Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
J Immunol ; 203(10): 2735-2745, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31578272

ABSTRACT

Therapeutic uses of mesenchymal stromal cells (MSCs) have emerged over the past decade. Yet, their effect on tumor growth remains highly debated, particularly in an immune competent environment. In this study, we wanted to investigate the impact of human umbilical cord-derived MSCs (hUC-MSCs) on tumor growth in humanized mice generated by the human adoptive transfer of PBMCs or the cotransplantation of hematopoietic stem cells and human thymic tissue (human BLT [Hu-BLT]). Our results showed that the growth and immune rejection of engineered human fibroblastic tumors was not altered by the injection of hUC-MSCs in immune-deficient or humanized mice, respectively. This was observed whether tumor cells were injected s.c. or i.v. and independently of the injection route of the hUC-MSCs. Moreover, only in Hu-BLT mice did hUC-MSCs have some effects on the tumor-immune infiltrate, yet without altering tumor growth. These results demonstrate that hUC-MSCs do not promote fibroblastic tumor growth and neither do they prevent tumor infiltration and rejection by immune cells in humanized mice.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Adoptive Transfer , Animals , Cell Line, Transformed/transplantation , Fibroblasts/transplantation , Genetic Vectors , Graft Rejection/immunology , Heterografts , Humans , Injections, Intravenous , Injections, Subcutaneous , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Radiation Chimera , Specific Pathogen-Free Organisms , Telomerase/genetics , Telomerase/physiology , Thymus Gland/transplantation , Wharton Jelly/cytology
2.
PLoS One ; 13(10): e0205666, 2018.
Article in English | MEDLINE | ID: mdl-30359388

ABSTRACT

BACKGROUND: Activation of Ras oncogene in human tumors is associated with radiation-associated metastatic potential. Although ionizing radiation is one important method of cancer treatments, it has been shown to enhance matrix metalloproteinases (MMPs) activity and facilitates a more aggressive cancer phenotype. Our previous studies showed that andrographolide with lower dose rates of radiation could inhibit RAS-transformed cancer metastasis in vivo; however, the molecular mechanisms are not yet clear. In this study, we aimed to explore the anti-metastatic effect of andrographolide combined with radiation on Ras-transformed cells. METHODS: RAS-transformed cells were treated with andrographolide in the presence or absence of irradiation (2-4 Gy) or angiotensin II to examine cell invasion. In vivo tumorigenesis assays were also performed. The MMP-2 activity was detected by using Gelatin zymography. Signal transduction of NF-κB subunit, p65 and phosphor-ERK 1/2, were examined by using Western blotting analysis. RESULTS: Treatment with andrographolide inhibited migration of Ras-transformed cells. Andrographolide treatment with radiation significantly inhibited cancer metastasis in vivo. We found that andrographolide exhibited anti-migration and anti-invasive ability against cancer metastasis via inhibition of MMP2 activity rather than affected MMP-9 and EMT. In addition, combined andrographolide with radiation appeared to be more effective in reducing MMP-2 expression, and this effect was accompanied by suppression of ERK activation that inhibits cancer cell migration and invasion. CONCLUSIONS: These findings suggest that andrographolide enhances the anti-metastatic effect of radiation in Ras-transformed cells via suppression of ERK-mediated MMP-2 activity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Diterpenes/pharmacology , Matrix Metalloproteinase 2/metabolism , Neoplasms/therapy , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Transformed/transplantation , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Transformation, Viral , Chemoradiotherapy/methods , Disease Models, Animal , Diterpenes/therapeutic use , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/prevention & control , Neoplasms/pathology , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Rats , Retroviridae/genetics , Retroviridae/metabolism
3.
Oncol Rep ; 33(4): 1675-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25633640

ABSTRACT

Bee venom (BV) therapy is a type of alternative medical treatment used to treat various diseases in oriental medicine. The mechanisms underlying the effects of BV remain poorly understood. In the present study, we evaluated the antiviral effect of BV on cervical carcinoma cell lines (CaSki, HeLa, C33A and TC-1). BV treatments resulted in a more significant suppression of cell growth in HPV 16-infected cells (CaSki) and a lesser suppression in HPV 18-infected cells (HeLa). However, less suppression was observed in HPV-negative C33A cells. In 10 µg/ml BV-treated CaSki cells, the mRNA expression and protein levels of HPV16 E6 and E7 were significantly decreased by BV, while HPV18 E6 and E7 mRNA expression levels were not significantly altered by 10 µg/ml BV-treated HeLa cells. The antitumor effects of BV were in accordance with in vitro data, in restricting tumor growth in vivo and were much more effective on the suppression of tumor growth. Furthermore, the mRNA and protein expression levels of HPV16 E6 and E7 were decreased by BV in TC-1 tumors. These findings demonstrated the antiviral effects of BV in HPV-infected cervical cancer cells and the anticancer effects of BV in HPV16 E6/E7-expressed TC-1 tumors. Collectively, BV plays a differential role in suppressing HPV16-infected cells (CaSki cells) and HPV18-infected cells (HeLa cells) by the downregulation of E6/E7 protein of HPV16/18.


Subject(s)
Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Bee Venoms/pharmacology , Biological Therapy , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Viral/drug effects , Human papillomavirus 16/drug effects , Human papillomavirus 18/drug effects , Neoplasm Proteins/biosynthesis , Oncogene Proteins, Viral/biosynthesis , Papillomavirus E7 Proteins/biosynthesis , Papillomavirus Infections/pathology , Repressor Proteins/biosynthesis , Uterine Cervical Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Bee Venoms/therapeutic use , Carcinoma, Squamous Cell/virology , Cell Line, Transformed/transplantation , Cell Line, Tumor , Cell Transformation, Viral , DNA-Binding Proteins/genetics , Down-Regulation , Drug Screening Assays, Antitumor , Female , Genes, ras , HeLa Cells , Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Humans , Lung/cytology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Neoplasms, Experimental/therapy , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/virology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Random Allocation , Repressor Proteins/genetics , Uterine Cervical Neoplasms/virology
4.
J Clin Invest ; 122(4): 1487-502, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22406538

ABSTRACT

Epstein-Barr virus (EBV) persistently infects more than 90% of the human population and is etiologically linked to several B cell malignancies, including Burkitt lymphoma (BL), Hodgkin lymphoma (HL), and diffuse large B cell lymphoma (DLBCL). Despite its growth transforming properties, most immune-competent individuals control EBV infection throughout their lives. EBV encodes various oncogenes, and of the 6 latency-associated EBV-encoded nuclear antigens, only EBNA3B is completely dispensable for B cell transformation in vitro. Here, we report that infection with EBV lacking EBNA3B leads to aggressive, immune-evading monomorphic DLBCL-like tumors in NOD/SCID/γc-/- mice with reconstituted human immune system components. Infection with EBNA3B-knockout EBV (EBNA3BKO) induced expansion of EBV-specific T cells that failed to infiltrate the tumors. EBNA3BKO-infected B cells expanded more rapidly and secreted less T cell-chemoattractant CXCL10, reducing T cell recruitment in vitro and T cell-mediated killing in vivo. B cell lines from 2 EBV-positive human lymphomas encoding truncated EBNA3B exhibited gene expression profiles and phenotypic characteristics similar to those of tumor-derived lines from the humanized mice, including reduced CXCL10 secretion. Screening EBV-positive DLBCL, HL, and BL human samples identified additional EBNA3B mutations. Thus, EBNA3B is a virus-encoded tumor suppressor whose inactivation promotes immune evasion and virus-driven lymphomagenesis.


Subject(s)
Cell Transformation, Viral/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/physiology , Genes, Tumor Suppressor , Genes, Viral , Herpesvirus 4, Human/physiology , Lymphoma, B-Cell/virology , Lymphoproliferative Disorders/virology , Postoperative Complications/virology , Tumor Suppressor Proteins/physiology , Tumor Virus Infections/virology , Animals , Cell Line, Transformed/transplantation , Cell Line, Transformed/virology , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/deficiency , Chemokine CXCL10/genetics , Chimera , DNA Mutational Analysis , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Deletion , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Lymphoma, B-Cell/genetics , Lymphoproliferative Disorders/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mutation , Postoperative Complications/genetics , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics , Tumor Virus Infections/genetics
5.
Int J Oncol ; 40(2): 487-93, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21956144

ABSTRACT

We investigated whether a genetic modification of BCR-ABL-transformed mouse cells that resulted in endostatin (ES) production altered their oncogenic potential. Mouse B210 cells, which express p210bcr-abl fusion protein and induce leukemia-like disease and extremely rarely solid tumors after intravenous (i.v.) administration, were used. The cells were transfected with a plasmid carrying genes for mouse ES and resistance to blasticidine. Transduced cells were isolated in media supplemented with blasticidine. Production of ES was determined by Western blotting. For further tests, two clones were selected, and their pathogenicity after i.v. inoculation was tested. Compared with the parental B210 cells, the capability of both gene-modified cell clones to induce lethal leukemia was reduced. However, mice that did not succumb to leukemia subsequently developed solid tumors. They were composed of poorly differentiated cells with irregular nuclei and roughly granular chromatin and were well vascularized. FISH revealed the presence of the BCR-ABL fusion gene both in tumors and spleens. Immunohistological investigation of the tumors demonstrated the production of ES in vivo and the cell lines derived from the tumors produced detectable amounts of ES, this demonstrating that the formation of solid tumors was not associated with the loss or silencing of the ES gene.


Subject(s)
Cell Line, Transformed/metabolism , Endostatins/metabolism , Fusion Proteins, bcr-abl/genetics , Animals , Cell Line, Transformed/pathology , Cell Line, Transformed/transplantation , Cell Proliferation , Cell Transformation, Neoplastic , Culture Media, Conditioned/pharmacology , Endostatins/pharmacology , Female , Fusion Proteins, bcr-abl/metabolism , Histocompatibility Antigens Class I/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Neoplasm Transplantation
6.
DNA Cell Biol ; 30(12): 975-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21649506

ABSTRACT

DNA vaccines are known to be lacking in immunogenicity in humans. Presently, electroporation (EP) is thought to overcome this limitation. Here, we investigate whether human papillomavirus 16 E7 DNA vaccines delivered by EP might elicit potent antitumor activity in animal cervical cancer models, with a focus on the underlying mechanism(s). Intramuscular (IM)-EP delivery of E7 DNA vaccines induced more potent antitumor therapeutic and antimetastatic activity compared with IM delivery. Moreover, the tumor-controlled animals by IM-EP possessed long-term memory responses to parental tumor cells. This improved antitumor effect was concomitant with augmented Ag-specific CTL activities. IM-EP also induced IgG and Th-cell responses higher than IM delivery. Finally, IM-EP resulted in more antigen production in and more attraction of immune cells into the site of DNA injection, suggesting that these biological and immunological changes made by IM-EP might be responsible for enhanced CTL activities and antitumor resistance. Thus, this study shows that IM-EP can induce more potent antitumor activity by augmenting CTL responses possibly through more antigen production in and more attraction of immune cells into the muscle sites. This study also suggests that IM-EP of E7 DNA vaccines might be a potential approach toward treating patients with cervical cancer.


Subject(s)
Cancer Vaccines/therapeutic use , Human papillomavirus 16/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/therapeutic use , Animals , Cancer Vaccines/administration & dosage , Cell Line, Transformed/transplantation , Cell Line, Transformed/virology , Disease Models, Animal , Drug Screening Assays, Antitumor , Electroporation , Epithelial Cells/transplantation , Epithelial Cells/virology , Female , Humans , Immunization Schedule , Injections, Intramuscular , Interferon-gamma/analysis , Lung/cytology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/secondary , Neoplasms, Experimental/therapy , Papillomavirus Vaccines/administration & dosage , Subcutaneous Tissue , Uterine Cervical Neoplasms , Vaccines, DNA/administration & dosage , Vaccines, DNA/therapeutic use
7.
Cancer Biol Ther ; 9(2): 88-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19901563

ABSTRACT

Gankyrin, a small and highly conserved protein which is identical to the p28 gene product, was found to be related with the malignant phenotypes in liver and esophageal carcinoma. However, the roles of gankyrin in colorectal carcinoma (CRC) are still unknown. In the present study, the gankyrin mRNA and protein expression in human CRC cell lines and clinical tissue samples were evaluated and correlated with clinicopathological features. Possible mechanisms by which gankyrin regulates the malignant phenotype of CRC cells were also investigated. The results demonstrated that gankyrin was obviously overexpressed in CRC tissues and cell lines compared to controls, and gankyrin expression was correlated with TNM stages and metastasis of CRC. Overexpression of gankyrin by PhkitNeo-hGankyrin plasmid transfected into Lovo cells could promote the cell proliferation and tumorigenicity. This finding was further strengthened by experiments that suppressing gankyrin expression by siRNA exerted the opposite effects on CRC cells SW620. In addition, our present study showed that the co-expression of cyclinD1 and beta-catenin were positive correlation with the alteration of gankyrin expression. This data suggested that gankyrin played significant roles in the pathogenesis of human CRC, and might be an important therapeutic target for CRC.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/physiology , Proteasome Endopeptidase Complex/physiology , Proto-Oncogene Proteins/physiology , Adenocarcinoma/pathology , Aged , Animals , Blotting, Western , Cell Differentiation , Cell Line, Transformed/metabolism , Cell Line, Transformed/transplantation , Colorectal Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Staging , Neoplasm Transplantation , Polymerase Chain Reaction , Proteasome Endopeptidase Complex/biosynthesis , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/biosynthesis , Transfection
8.
Restor Neurol Neurosci ; 27(1): 67-78, 2009.
Article in English | MEDLINE | ID: mdl-19164854

ABSTRACT

PURPOSE: By using a nerve amputee model of the rat sciatic nerve (Lago and Navarro, 2007), we have tested a strategy for the long-term maintenance of regenerated axons without distal target reinnervation, by grafting Schwann cells (SCs) into a capped silicone chamber containing the ending nerve stump. METHODS: The sciatic nerve of rats was transected and repaired with a silicone tube, the distal nerve was again cut at 10 mm and inserted in a capped tube that was filled with saline or with a suspension of cultured SCs. Transplants of SCs obtained from primary cultures have been compared with those of an immortalized SC line (SCTM41) or the same line overexpressing GDNF. RESULTS: The histological results show that nerve fibers were able to regenerate through a short distal nerve segment ending into the capped chamber, and sustain distal branches without degenerating for several months. There was abundant axonal sprouting forming an ending neuroma, and the caliber of myelinated fibers remained far thinner than normal during the 9 months investigated. With a distal transplant of primary SCs there were significantly more regenerated myelinated fibers than in the control group at 9 months, indicating that the grafted cells stimulated the axonal growth response and helped to maintain survival of axon branches. In contrast, axonal regeneration was significantly reduced with grafts of SCTM41 cells, probably due to physical competition between cell proliferation and axonal growth. SCTM41 cells overexpressing GDNF improved the regenerative response with respect to the parent SCTM41 cells, although not to the same extent as the primary SCs. CONCLUSION: A graft of primary SCs in the capped chamber stimulated axonal growth response and/or maintained survival of axonal branches on the long term in the nerve amputee model.


Subject(s)
Nerve Regeneration , Schwann Cells/physiology , Schwann Cells/transplantation , Sciatic Neuropathy/surgery , Analysis of Variance , Animals , Axotomy/methods , Cell Line, Transformed/transplantation , Cells, Cultured , Disease Models, Animal , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Guided Tissue Regeneration , Nerve Fibers, Myelinated/physiology , Rats , Rats, Sprague-Dawley , S100 Proteins/metabolism , Sciatic Neuropathy/physiopathology , Time Factors
9.
Cancer Immunol Immunother ; 58(5): 737-48, 2009 May.
Article in English | MEDLINE | ID: mdl-18815785

ABSTRACT

Current therapeutic approaches to treatment of patients with bulky cervical cancer are based on conventional in situ ablative modalities including cisplatin-based chemotherapy and radiation therapy. The 5-year survival of patients with nonresectable disease is dismal. Because over 99% of squamous cervical cancer is caused by persistent infection with an oncogenic strain of human papillomavirus (HPV), particularly type 16 and viral oncoproteins E6 and E7 are functionally required for disease initiation and persistence, HPV-targeted immune strategies present a compelling opportunity in which to demonstrate proof of principle. Sublethal doses of radiation and chemotherapeutic agents have been shown to have synergistic effect in combination with either vaccination against cancer-specific antigens, or with passive transfer of tumor-specific cytotoxic T lymphocytes (CTLs). Here, we explored the combination of low-dose radiation therapy with DNA vaccination with calreticulin (CRT) linked to the mutated form of HPV-16 E7 antigen (E7(detox)), CRT/E7(detox) in the treatment of E7-expressing TC-1 tumors. We observed that TC-1 tumor-bearing mice treated with radiotherapy combined with CRT/E7(detox) DNA vaccination generated significant therapeutic antitumor effects and the highest frequency of E7-specific CD8(+) T cells in the tumors and spleens of treated mice. Furthermore, treatment with radiotherapy was shown to render the TC-1 tumor cells more susceptible to lysis by E7-specific CTLs. In addition, we observed that treatment with radiotherapy during the second DNA vaccination generated the highest frequency of E7-specific CD8(+) T cells in the tumors and spleens of TC-1 tumor-bearing mice. Finally, TC-1 tumor-bearing mice treated with the chemotherapy in combination with radiation and CRT/E7(detox) DNA vaccination generate significantly enhanced therapeutic antitumor effects. The clinical implications of the study are discussed.


Subject(s)
Immunotherapy, Active , Neoplasms, Experimental/therapy , Papillomavirus Vaccines/therapeutic use , Radiotherapy, Adjuvant/methods , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/therapeutic use , Animals , Apoptosis , Calreticulin/genetics , Calreticulin/immunology , Cell Line, Transformed/immunology , Cell Line, Transformed/transplantation , Cell Transformation, Viral , Combined Modality Therapy , Cytokines/analysis , Disease Models, Animal , Female , Lung , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/radiotherapy , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins , Radiotherapy Dosage , Recombinant Fusion Proteins/immunology , Uterine Cervical Neoplasms
10.
Behav Brain Res ; 194(2): 236-41, 2008 Dec 12.
Article in English | MEDLINE | ID: mdl-18672005

ABSTRACT

A human neuronal cell line, hNT2.19, which secretes serotonin (5-HT) after differentiation, was used as a transplant source to improve motor dysfunction following severe contusive spinal cord injury (SCI). Also, environmental enrichment (EE) was added to the interspinal transplant treatment paradigm. Motor testing was performed weekly before and following SCI, with and without EE and/or cell transplant conditions. Motor recovery was maximal when both cell transplant and EE were used. Individual treatment paradigms also significantly improved foot rotation and reduced footfall errors but not stride length or base of support dysfunction. This recovery of motor function after SCI suggests that the combinatory use of serotonergic hNT2.19 cell grafts plus EE is a meaningful strategy to modestly improve motor dysfunction that accompanies contusive SCI.


Subject(s)
Cell Transplantation/methods , Environment , Neurons/transplantation , Serotonin/metabolism , Spinal Cord Injuries/therapy , Animals , Cell Line, Transformed/transplantation , Disease Models, Animal , Exploratory Behavior/physiology , Female , Humans , Neurons/physiology , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Spinal Cord Injuries/physiopathology , Time Factors
11.
J Hepatol ; 47(1): 74-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17434229

ABSTRACT

BACKGROUND/AIMS: Hepatocyte transplantation and bioartificial liver treatment are attractive alternatives to liver transplantation. The availability of well-characterized human hepatocyte lines facilitates such cell therapies. METHODS: Human hepatocytes were immortalized with a retroviral vector SSR#197 expressing catalytic subunit of human telomerase reverse transcriptase (hTERT) and enhanced green fluorescent protein (EGFP) cDNAs flanked by a pair of loxP recombination targets. Then, Tamoxifen-dependent Cre recombinase was expressed in SSR#197-immortalized hepatocytes. Cre/LoxP recombination was performed in the established cells by simple exposure to 500 nM Tamoxifen for a week. Then, the reverted population of the cells was recovered by EGFP-negative cell sorting and characterized in vitro and in vivo using a pig model of acute liver failure (ALF) induced by d-galactosamine (0.5 g/kg) injection. RESULTS: A human hepatocyte cell line 16T-3 was established. Reverted 16-T3 cells showed the increased expression of hepatic markers in association with enhanced levels of transcriptional factors. Compared to normal human hepatocytes, albumin production and lidocaine-metabolizing activities of reverted 16-T3 cells were 0.32 and 0.50-fold, respectively. Transplantation of reverted 16T-3 cells significantly prolonged the survival of ALF pigs. CONCLUSIONS: Here we demonstrate the usefulness of Cre/LoxP -mediated reversible immortalization of human hepatocytes with Tamoxifen-mediated self-recombination.


Subject(s)
Cell Line, Transformed/transplantation , Hepatocytes/transplantation , Liver Failure/surgery , Animals , Biomarkers/analysis , Cell Line, Transformed/chemistry , Cell Line, Transformed/cytology , Hepatocytes/chemistry , Hepatocytes/drug effects , Humans , Integrases/genetics , Liver Failure/pathology , Recombination, Genetic , Retroviridae/genetics , Sus scrofa , Tamoxifen/pharmacology , Treatment Outcome
12.
PLoS One ; 2(2): e260, 2007 Feb 28.
Article in English | MEDLINE | ID: mdl-17327920

ABSTRACT

Inflammation in the tumor bed can either promote or inhibit tumor growth. Peroxisome proliferator-activated receptor (PPAR)alpha is a central transcriptional suppressor of inflammation, and may therefore modulate tumor growth. Here we show that PPARalpha deficiency in the host leads to overt inflammation that suppresses angiogenesis via excess production of the endogenous angiogenesis inhibitor thrombospondin-1 and prevents tumor growth. Bone marrow transplantation and granulocyte depletion show that PPARalpha expressing granulocytes are necessary for tumor growth. Neutralization of thrombospondin-1 restores tumor growth in PPARalpha-deficient mice. These findings suggest that the absence of PPARalpha activity renders inflammatory infiltrates tumor suppressive and, thus, may provide a target for inhibiting tumor growth by modulating stromal processes, such as angiogenesis.


Subject(s)
Granulocytes/physiology , Neoplasm Proteins/physiology , Neoplasms, Experimental/pathology , PPAR alpha/physiology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Line, Transformed/transplantation , Corneal Neovascularization/genetics , Inflammation , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/physiopathology , Neovascularization, Pathologic/prevention & control , PPAR alpha/deficiency , PPAR alpha/genetics , Radiation Chimera , Thrombospondin 1/physiology , Vascular Endothelial Growth Factor A/physiology
13.
Neoplasia ; 8(3): 231-41, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16611417

ABSTRACT

Exclude experimental models of malignant transformation employ chemical and physical carcinogens or genetic manipulations to study tumor progression. In this work, different melanoma cell lines were established after submitting a nontumorigenic melanocyte lineage (melan-a) to sequential cycles of forced anchorage impediment. The great majority of these cells underwent anoikis when maintained in suspension. After one deadhesion cycle, phenotypic alterations were noticeable in the few surviving cells, which became more numerous and showed progressive alterations after each adhesion impediment step. No significant differences in cell surface expression of integrins were detected, but a clear electrophoretic migration shift, compatible with an altered glycosylation pattern, was observed for beta1 chain in transformed cell lines. In parallel, a progressive enrichment of tri- and tetra-antennary N-glycans was apparent, suggesting increased N-acetylglucosaminyltransferase V activity. Alterations both in proteoglycan glycosylation pattern and core protein expression were detected during the transformation process. In conclusion, this model corroborates the role of adhesion state as a promoting agent in transformation process and demonstrates that cell adhesion disturbances may act as carcinogenic stimuli, at least for a nontumorigenic immortalized melanocyte lineage. These findings have intriguing implications for in vivo carcinogenesis, suggesting that anchorage independence may precede, and contribute to, neoplastic conversion.


Subject(s)
Anoikis , Cell Transformation, Neoplastic , Melanocytes/cytology , Melanoma, Experimental/pathology , Animals , Cell Adhesion , Cell Line, Transformed/transplantation , Cell Lineage , Cells, Cultured/cytology , Chondroitin Sulfate Proteoglycans/biosynthesis , Chondroitin Sulfate Proteoglycans/genetics , Culture Media, Serum-Free , Decorin , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Female , Fibronectins , Flow Cytometry , Gene Expression Regulation, Neoplastic , Glucuronidase/biosynthesis , Glucuronidase/genetics , Glycosaminoglycans/analysis , Heparan Sulfate Proteoglycans/biosynthesis , Heparan Sulfate Proteoglycans/genetics , Integrins/metabolism , Laminin , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Melanocytes/metabolism , Melanocytes/transplantation , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Phenotype , Proteoglycans/biosynthesis , Proteoglycans/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Versicans
14.
Cancer Lett ; 235(1): 53-9, 2006 Apr 08.
Article in English | MEDLINE | ID: mdl-15899546

ABSTRACT

Several tumor animal models have been provided as a tool for developing cancer therapy. Here, we developed rapid, easy-to use, and cost-effective new rat animal model for invasion and metastasis of cancer using genetically k-ras-induced rat kidney cells (RK3E-ras). We observed tumor as early as 3 days after injection of RK3E-ras cells in subcutaneous of Sprague-Dawley rats. Tumor size and volume were increased exponentially for 2 weeks. The tail vein injected rats obtained the lethal infiltration in the lung within 2 weeks. This tumor animal model has great potential for studying cancer processes and short-term screening of variable cancer therapy strategy.


Subject(s)
Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Genes, ras/physiology , Kidney/metabolism , Neoplasms, Experimental/pathology , Animals , Cell Line, Transformed/transplantation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Injections, Subcutaneous , Kidney/pathology , Male , Neoplasm Invasiveness , Neoplasms, Experimental/etiology , Rats , Rats, Sprague-Dawley
15.
J Neurosci Methods ; 152(1-2): 250-4, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16257450

ABSTRACT

A loss of neostriatal neurons is a characteristic of Huntington's disease (HD), and neural tissue transplantation has been performed directly into the striatum. Since the neural stem cells have ability to migrate into the lesion site, we administered immortalized neural stem-like cells (NSC) into the ventricle or via a tail vein following unilateral intrastriatal quinolinic acid lesioning in Sprague-Dawley rats. To identify transplanted NSC, cells were encoded with lac Z and beta-galactosidase histochemistry was performed. lac Z+ cells were detected in the lesioned striatum but tissue damage or tumor formation was not observed. This study shows that NSC migrate into the striatum, either from ventricle or from the systemic circulation, providing less invasive routes for stem cell application in HD.


Subject(s)
Cell Line, Transformed/transplantation , Huntington Disease/therapy , Neurons/transplantation , Stem Cell Transplantation/methods , Animals , Cell Movement , Humans , Huntington Disease/chemically induced , Immunohistochemistry , Immunosuppression Therapy , Injections, Intraventricular , Lac Operon/genetics , Male , Quinolinic Acid/toxicity , Rats , Rats, Sprague-Dawley , Stereotaxic Techniques
16.
Antivir Ther ; 11(6): 717-27, 2006.
Article in English | MEDLINE | ID: mdl-17310816

ABSTRACT

In this study, we demonstrate that fusion-active virosomes, containing recombinant human papillomavirus type 16 (HPV16) E7 protein antigen, are capable of inducing a robust class I MHC-restricted cytotoxic T-lymphocyte (CTL) response against HPV-transformed tumour cells in a murine model system. Virosomes are reconstituted viral envelopes, which do not contain the genetic material of the native virus. During the reconstitution process, protein antigens can be encapsulated within the virosomes. In the present study, we used virosomes derived from influenza virus. These virosomes retain the cell binding and membrane fusion characteristics of native influenza virus, and have the capacity to deliver encapsulated antigens to the cytosol of antigen-presenting cells through fusion from within acidic endosomes. After immunization of mice with virosomes containing encapsulated HPV16 E7 protein, the animals developed a strong E7-specific CTL response as assessed by 51Cr release measurements and MHC tetramer staining of spleen cells. Immunization with E7-containing virosomes also resulted in E7-specific antibody responses. In tumour challenge experiments, immunization of mice with E7-containing virosomes prevented tumour outgrowth in >70% of the animals. Thus, influenza-derived virosomes with encapsulated HPV E7 protein antigen act as an excellent vaccine delivery system for induction of cellular immunity against HPV-transformed cells and represent a promising immunotherapeutic vaccine for the treatment of (precursor lesions of) cervical cancer.


Subject(s)
Cancer Vaccines/administration & dosage , Oncogene Proteins, Viral/administration & dosage , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Dysplasia/prevention & control , Uterine Cervical Neoplasms/prevention & control , Vaccines, Virosome/administration & dosage , Animals , Antibodies, Viral/blood , Cancer Vaccines/immunology , Cell Line, Transformed/transplantation , Cell Line, Tumor , Female , Histocompatibility Antigens Class I/metabolism , Human papillomavirus 16/immunology , Humans , Immunization , Mice , Mice, Inbred C57BL , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , T-Lymphocytes, Cytotoxic/immunology , Uterine Cervical Neoplasms/virology , Vaccines, Virosome/immunology , Uterine Cervical Dysplasia/virology
17.
Folia Biol (Praha) ; 51(1): 12-8, 2005.
Article in English | MEDLINE | ID: mdl-15783087

ABSTRACT

In an effort to develop an experimental system suitable for immunological studies in which Bcr-Abl-positive cells are to be used as antigens, we examined the properties of two mouse (Balb/c) established cell lines that express the Bcr-Abl protein and are oncogenic for syngeneic animals. Under standard conditions the two cell lines, viz. Ba-p210 (B210) and 12B1, expressed comparable amounts of the Bcr-Abl protein. However, they differed in a number of characteristics. From the morphological point of view, B210 cells were the more homogeneous, being mainly represented by leukaemic blastic cells with a large number of AgNORs as markers indicating a high proliferative activity. 12B1 cells were more polymorphic and giant cells were detected within their populations. Many 12B1 cells exhibited nuclear segmentation and "band-like" structures. Markers of proliferation were less frequent in 12B1 and the tendency for aging was more pronounced in these cells. The 12B1 cells were slightly more sensitive to imatinib mesylate than B210 cells. In B210 cells, the expression of MHC class I was downregulated, which was not the case with 12B1 cells. Both cell lines induced leukaemia-like disease in mice after intravenous application but, as compared with B210, 12B1 cells were about 100 times more oncogenic and the disease they induced was more aggressive. Moreover, 12B1, but not B210, induced tumours after subcutaneous or intraperitoneal inoculation.


Subject(s)
Cell Line, Transformed/metabolism , Cell Line, Transformed/transplantation , Cell Transformation, Neoplastic/metabolism , Fusion Proteins, bcr-abl/metabolism , Leukemia/metabolism , Animals , Antineoplastic Agents/pharmacology , Benzamides , Biomarkers, Tumor/metabolism , Cell Line, Transformed/pathology , Cell Proliferation/drug effects , Cell Shape , Cellular Senescence/physiology , Down-Regulation/physiology , Drug Resistance, Neoplasm/physiology , Fusion Proteins, bcr-abl/genetics , Histocompatibility Antigens Class I/metabolism , Imatinib Mesylate , Leukemia/drug therapy , Leukemia/pathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness/physiopathology , Neoplasm Transplantation , Piperazines/pharmacology , Pyrimidines/pharmacology
18.
Mol Syst Biol ; 1: 2005.0022, 2005.
Article in English | MEDLINE | ID: mdl-16729057

ABSTRACT

Deciphering regulatory events that drive malignant transformation represents a major challenge for systems biology. Here, we analyzed genome-wide transcription profiling of an in vitro cancerous transformation process. We focused on a cluster of genes whose expression levels increased as a function of p53 and p16(INK4A) tumor suppressors inactivation. This cluster predominantly consists of cell cycle genes and constitutes a signature of a diversity of cancers. By linking expression profiles of the genes in the cluster with the dynamic behavior of p53 and p16(INK4A), we identified a promoter architecture that integrates signals from the two tumor suppressive channels and that maps their activity onto distinct levels of expression of the cell cycle genes, which, in turn, correspond to different cellular proliferation rates. Taking components of the mitotic spindle as an example, we experimentally verified our predictions that p53-mediated transcriptional repression of several of these novel targets is dependent on the activities of p21, NFY, and E2F. Our study demonstrates how a well-controlled transformation process allows linking between gene expression, promoter architecture, and activity of upstream signaling molecules.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Gene Expression Profiling , Genes, Tumor Suppressor , Genes, cdc , Promoter Regions, Genetic/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cell Cycle Proteins/physiology , Cell Division , Cell Line, Transformed/metabolism , Cell Line, Transformed/transplantation , Computational Biology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Genes, p16 , Genes, p53 , Humans , Mice , Mice, Nude , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/physiology , Regulatory Sequences, Nucleic Acid , Spindle Apparatus/metabolism , Telomerase/genetics , Telomerase/physiology , Transcription, Genetic , Transplantation, Heterologous
19.
Circulation ; 110(11 Suppl 1): II219-24, 2004 Sep 14.
Article in English | MEDLINE | ID: mdl-15364866

ABSTRACT

BACKGROUND: Poor survival of grafted cells is a major factor hindering the therapeutic effect of cell transplantation; however, the causes of cell death remain unclear. We hypothesized that interleukin-1beta (IL-1beta) might play a role in the acute inflammatory response and graft death after cell transplantation and that inhibition of IL-1beta might improve graft survival. METHODS AND RESULTS: 14C-labeled male skeletal muscle precursor cells were implanted into female mouse hearts by direct intramuscular injection. The amount of 14C-label provides an estimate of the surviving cell number, whereas the amount of male-specific Smcy gene measured by polymerase chain reaction indicates the total (surviving+proliferated) number of donor-derived cells. At 10 minutes after implantation, 44.8+/-2.4% of the grafted cells survived and this steadily decreased to 14.6+/-1.1% by 24 hours, and to 7.9+/-0.6% by 72 hours (n=6 in each point). Proliferation of the surviving cells, which began after 24 hours, resulted in an increase in the total cell number from 15.5+/-0.8% at 24 hours to 24.4+/-1.6% at 72 hours. Acute inflammation was prominent at 24 hours and was reduced by 72 hours, in parallel with IL-1beta expression. Administration of anti-IL-1beta antibody improved graft survival at both 24 (25.6+/-1.6%) and 72 hours (14.8+/-1.1%) and resulted in a 2-fold increase in the total cell number at 72 hours (45.8+/-2.4%). The effects of IL-1beta inhibition corresponded with a reduced inflammatory response. CONCLUSIONS: IL-1beta is involved in acute inflammation and graft death after direct intramyocardial cell transplantation. Targeted inhibition of IL-1beta may be a useful strategy to improve graft survival.


Subject(s)
Interleukin-1/physiology , Myoblasts/transplantation , Myocarditis/etiology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biomarkers , Cell Differentiation , Cell Division , Cell Line, Transformed/transplantation , Cell Survival/drug effects , Cell Transplantation/adverse effects , Female , Graft Survival/drug effects , Histone Demethylases , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Interleukin-1/genetics , Male , Mice , Myoblasts/pathology , Myocarditis/drug therapy , Myocarditis/prevention & control , Myocardium/metabolism , Peroxidase/analysis , Proteins/analysis , Reverse Transcriptase Polymerase Chain Reaction
20.
Expert Opin Biol Ther ; 4(9): 1423-34, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15335310

ABSTRACT

Chronic granulomatous disease (CGD) is a congenital immune deficiency that is a promising therapeutic target for gene replacement into haematopoietic stem cells (HSCs). CGD results from mutations in any one of four genes encoding subunits of the superoxide-generating NADPH oxidase of phagocytes. Life-threatening, recurrent bacterial and fungal infections, as well as inflammatory granulomas, are the hallmarks of the disease. NADPH oxidase activity can be reconstituted by retroviral- or lentiviral-mediated gene transfer to human CGD marrow in vitro and in xenograft transplant models. Gene transfer studies in knockout mouse models that resemble the human disease suggest that correction of oxidase activity in a minority of phagocytes will be of clinical benefit. Phase I clinical studies in unconditioned CGD patients showed transient expression of small numbers of gene-corrected neutrophils. Areas of research at present include efforts to enhance gene transfer rates into repopulating HSCs using vectors that transduce quiescent cells, and to increase the engraftment of genetically corrected HSCs using non-myeloablative conditioning and drug resistance genes for selection.


Subject(s)
Genetic Therapy , Granulomatous Disease, Chronic/therapy , Animals , Bone Marrow Cells/enzymology , Bone Marrow Transplantation , Cell Line, Transformed/enzymology , Cell Line, Transformed/transplantation , Cells, Cultured/enzymology , Cells, Cultured/transplantation , Clinical Trials, Phase I as Topic , Disease Susceptibility , Dosage Compensation, Genetic , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/immunology , Hematopoietic Stem Cell Transplantation , Humans , Infections/etiology , Inflammation/etiology , Mice , Mice, Knockout , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Phagocytes/enzymology , Phagocytosis , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...