Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Xenotransplantation ; 28(3): e12673, 2021 05.
Article in English | MEDLINE | ID: mdl-33522023

ABSTRACT

BACKGROUND: Alginate-encapsulated islet xenografts have restored normoglycemia in diabetic animals for various periods of time. Plausible mechanisms of graft failure in vivo include immune rejection and hypoxia. We sought to understand the effects of encapsulated adult porcine islet (API) dosage on the peritoneal dissolved oxygen (DO) level in correlation to the achieved glycemic regulation in diabetic mice. METHODS: Adult porcine islets encapsulated in barium alginate were transplanted intraperitoneally in streptozotocin diabetic BALB/c mice at 6000 and 4000 islet equivalents (IEQ) and in normal mice at 500 IEQ; APIs encapsulated in calcium alginate were transplanted at 6000 IEQ in diabetic mice. In all cases, cell-free barium alginate capsules containing a perfluorocarbon emulsion were co-implanted for DO measurements using 19 F NMR spectroscopy. Blood glucose levels and peritoneal DO were measured over 60 days or until graft failure. Explanted capsules were evaluated microscopically and histologically. RESULTS: Both barium and calcium alginate-encapsulated APIs at 6000 IEQ reversed diabetes until day 60; barium alginate-encapsulated APIs at 4000 IEQ also reversed diabetes but with a higher failure rate. Transplanted APIs significantly reduced the peritoneal DO, approximately in a dose-dependent manner. The number of viable islets and the insulin content per capsule decreased over time. Capsules retrieved from normoglycemic mice exhibited minimal host cell adherence. CONCLUSIONS: Transplantation of encapsulated APIs can reduce peritoneal DO to severely hypoxic levels. Although normoglycemia could be maintained within the study period, the DO levels suggest that hypoxia is a factor contributing to loss of islet viability and insulin secretion with time in mice.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans Transplantation , Islets of Langerhans , Alginates , Animals , Graft Survival , Mice , Mice, Inbred BALB C , Oxygen , Streptozocin , Swine , Transplantation, Heterologous
2.
World J Surg ; 38(6): 1251-61, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24549997

ABSTRACT

BACKGROUND: Insulin-secreting beta-like cells are vulnerable to diabetic autoimmunity. We hypothesized that human thyroid neuroendocrine (NE) cells could be engineered to secrete human insulin, be glucose-responsive, and avoid autoimmunity. METHODS: Collagenase-digested thyroid tissue was cultured and subjected to size-based fluorescence-activated cell sorting. Insulin secretion and storage in NE cells transduced with viral vectors carrying an insulin sequence was assessed by enzyme-linked immunosorbent assay (ELISA) and immunogold transmission electron microscopy (TEM). Baseline mRNA expression was assessed by Illumina expression array analysis. Transduction with retrovirus expressing transcription factors PDX1, NGN3, MAFA, or HNF6 altered mRNA expression in a custom polymerase chain reaction (PCR) array. Gastrin-releasing peptide (GRP) in conditioned medium and cell lysates was determined by reverse transcription (RT)-PCR, ELISA, and immunohistochemistry. RESULTS: Isolation yielded an average of 2.2 × 10(6) cells/g thyroid tissue, which stained for calcitonin/calcitonin gene-related protein, expressed genes consistent with NE origins, and secreted GRP. Transduced cells secreted 56 % and retained 48 % of total insulin produced. Immunogold TEM revealed insulin in secretory vesicles. PDX1, NGN3, and MAFA overexpression increased expression of genes typical for hepatocytes and beta cells. Overexpression of HNF6 also increased the message of genes critical for glucose sensing. CONCLUSIONS: Human thyroid NE cells can produce human insulin, fractions of which are both secreted and retained in secretory granules. Overexpression of HNF6, PDX1, or NGN3 enhances expression of both hepatocyte and beta cell typical mRNAs, including the message of proteins critical for glucose sensing. These data suggest that reimplantation of engineered autologous NE cells may develop as a viable treatment for diabetes mellitus type 1.


Subject(s)
Bioengineering/methods , Hepatocyte Nuclear Factor 6/metabolism , Insulin/pharmacology , Neuroendocrine Cells/metabolism , Thyroid Gland/cytology , Cells, Cultured , Diabetes Mellitus, Type 1/drug therapy , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Hepatocyte Nuclear Factor 6/genetics , Humans , Insulin/therapeutic use , Insulin-Secreting Cells/metabolism , Microscopy, Electron, Transmission , Neuroendocrine Cells/cytology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Thyroid Gland/metabolism
3.
Surgery ; 154(6): 1385-93; discussion 1393, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24206618

ABSTRACT

BACKGROUND: Human pituitary adenomas express folate receptors (FR); therefore, we hypothesized that parathyroid (PT) tumors also might express FR, whereas normal human thyroids might not. The purpose of our study was to characterize the functionality of FRs on human PT tumors, with the goal of developing an imaging tool that would concentrate in PT more than in the thyroid. METHODS: Human PTs and thyroids were evaluated for FR expression by immunohistochemistry. Expression of genes for FRα and FRß was measured with the Illumina Human HT-12 Expression Bead Chips and verified by quantitative reverse-transcription polymerase chain reaction. Folate incorporation by PT cells versus normal thyroid cells was determined by incubation with (99m)Technetium ((99m)Tc)(CO)3-folate and (99m)Tc-Etarfolatide, and uptake was determined by gamma counting. Specific targeting of FRs was demonstrated by blocking with cold folate. A549 cells and Jurkat cells served as FR-negative controls, and KB cells and HeLa cells were FR-positive controls. RESULTS: On immunohistochemistry and Western blotting, human PT cells expressed FRs, whereas human thyroid cells did not. The FRα gene was expressed in all PTs analyzed, and the FRß gene was expressed by most. Uptake of (99m)Tc(CO)3-folate was increased in PT cells versus thyroid cells. There was dose-dependent uptake of (99m)Tc-etarfolatide, and uptake was inhibited by preincubation with cold folate, confirming FR-mediated binding. CONCLUSION: This is the first report of the expression and functionality of FRs on human PT cells. These findings suggest that (99m)Tc-folate holds potential for localization of PT tumors preoperatively and their treatment.


Subject(s)
Folate Receptor 1/genetics , Folate Receptor 1/metabolism , Folate Receptor 2/genetics , Folate Receptor 2/metabolism , Parathyroid Glands/metabolism , Animals , CHO Cells , Cell Line , Cricetulus , Folic Acid/analogs & derivatives , Folic Acid/metabolism , Gene Expression , HeLa Cells , Humans , Jurkat Cells , KB Cells , Organotechnetium Compounds/metabolism , Parathyroid Glands/diagnostic imaging , Parathyroid Neoplasms/diagnostic imaging , Parathyroid Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Radionuclide Imaging , Radiopharmaceuticals/metabolism , Thyroid Gland/diagnostic imaging , Thyroid Gland/metabolism , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/metabolism
4.
Cancer Epidemiol ; 34(4): 461-71, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20466611

ABSTRACT

BACKGROUND: The purpose of our study was to show the distinction between the apoptotic and anti-proliferative signaling of phytosterols and cholesterol-enrichment in prostate cancer cell lines, mediated by the differential transcription of caveolin-1, and N-myc downstream-regulated gene 1 (NDRG1), a pro-apoptotic androgen-regulated tumor suppressor. METHODS: PC-3 and DU145 cells were treated with sterols (cholesterol and phytosterols) for 72h, followed by trypan blue dye-exclusion measurement of necrosis and cell growth measured with a Coulter counter. Sterol induction of cell growth-suppressor gene expression was evaluated by mRNA transcription using RT-PCR, while cell cycle analysis was performed by FACS analysis. Altered expression of Ndrg1 protein was confirmed by Western blot analysis. Apoptosis was evaluated by real time RT-PCR amplification of P53, Bcl-2 gene and its related pro- and anti-apoptotic family members. RESULTS: Physiological doses (16microM) of cholesterol and phytosterols were not cytotoxic in these cells. Cholesterol-enrichment promoted cell growth (P<0.05), while phytosterols significantly induced growth-suppression (P<0.05) and apoptosis. Cell cycle analysis showed that contrary to cholesterol, phytosterols decreased mitotic subpopulations. We demonstrated for the first time that cholesterols concertedly attenuated the expression of caveolin-1 (cav-1) and NDRG1 genes in both prostate cancer cell lines. Phytosterols had the opposite effect by inducing overexpression of cav-1, a known mediator of androgen-dependent signals that presumably control cell growth or apoptosis. CONCLUSIONS: Cholesterol and phytosterol treatment differentially regulated the growth of prostate cancer cells and the expression of p53 and cav-1, a gene that regulates androgen-regulated signals. These sterols also differentially regulated cell cycle arrest, downstream pro-apoptotic androgen-regulated tumor suppressor, NDRG1 suggesting that cav-1 may mediate pro-apoptotic NDRG1 signals. Elucidation of the mechanism for sterol modulation of growth and apoptosis signaling may reveal potential targets for cancer prevention and/or chemotherapeutic intervention. Sterol regulation of NDRG1 transcription suggests its potential as biomarker for prediction of neoplasms that would be responsive to chemoprevention by phytosterols.


Subject(s)
Caveolin 1/genetics , Cell Cycle Proteins/genetics , Cholesterol/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Phytosterols/pharmacology , Prostatic Neoplasms/genetics , Apoptosis/drug effects , Blotting, Western , Caveolin 1/metabolism , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
5.
Cancer Detect Prev ; 32(4): 319-28, 2009.
Article in English | MEDLINE | ID: mdl-19186008

ABSTRACT

BACKGROUND: The purpose of our study was to show the apoptotic and anti-proliferative effects of phytosterols as distinct from cholesterol effects on prostate cancer cell lines, and also their differential expression of caveolin-1, and a prostate specific gene, PCGEM1. METHODS: PC-3 and DU145 cells were treated with sterols (cholesterol and phytosterols) for 48h, followed by trypan blue dye exclusion measurement of cytotoxicity and MTT cell proliferation assays, respectively. Cell cycle analysis was carried out microscopically, and by propidium iodide uptake using flow cytometry. Sterol induction of oncogenic gene expression was evaluated by RT-PCR. Apoptotic cells were identified by immunocytochemistry using DNA fragmentation method, and by annexin V adhesion using flow cytometry. RESULTS: Physiological doses (16microM) of these sterols were not cytotoxic in these cells. Cholesterol-enrichment promoted mitosis (54 and 61% by microscopy; 40.8 and 34.08% by FACS analysis in PC-3 and DU145, respectively) and cell growth (P<0.05), while phytosterols suppressed mitosis (29 and 35% by microscopy; 27.71 and 17.37% by FACS analysis in PC-3 and DU145, respectively), and significantly induced tumor-suppression (P<0.05) and apoptosis. We demonstrated for the first time that cholesterols upregulated the expression of PCGEM1 even in androgen-insensitive prostate cancer cell lines. Phytosterols reversed this effect, while upregulating the expression of caveolin-1, a known mediator of androgen-dependent proto-oncogene signals that presumably control growth and anti-apoptosis. CONCLUSIONS: Phytosterol inhibition of PCGEM1 and cell growth and the overexpression of caveolin-1, suggests that poor disease prognosis anchors on the ability of caveolin-1 to regulate downstream oncogene(s) and apoptosis genes. Sterol intake may contribute to the disparity in incidence of prostate cancer, and elucidation of the mechanism for modulation of growth and apoptosis signaling may reveal potential targets for cancer prevention and/or chemotherapeutic intervention. Sterol regulation of PCGEM1 expression suggests its potential as biomarker for prediction of neoplasms that would be responsive to chemoprevention by phytosterols.


Subject(s)
Cholesterol/pharmacology , Phytosterols/pharmacology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Untranslated/drug effects , Apoptosis/drug effects , Caveolin 1/drug effects , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Cycle/drug effects , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cholesterol/metabolism , Flow Cytometry , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Phytosterols/metabolism , Proto-Oncogene Mas , RNA, Long Noncoding , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...