Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 495(1): 962-968, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29155177

ABSTRACT

Although gemcitabine is an effective chemotherapeutic for pancreatic cancer, severe side effects often accompany its use. Since we have discovered that locally administered C1B domain peptides effectively control tumor growth without any side effects, the efficacy of co-treatment with this peptide and a low dose of gemcitabine on the growth of pancreatic cancer was examined. Two- and three-dimensional cell culture studies clarified that a co-treatment with C1B5 peptide and gemcitabine significantly attenuated growth of PAN02 mouse and PANC-1 human pancreatic cancer cells in 2D and 3D cultures. Although treatment with the low dose of gemcitabine alone (76%) or the C1B5 peptide alone (39%) inhibited tumor growth moderately, a co-treatment with C1B5 peptide and a low dose of gemcitabine markedly inhibited the growth of PAN02 autografts in the mouse peritoneal cavity (94% inhibition) without any noticeable adverse effect. The number of peritoneal cavity-infiltrating neutrophils and granzyme B+ lymphocytes was significantly higher in the co-treatment group than in the control group. A significant increase of granzyme B mRNA expression was also detected in human T cells by the co-treatment. Taken together, the current study suggests that C1B5 peptide offers a remarkably effective combination treatment strategy to reduce side effects associated with gemcitabine, without losing its tumoricidal effect.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Peptide Fragments/administration & dosage , Protein Kinase C/administration & dosage , T-Lymphocytes/drug effects , Animals , Cell Line, Tumor , Deoxycytidine/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Protein Kinase C/chemistry , Gemcitabine
2.
PLoS One ; 12(4): e0175064, 2017.
Article in English | MEDLINE | ID: mdl-28380056

ABSTRACT

The newly purified extracellular polysaccharides (exopolysaccharides) from Parachlorella kessleri (PCEPS) were evaluated on their antitumor and immunomodulatory effects in cell culture and mouse colon carcinoma peritoneal dissemination model. In two-dimensional cell culture, the PCEPS treatment inhibited cell growth of both murine and human colon carcinoma cells in a dose- and time-dependent manner. In contrast, the growth of mouse splenocytes (SPLs) and bone marrow cells (BMCs) were stimulated by the treatment with PCEPS. The treatment with PCEPS also increased specific subpopulations of the cells in BMCs: antigen presenting cells (CD19+ B cells, 33D1+ dendritic cells and CD68+ macrophage) and CD8+ cytotoxic T cells. In three-dimensional spheroid culture, spheroid growth of CT26 cells co-cultured with HL-60 human neutrophilic promyeloblasts and Jurkat cells (human lymphoblasts), but not THP-1 human monocyte/macrophage was significantly attenuated by PCEPS treatment. In a mouse CT26 colon carcinoma peritoneal dissemination model, intraperitoneal injection of PCEPS (10 mg/kg, twice per week) significantly attenuated the growth of CT26 colon carcinoma in syngeneic mice. The present study suggests that PCEPS inhibits colon carcinoma growth via direct cell growth inhibition and a stimulation of the host antitumor immune responses. Taken together, the current study suggests that exopolysaccharides derived from Parachlorella kessleri contain significant bioactive materials that inhibit colon carcinoma growth.


Subject(s)
Antineoplastic Agents/therapeutic use , Chlorella/chemistry , Colonic Neoplasms/drug therapy , Immunologic Factors/therapeutic use , Plant Extracts/therapeutic use , Polysaccharides/therapeutic use , Animals , Bone Marrow Cells/drug effects , Cell Line, Tumor , Chlorophyta , Dose-Response Relationship, Drug , Female , Humans , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/drug effects
3.
J Pharm Sci ; 106(1): 385-394, 2017 01.
Article in English | MEDLINE | ID: mdl-27769520

ABSTRACT

To evaluate the potential of cell-penetrating peptide-based delivery of apoptosis-inducer gene in cancer therapy, a modified HIV-1 TAT peptide (dimerized TAT peptide, dTAT) was studied. The dTAT and plasmid DNA (pDNA) complexes (dTAT-pDNA) were condensed using calcium chloride (dTAT-pDNA-Ca2+). This simple nonviral formulation approach showed high levels of gene expression in vitro without any cytotoxicity. In mouse studies, a single intratracheal (IT) aerosol spray or 2 intravenous (IV) injections of the dTAT, apoptosis-inducer gene, angiotensin II type 2 receptor (AT2R), and Ca2+ complexes (dTAT-pAT2R-Ca2+) significantly attenuated the acutely growing mouse Lewis lung carcinoma allografts in mouse lungs. Furthermore, single IT (p = 0.054) and the combination of IT and IV (p < 0.05) administrations of dTAT-pAT2R-Ca2+ markedly attenuated slowly growing and relatively large-sized H358 human bronchioloalveolar carcinoma xenografts in mouse lungs. These results indicate that the dTAT-pDNA-Ca2+ effectively delivered the gene to cancer cells by either IT or IV administration although the local pulmonary delivery of the dTAT-pAT2R-Ca2+ showed more effective growth inhibition of orthotopic lung cancer grafts. Thus, the present study offers preclinical proof of concept that a dTAT-based nonviral gene delivery method via IT administration may be an effective lung cancer gene therapy.


Subject(s)
Carcinoma, Lewis Lung/therapy , DNA/administration & dosage , Gene Transfer Techniques , Lung Neoplasms/therapy , Nanoparticles/chemistry , Receptor, Angiotensin, Type 2/genetics , tat Gene Products, Human Immunodeficiency Virus/chemistry , Animals , Carcinoma, Lewis Lung/genetics , Cell Line, Tumor , DNA/genetics , DNA/therapeutic use , Gene Expression , Genetic Therapy , HIV-1/chemistry , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/therapeutic use
4.
Pharm Res ; 33(10): 2517-29, 2016 10.
Article in English | MEDLINE | ID: mdl-27335023

ABSTRACT

PURPOSE: To determine aerosol administration capability and therapeutic efficacy of the new formulation of hyaluronan cisplatin conjugates, HylaPlat™ (HA-Pt), for lung cancer treatment. METHODS: In vitro formulation stability test, 2D and 3D spheroid cell culture and in vivo efficacy studies using mouse orthotopic allograft models were conducted. RESULTS: The HA-Pt effectively attenuated cell growth in 2D and 3D cultures with IC50 of 2.62 and 5.36 µM, respectively, which were comparable to those with unconjugated control cisplatin-dependent growth inhibition (IC50 1.64 and 4.63 µM, respectively). A single dose of either 7.5 or 15 mg/kg HA-Pt (cisplatin equivalent) by intratracheal aerosol spray 7 days after Lewis lung carcinoma (LLC) cell inoculation markedly inhibited growth of LLC allografts in mouse lungs and resulted in a 90 or 94% reduction of tumor nodule numbers, respectively, as compared to those from the PBS control. Cancer stem cells and cisplatin resistant cells marker, CD44 expression decreased in the tumor nodules of the HA-Pt but not in those of cisplatin treated groups. CONCLUSIONS: The current study suggests that an intratracheal aerosol administration of the HA-Pt nanoparticles offers an effective strategy for lung cancer treatment and this treatment may induce only limited cisplatin resistance.


Subject(s)
Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Hyaluronic Acid/administration & dosage , Lung Neoplasms/drug therapy , Nanoparticles/administration & dosage , Trachea/metabolism , A549 Cells , Administration, Inhalation , Animals , Antineoplastic Agents/metabolism , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cisplatin/metabolism , Drug Administration Routes , Female , Humans , Hyaluronic Acid/metabolism , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Nanoparticles/metabolism , Trachea/drug effects , Tumor Cells, Cultured
5.
Mol Cancer Ther ; 15(1): 209-18, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26637367

ABSTRACT

Transfection efficiency and toxicity concerns remain a challenge for gene therapy. Cell-penetrating peptides (CPP) have been broadly investigated to improve the transfection of genetic material (e.g., pDNA and siRNA). Here, a synthetic CPP (polylysine, K9 peptide) was complexed with angiotensin II type 2 receptor (AT2R) plasmid DNA (pAT2R) and complexes were condensed using calcium chloride. The resulting complexes were small (∼150 nm) and showed high levels of gene expression in vitro and in vivo. This simple nonviral formulation approach showed negligible cytotoxicity in four different human cell lines (cervix, breast, kidney, and lung cell lines) and one mouse cell line (a lung cancer cell line). In addition, this K9-pDNA-Ca(2+) complex demonstrated cancer-targeted gene delivery when administered via intravenous injection or intratracheal spray. The transfection efficiency was evaluated in Lewis lung carcinoma (LLC) cell lines cultured in vitro and in orthotopic cancer grafts in syngeneic mice. Immunohistochemical analysis confirmed that the complex effectively delivered pAT2R to the cancer cells, where it was expressed mainly in cancer cells along with bronchial epithelial cells. A single administration of these complexes markedly attenuated lung cancer growth, offering preclinical proof-of-concept for a novel nonviral gene delivery method exhibiting effective lung tumor gene therapy via either intravenous or intratracheal administration.


Subject(s)
Carcinoma, Lewis Lung/genetics , Gene Transfer Techniques , Plasmids/genetics , Polylysine , Receptor, Angiotensin, Type 2/genetics , Allografts , Animals , Apoptosis/drug effects , Calcium/chemistry , Carcinoma, Lewis Lung/pathology , Carcinoma, Lewis Lung/therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Carriers , Gene Expression , Genetic Therapy , Humans , Imines/chemistry , Injections, Intravenous , Male , Mice , Plasmids/administration & dosage , Polyethylenes/chemistry , Polylysine/chemistry , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
6.
PLoS One ; 10(5): e0123756, 2015.
Article in English | MEDLINE | ID: mdl-25942583

ABSTRACT

Human and rat umbilical cord matrix mesenchymal stem cells (UCMSC) possess the ability to control the growth of breast carcinoma cells. Comparative analyses of two types of UCMSC suggest that rat UCMSC-dependent growth regulation is significantly stronger than that of human UCMSC. Their different tumoricidal abilities were clarified by analyzing gene expression profiles in the two types of UCMSC. Microarray analysis revealed differential gene expression between untreated naïve UCMSC and those co-cultured with species-matched breast carcinoma cells. The analyses screened 17 differentially expressed genes that are commonly detected in both human and rat UCMSC. The comparison between the two sets of gene expression profiles identified two tumor suppressor genes, adipose-differentiation related protein (ADRP) and follistatin (FST), that were specifically up-regulated in rat UCMSC, but down-regulated in human UCMSC when they were co-cultured with the corresponding species' breast carcinoma cells. Over-expression of FST, but not ADRP, in human UCMSC enhanced their ability to suppress the growth of MDA-231 cells. The growth of MDA-231 cells was also significantly lower when they were cultured in medium conditioned with FST, but not ADRP over-expressing human UCMSC. In the breast carcinoma lung metastasis model generated with MDA-231 cells, systemic treatment with FST-over-expressing human UCMSC significantly attenuated the tumor burden. These results suggest that FST may play an important role in exhibiting stronger tumoricidal ability in rat UCMSC than human UCMSC and also implies that human UCMSC can be transformed into stronger tumoricidal cells by enhancing tumor suppressor gene expression.


Subject(s)
Apoptosis/physiology , Breast Neoplasms/physiopathology , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cells, Cultured , Cluster Analysis , Female , Follistatin/metabolism , Humans , Membrane Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Perilipin-2 , Rats , Real-Time Polymerase Chain Reaction
7.
Mol Pharm ; 8(5): 1549-58, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21851062

ABSTRACT

Rat umbilical cord matrix stem cells (UCMSC) have been shown to exhibit a remarkable ability to control rat mammary adenocarcinoma (Mat B III) cell proliferation both in vivo and in vitro. To study the underlying mechanisms and genes involved in Mat B III growth attenuation, total RNA was extracted from the naive rat UCMSC alone and those cocultured with Mat B III in Transwell culture dishes. Gene expression profiles of naive rat UCMSC alone and those cocultured with Mat B III cells were investigated by microarray analysis using an Illumina RatRef-12 Expression BeadChip. The comparison of gene expression profiles between untreated and cocultured rat UCMSC identified five upregulated candidate genes (follistatin (FST), sulfatase1 (SULF-1), glucose phosphate isomerase (GPI), HtrA serine peptidase (HTRA1), and adipocyte differentiation-related protein (ADRP)) and two downregulated candidate genes (transforming growth factor, beta-induced, 68 kDa (TGFßI) and podoplanin (PDPN)) based upon the following screening criteria: (1) expression of the candidate genes should show at least a 1.5-fold change in rat UCMSC cocultured with Mat B III cells; (2) candidate genes encode secretory proteins; and (3) they encode cell growth-related proteins. Following confirmation of gene expression by real-time PCR, ADRP, SULF-1 and GPI were selected for further analysis. Addition of specific neutralizing antibodies against these three gene products or addition of gene-specific siRNA's individually in cocultures of 1:20 rat UCMSC:Mat B III cells significantly increased cell proliferation, implying that these gene products are produced under the cocultured condition and functionally attenuate cell growth. Immunoprecipitation followed by Western blot analysis demonstrated that these proteins are indeed secreted into the culture medium. Individual overexpression of these three genes in rat UCMSC significantly enhanced UCMSC-dependent inhibition of cell proliferation in coculture. These results suggest that ADRP, SULF-1 and GPI act as tumor suppressor genes, and these genes might be involved in rat UCMSC-dependent growth attenuation of rat mammary tumors.


Subject(s)
Embryonic Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism , Umbilical Cord/cytology , Adenocarcinoma/therapy , Animals , Breast Neoplasms/therapy , Cell Communication , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Embryonic Stem Cells/transplantation , Female , Gene Expression Profiling , Glucose-6-Phosphate Isomerase/antagonists & inhibitors , Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Perilipin-2 , Pregnancy , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Rats , Rats, Inbred F344 , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/genetics , Sulfotransferases/metabolism , Tumor Suppressor Proteins/genetics
8.
BMC Cancer ; 10: 67, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181281

ABSTRACT

BACKGROUND: Pancreatic cancer is one of the most aggressive human malignancies, with a very poor prognosis. To evaluate the effect of angiotensin II (Ang II) type 2 receptor (AT2) expression in the host's body on the growth of pancreatic carcinoma, we have investigated the growth of mouse pancreatic ductal carcinoma grafts in syngeneic wild type and AT2 receptor-deficient (AT2-KO) mice. METHODS: The role of AT2 receptor-signaling in stromal cells on the growth of murine pancreatic carcinoma cells (PAN02) was studied using various in vitro and in vivo assays. In vivo cell proliferation, apoptosis, and vasculature in tumors were monitored by Ki-67 immunostaining, TUNEL assay, and von Willebrand factor immunostaining, respectively. In the co-culture study, cell proliferation was measured by MTT cell viability assay. All the data were analyzed using t-test and data were treated as significant when p < 0.05. RESULTS: Our results show that the growth of subcutaneously transplanted syngeneic xenografts of PAN02 cells, mouse pancreatic ductal carcinoma cells derived from the C57/BL6 strain, was significantly faster in AT2-KO mice compared to control wild type mice. Immunohistochemical analysis of tumor tissue revealed significantly more Ki-67 positive cells in xenografts grown in AT2-KO mice than in wild type mice. The index of apoptosis is slightly higher in wild type mice than in AT2-KO mice as evaluated by TUNEL assay. Tumor vasculature number was significantly higher in AT2-KO mice than in wild type mice. In vitro co-culture studies revealed that the growth of PAN02 cells was significantly decreased when grown with AT2 receptor gene transfected wild type and AT2-KO mouse-derived fibroblasts. Faster tumor growth in AT2-KO mice may be associated with higher VEGF production in stromal cells. CONCLUSIONS: These results suggest that Ang II regulates the growth of pancreatic carcinoma cells through modulating functions of host stromal cells; Moreover, Ang II AT2 receptor signaling is a negative regulator in the growth of pancreatic carcinoma cells. These findings indicate that the AT2 receptor in stromal fibroblasts is a potentially important target for chemotherapy for pancreatic cancer.


Subject(s)
Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/pathology , Receptor, Angiotensin, Type 2/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Coculture Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...