancer immunotherapy.


Subject(s)
Mucin-1/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Disease Models, Animal , Disease Progression , Immune Tolerance , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mucin-1/biosynthesis , Mucin-1/genetics , Pancreatic Neoplasms/metabolism , Survival Rate , Transforming Growth Factor beta/metabolism , Tumor Escape/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...