Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 349
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Gynecol Oncol ; 162(1): 173-181, 2021 07.
Article in English | MEDLINE | ID: mdl-33972086

ABSTRACT

BACKGROUND: The patient-derived xenograft (PDX) model is a promising translational platform for duplicating the characteristics of primary tumors. Here, we established and characterized PDX models of uterine cancer to demonstrate their utility for preclinical drug testing. MATERIALS AND METHODS: We generated PDX tumors surgically derived from 58 cases of uterine cancer. Subrenal capsule xenografts and primary tumors were compared using microscopic examination, short tandem repeat analyses, and targeted sequencing analyses. A phosphatidylinositol 3-kinase (PI3K) inhibitor was administered to mice whose PDX tumors harbored a PTEN deletion or PIK3CA mutation. We also generated an orthotopic PDX model using uterine horn implantation. RESULTS: Thirty-three (56.9%) PDXs were successfully generated and passaged to maintain tumors. The histological features of the PDX tumors were stable over subsequent passages. By contrast, the proportions of epithelial and mesenchymal components of carcinosarcoma PDX models varied by generation. Targeted sequencing analyses revealed that all mutated cancer-related genes were stable during establishment and subgrafting. Treatment with a PI3K inhibitor cased a significant decrease in tumor weight in the clear cell carcinoma PDX harboring a frameshift PTEN deletion (p = 0.049) and in the serous carcinoma PDX harboring a missense PI3KCA mutation (p = 0.003) compared with matched controls. We also successfully established orthotopic PDX models (3/3; 100.0%). CONCLUSIONS: The histological and genetic features of PDXs were similar to those of primary tumors. This model is a promising translational platform for preclinical testing of new anticancer drugs and will enable the personalized development of therapeutic options for uterine cancer.


Subject(s)
Subrenal Capsule Assay/methods , Uterine Neoplasms/drug therapy , Uterine Neoplasms/pathology , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Graft Survival , Heterografts , Humans , Mice , Neoplasm Staging , Neoplasm Transplantation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Point Mutation , Transplantation, Heterologous , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism
2.
Blood ; 127(18): 2203-13, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26773040

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease defined by transcriptional classifications, specific signaling and survival pathways, and multiple low-frequency genetic alterations. Preclinical model systems that capture the genetic and functional heterogeneity of DLBCL are urgently needed. Here, we generated and characterized a panel of large B-cell lymphoma (LBCL) patient-derived xenograft (PDX) models, including 8 that reflect the immunophenotypic, transcriptional, genetic, and functional heterogeneity of primary DLBCL and 1 that is a plasmablastic lymphoma. All LBCL PDX models were subjected to whole-transcriptome sequencing to classify cell of origin and consensus clustering classification (CCC) subtypes. Mutations and chromosomal rearrangements were evaluated by whole-exome sequencing with an extended bait set. Six of the 8 DLBCL models were activated B-cell (ABC)-type tumors that exhibited ABC-associated mutations such as MYD88, CD79B, CARD11, and PIM1. The remaining 2 DLBCL models were germinal B-cell type, with characteristic alterations of GNA13, CREBBP, and EZH2, and chromosomal translocations involving IgH and either BCL2 or MYC Only 25% of the DLBCL PDX models harbored inactivating TP53 mutations, whereas 75% exhibited copy number alterations of TP53 or its upstream modifier, CDKN2A, consistent with the reported incidence and type of p53 pathway alterations in primary DLBCL. By CCC criteria, 6 of 8 DLBCL PDX models were B-cell receptor (BCR)-type tumors that exhibited selective surface immunoglobulin expression and sensitivity to entospletinib, a recently developed spleen tyrosine kinase inhibitor. In summary, we have established and characterized faithful PDX models of DLBCL and demonstrated their usefulness in functional analyses of proximal BCR pathway inhibition.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/genetics , Animals , Cell Lineage , Chromosome Aberrations , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Genetic Heterogeneity , Heterografts , Humans , Immunophenotyping , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Sequence Analysis, DNA , Subrenal Capsule Assay , Transcriptome
3.
Stem Cells ; 35(7): 1719-1732, 2017 07.
Article in English | MEDLINE | ID: mdl-28520232

ABSTRACT

Mesenchymal stem cells (MSCs) negatively modulate immune properties. Induced pluripotent stem cells (iPSCs)-derived MSCs are alternative source of MSCs. However, the effects of iPSC-MSCs on T cells phenotypes in vivo remain unclear. We established an iPSC-MSC-transplanted host versus graft reaction mouse model using subcapsular kidney injection. Th1, Th2, regulatory T cells (Treg), and Th17 phenotypes and their cytokines were investigated in vivo and in vitro. The role of caspases and the soluble factors involved in the effects of MSCs were examined. We found that iPSC-MSC grafts led to more cell survival and less infiltration of inflammatory cells in mice. iPSC-MSC transplantation inhibited T cell proliferation, decreased Th1 and Th2 phenotypes and cytokines, upregulated Th17 and Treg subsets. Moreover, iPSC-MSCs inhibited the cleavage of caspases 3 and 8 and inhibition of caspases downregulated Th1, Th2 responses and upregulated Th17, Treg responses. Soluble factors were determined using protein array and TGF-ß1/2/3, IL-10, and MCP-1 were found to be highly expressed in iPSC-MSCs. The administration of the soluble factors decreased Th1/2 response, upregulated Treg response and inhibited the cleavage of caspases. Our results demonstrate that iPSC-MSCs regulate T cell responses as a result of a combined action of the above soluble factors secreted by iPSC-MSCs. These factors suppress T cell responses by inhibiting the cleavage of caspases. These data provide a novel immunomodulatory mechanism for the underlying iPSC-MSC-based immunomodulatory effects on T cell responses. Stem Cells 2017;35:1719-1732.


Subject(s)
Caspases/immunology , Immunomodulation , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Caspases/genetics , Cell Differentiation , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Female , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred C57BL , Signal Transduction , Subrenal Capsule Assay , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transplantation, Heterologous
4.
Int J Cancer ; 140(6): 1356-1363, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27935045

ABSTRACT

Patient-derived cancer xenografts (PDX) are widely used to identify and evaluate novel therapeutic targets, and to test therapeutic approaches in preclinical mouse avatar trials. Despite their widespread use, potential caveats of PDX models remain considerably underappreciated. Here, we demonstrate that EBV-associated B-lymphoproliferations frequently develop following xenotransplantation of human colorectal and pancreatic carcinomas in highly immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ (NSG) mice (18/47 and 4/37 mice, respectively), and in derived cell cultures in vitro. Strikingly, even PDX with carcinoma histology can host scarce EBV-infected B-lymphocytes that can fully overgrow carcinoma cells during serial passaging in vitro and in vivo. As serial xenografting is crucial to expand primary tumor tissue for biobanks and cohorts for preclinical mouse avatar trials, the emerging dominance of B-lymphoproliferations in serial PDX represents a serious confounding factor in these models. Consequently, repeated phenotypic assessments of serial PDX are mandatory at each expansion step to verify "bona fide" carcinoma xenografts.


Subject(s)
B-Lymphocytes/transplantation , Carcinoma, Pancreatic Ductal/pathology , Colorectal Neoplasms/pathology , Epstein-Barr Virus Infections/pathology , Lymphoproliferative Disorders/etiology , Pancreatic Neoplasms/pathology , Subrenal Capsule Assay , Animals , Antigens, Neoplasm/analysis , B-Lymphocytes/pathology , B-Lymphocytes/virology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/virology , Cell Division , Colorectal Neoplasms/immunology , Colorectal Neoplasms/virology , Culture Media, Serum-Free , Epstein-Barr Virus Infections/immunology , Heterografts/immunology , Heterografts/pathology , Humans , Immunocompromised Host , Leukocyte Common Antigens/analysis , Lymphoproliferative Disorders/pathology , Lymphoproliferative Disorders/virology , Mice , Mice, Inbred NOD , Organ Specificity , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/virology , Spheroids, Cellular , Subrenal Capsule Assay/methods
5.
Pancreatology ; 15(4): 397-404, 2015.
Article in English | MEDLINE | ID: mdl-26026767

ABSTRACT

BACKGROUND: Although gemcitabine is commonly used as adjuvant therapy for pancreatic adenocarcinoma and pancreaticobiliary-type periampullary cancers, not all patients appear to benefit. This translational study evaluates the potential of a patient-derived subrenal capsule pancreatic cancer xenograft (SRCPCX) model to identify within eight weeks after surgery those tumours which will respond to gemcitabine. METHODS: SRCPCXs from 32 pancreatectomy patients were established in six to ten NOD/SCID mice per patient. After four weeks the mice were randomly assigned to receive gemcitabine or saline for four more weeks. After eight weeks, gemcitabine response in the grafts was evaluated by the percentage of tumour growth inhibition (%TGI), histological morphology and immunohistochemical markers (Ki-67, CK7 and cleaved caspase-3). These were collated into an Overall Response. Survival was assessed by Kaplan-Meier and Cox multivariate analyses. RESULTS: 375 of 450 pieces of tissue from 27 of 31 patients were evaluable. In 90% of patients, histopathological and immunostaining features of saline-treated control grafts were concordant with their original tumours. At follow up, six of 15 patients whose tumours had an Overall Response to gemcitabine died, compared with ten of 12 whose tumours did not respond (P = 0.025, Fisher's exact test). This was associated with improved survival on Kaplan-Meier analysis (P = 0.013). Cox multivariate analysis indicated that Overall Response, stage and grade were independent predictors of survival. CONCLUSION: This SRCPCX model retains major histopathological and immunohistochemical characteristics of the original tumour and when a combination of measures is used, enables early assessment of tumour sensitivity to gemcitabine in pancreatic cancers.


Subject(s)
Chemoradiotherapy, Adjuvant/methods , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/therapy , Subrenal Capsule Assay/methods , Adult , Aged , Aged, 80 and over , Animals , Antibiotics, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred NOD , Mice, SCID , Pancreatectomy , Pancreatic Neoplasms/surgery , Predictive Value of Tests , Survival Analysis , Treatment Outcome , Gemcitabine
6.
In Vivo ; 36(4): 1667-1675, 2022.
Article in English | MEDLINE | ID: mdl-35738627

ABSTRACT

BACKGROUND/AIM: Changes in the expression of neo-angiogenic molecules in the primary tumor and its metastases may significantly affect the efficacy of therapies. The aim of this study was to evaluate the alterations in aminopeptidase N (APN/CD13) and αvß3 integrin receptor expression in serially transplanted mesoblastic nephroma tumor (Ne/De) metastases using 68Gallium (68Ga)-labeled NOTA-cNGR and NODAGA-RGD radiotracers and preclinical positron emission tomography (PET) imaging. MATERIALS AND METHODS: Primary and metastatic mesoblastic nephroma (Ne/De) tumors were induced by subrenal capsule assay (SRCA) in Fischer-344 rats. In vivo PET imaging experiments were performed 8±1 days after the SRCA surgery using intravenously injected 68Ga-NOTA-c(NGR), 68Ga-NODAGA-RGD, and [18F]FDG radiotracers. RESULTS: Among the examined neo-angiogenic molecules, the expression of αvß3 integrin in the tumors was significantly lower than that of APN/CD13. This observation was confirmed by the PET data analysis, where a 2-6-fold higher APN/CD13-specific 68Ga-NOTA-cNGR accumulation was observed in both primary malignancies and metastases. However, a steadily increased accumulation of [18F]FDG, 68Ga-NODAGA-RGD, and 68Ga-NOTA-cNGR was observed in the tumors growing under the renal capsule and in the metastatic parathymic lymph nodes during serial transplantations. The observed increase in 68Ga- NOTA-cNGR accumulation during serial transplantations correlated well with the western blot analysis, where APN/CD13 protein levels were also elevated in the metastatic parathymic lymph nodes. CONCLUSION: The observed increase in glucose metabolism and the up-regulated expression of αvß3 integrin and APN/CD13 during serial transplantations of metastases may indicate enhanced malignancy.


Subject(s)
Kidney Neoplasms , Nephroma, Mesoblastic , Animals , Cell Line, Tumor , Fluorodeoxyglucose F18 , Gallium Radioisotopes/chemistry , Integrins , Kidney Neoplasms/diagnostic imaging , Oligopeptides/chemistry , Oligopeptides/metabolism , Positron-Emission Tomography/methods , Rats , Rats, Inbred F344 , Subrenal Capsule Assay
7.
Prostate ; 71(7): 675-81, 2011 May 15.
Article in English | MEDLINE | ID: mdl-20949523

ABSTRACT

BACKGROUND: Prostatic small cell carcinoma (SCC) is a rare variant of prostate cancer. It is extremely aggressive and resistant to available therapies with a median survival range of 5-17 months. No standard chemotherapeutic regimen has been established for its treatment. In search of a new therapeutic approach, we examined the response of patient-derived prostatic SCC tissue xenografts to irinotecan, a topoisomerase I inhibitor. METHODS: A tumor tissue line was established from a patient's prostatic SCC by subrenal capsule grafting using NOD-SCID mice. Mice carrying subcutaneous transplants of the tumor line were then treated for 2 weeks with irinotecan alone and in combination with cisplatin. The effect on tumor volume, histopathology, and apoptosis were determined. RESULTS: The prostatic SCC tissue line resembled the donor tissue in morphologic and immunohistochemical features. Irinotecan (20 mg/kg/day; days 1-3, 8-10) completely arrested xenograft growth with a small reduction in tumor volume and only minor weight loss of the hosts (7%); irinotecan (12 mg/kg; same schedule) + cisplatin (2.5 mg/kg/day; days 1 and 8) had a similar effect, but with lower weight loss. While the growth inhibition involved apoptosis, it was also associated with a marked increase in autophagy. CONCLUSIONS: Tumor tissue lines established via subrenal capsule xenografting provide models with clinical relevance and the present study suggests that irinotecan could be useful for therapy of refractory prostatic SCC, in particular in combination with cisplatin.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Carcinoma, Small Cell/drug therapy , Prostatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Autophagy/drug effects , Camptothecin/pharmacology , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Cell Survival , Cisplatin/pharmacology , Drug Therapy, Combination , Humans , Irinotecan , Male , Mice , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Subrenal Capsule Assay
8.
J Clin Invest ; 117(4): 997-1007, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17364025

ABSTRACT

We previously demonstrated that artificial lymph nodes (aLNs) could be generated in mice by the implantation of stromal cell-embedded biocompatible scaffolds into their renal subcapsular spaces. T and B cell domains that form in aLNs have immune response functions similar to those of follicles of normal lymphoid tissue. In the present study, we show that the aLNs were transplantable to normal as well as SCID mice, where they efficiently induced secondary immune responses. Antigen-specific secondary responses were strongly induced in aLNs even 4 weeks after their transplantation. The antigen-specific antibody responses in lymphocyte-deficient SCID mice receiving transplanted aLNs were substantial. The cells from the aLNs migrated to the SCID mouse spleen and BM, where they expanded to generate large numbers of antigen-specific antibody-forming cells. Secondary responses were maintained over time after immunization (i.e., antigen challenge), indicating that aLNs can support the development of memory B cells and long-lived plasma cells. Memory CD4(+) T cells were enriched in the aLNs and spleens of aLN-transplanted SCID mice. Our results indicate that aLNs support strong antigen-specific secondary antibody responses in immunodeficient mice and suggest the possibility of future clinical applications.


Subject(s)
Lymph Nodes/immunology , Lymph Nodes/transplantation , Animals , Antibody Formation , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Genes, Reporter , Green Fluorescent Proteins/genetics , Immunoglobulin G/analysis , Immunologic Memory , Mice , Mice, Inbred BALB C , Mice, SCID , Mice, Transgenic , Subrenal Capsule Assay , T-Lymphocytes/immunology
9.
Ophthalmic Res ; 43(2): 79-91, 2010.
Article in English | MEDLINE | ID: mdl-19829014

ABSTRACT

OBJECTIVE: It is theoretically possible to induce various cell types, including retinal neurons, from embryonic stem cells (ESCs). pax6 regulates early events in eye development, including the generation of retinal ganglion cells (RGCs). We previously reported the successful induction of corneal epithelial cells from ESCs transfected with the pax6 gene. Here, we attempted to establish cloned RGC-like cells from ESCs transfected with the pax6 gene. METHODS: Undifferentiated mouse ESCs were transfected with pax6 cDNA by electroporation, followed by selection with G418. We conducted limiting-dilution culture of pax6-transfected cells. We expanded the cloned pax6-transfected cells, which expressed nestin and musashi-1, for further characterization in culture media containing fibronectin. The cells were characterized using RT-PCR, immunostaining, electron microscopy, renal subcapsular transplantation assay and Ca imaging. RESULTS: We obtained clonally expanding pax6-transfected cells, all of which were positive for six3, sonic hedgehog (shh), math5, brn3, thy1 and melanopsin, by using several ESCs. When transplanted into a mouse renal capsule, they differentiated into neurons with elongated axons, expressing betaIII tubulin and neurofilament middle chain, and were free from teratoma development. Electron-microscopic examination showed neurotubules and neurofilaments in the axon-like processes of the cloned pax6-transfected cells. High KCl stimulation increased free Ca influx on Ca2+ imaging. CONCLUSIONS: ESCs were applicable for the induction of retinal progenitor cells, including RGC-like cells, by transfection with the pax6 gene and subsequent limiting-dilution culture. Cloned cell lines may be useful to analyze the requirements for retinal progenitor cell differentiation, and our study suggests the clinical application of this cell type.


Subject(s)
Embryonic Stem Cells/metabolism , Eye Proteins/genetics , Homeodomain Proteins/genetics , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Retinal Ganglion Cells/cytology , Retinal Neurons/cytology , Stem Cells/cytology , Transfection , Animals , Biomarkers/metabolism , Calcium/metabolism , Cell Differentiation , Cell Line , Cloning, Molecular , Electroporation , Female , Fibronectins/pharmacology , Intermediate Filament Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Nerve Tissue Proteins/metabolism , Nestin , PAX6 Transcription Factor , RNA-Binding Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Transplantation , Subrenal Capsule Assay
10.
Zhonghua Yi Xue Za Zhi ; 90(30): 2136-9, 2010 Aug 10.
Article in Zh | MEDLINE | ID: mdl-21029631

ABSTRACT

OBJECTIVE: To establish the murine xenograft model of human prostate cancer by grafting tumor tissues beneath the renal capsule of intact male athymic mouse. METHODS: Fifteen SCID mice were randomly divided into 3 groups (n = 5 each). Tissue recombinants were prepared in vitro with newborn BALB/c murine seminal vesicle mesenchyme (SVM) and surgical isolated human prostate cancer tissues by using recombination technique and then grafted beneath the renal capsule of intact male athymic mouse. At Week 4 after initial implantation, grafts were harvested and tumor sizes calculated. The expressions of human specific markers CK8/18 and vimentin were evaluated by immunohistochemistry to identify the human prostatic origin in grafts. P63 protein, a basal cell marker, was detected in prostate basal membrane to identify whether it was benign or malignant tissue. And the study control was prepared by implanting prostate cancer tissues alone under the renal capsule in SCID mouse. RESULTS: Of all 78 implantation cases in 15 mice, the tumor-forming rates were 100% (39/39) and 94.1% (37/39) respectively in the recombination and prostate cancer alone grafting groups. The recombination group was shown to be more efficient in terms of tumor size and weight in comparison with the prostate cancer alone group [(9.7 ± 3.1) vs (6.8 ± 2.0) mm(3), (12.1 ± 3.6) vs (8.2 ± 2.2) µg, P < 0.01]. There was no difference in serum PSA level between two groups. Grafts were confirmed as human prostate cancer tissues with the expressions of CK8/18 and vimentin. No expression of P63 was detected. CONCLUSION: The xenograft murine model of human prostate cancer is successfully established. It contains stroma components and is particularly suitable for studying the interaction of stroma and epithelia in the in vivo progression of prostate cancer.


Subject(s)
Disease Models, Animal , Prostatic Neoplasms , Subrenal Capsule Assay/methods , Animals , Humans , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Tumor Cells, Cultured
11.
Dev Biol ; 316(2): 302-11, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18325490

ABSTRACT

The mammalian prostate arises from the urogenital sinus and few factors have been identified to be important in the early stages of prostate development. In this study we show that the transcription factor Sox9 is expressed in the epithelia of all mouse prostatic lobes from the initial stages of their development. We used a conditional approach with mice expressing Cre recombinase under the control of Nkx3.1 regulatory sequences to delete Sox9 from the developing prostate. Mice with a prostate specific deletion of Sox9 showed a lack of ventral prostate development and abnormal anterior prostate differentiation. Analysis of these mutant animals revealed an early loss of expression of genes specific to the prostate epithelia such as Nkx3.1 and Shh and a marked reduction in proliferation in the ventral prostate but not in other lobes. Fgf signalling, through the MAPK pathway, has been shown to be important in prostate development and a lobe specific phenotype was reported for a prostate specific Fgfr2 mutant mouse model. Here we show that the levels of Fgfr2 and Sprouty2, a downstream target of Fgf signalling, were severely reduced in the ventral prostate of Sox9 mutant animals but not in other lobes. Prostate organ culture studies with a Mek inhibitor, U0126, and a Fgf receptor inhibitor, SU5402, indicate that the timing of expression of Cre in the mutant animals could account for the lobe specific phenotype in the Sox9 and Fgfr2 mutants. These studies imply that Sox9 is required for the early differentiation of the prostate bud epithelia.


Subject(s)
Gene Expression Regulation, Developmental , High Mobility Group Proteins/genetics , Prostate/embryology , Transcription Factors/genetics , Animals , Apoptosis , Cell Division , DNA Primers , Epithelial Cells/physiology , In Situ Hybridization , Male , Mice , Organ Culture Techniques , Prostate/cytology , Prostate/physiology , SOX9 Transcription Factor , Subrenal Capsule Assay
12.
Cells Tissues Organs ; 190(6): 336-46, 2009.
Article in English | MEDLINE | ID: mdl-19590164

ABSTRACT

BACKGROUND/AIMS: Craniosynostosis, the premature fusion of cranial sutures, is a common congenital defect. In vivo models for studying cranial suture biology impose inherent restrictions on tissue accessibility and manipulation. The present study was performed to investigate the utility of the renal capsule assay in overcoming these limitations and providing a reproducible model system for studying cranial suture morphogenesis and fate. MATERIALS AND METHODS: The posterior frontal suture, which fuses physiologically, and the coronal and sagittal sutures, which remain patent, were dissected from postnatal and embryonic mouse calvaria and placed under the renal capsule of syngeneic recipient mice (n = 72 in total). Sutures were harvested from 1-14 days after transplantation for histological and morphometric analysis. Suture transplants were compared with nonmanipulated sutures at equivalent developmental stages. The derivation of cells associated with the growing transplants was analyzed using beta-actin-GFP (green fluorescent protein) transgenic mice. RESULTS: Sutures transplanted under the renal capsule maintained normal suture morphology and fate with the posterior frontal suture fusing and the coronal and sagittal sutures remaining patent. In posterior frontal suture transplants, the fusion process mimicked in vivo suture fusion with a delay of 1-2 days. In comparison to in vivo suture complexes, transplant thickness and trabeculation were significantly increased. In addition, we found that osteoblasts within the growing transplant were derived from the transplant itself rather than the host. CONCLUSION: The renal capsule supports the growth of cranial sutures. In this system transplanted sutures recapitulate the anatomical development and fate (fusion or patency) of cranial sutures in vivo. This model system will facilitate controlled ex vivo manipulations of both embryonic and postnatal sutures.


Subject(s)
Cranial Sutures/growth & development , Morphogenesis , Subrenal Capsule Assay/methods , Animals , Cranial Sutures/cytology , Cranial Sutures/transplantation , Craniosynostoses , Disease Models, Animal , In Vitro Techniques , Mice , Osteoblasts/cytology , Osteoblasts/physiology
13.
Anticancer Res ; 29(6): 2121-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19528472

ABSTRACT

BACKGROUND: The ultimate cause of cancer death is, in most cases, the appearance of metastases. The aim of the present study was to contribute to animal experimental investigations of metastatic tumor development. MATERIALS AND METHODS: Rat hepatocarcinoma (He/De), mesoblastic nephroma (Ne/De) cells, and in other cases tumor-bearing lymph nodes were transplanted under the renal capsule of F344 rats. Metastatic potential of tumor cells was examined by whole body autoradiography and phosphor image analysis. The organ distribution of cells was also investigated. RESULTS: Transplanted tumor cells resulted in metastases in the parathymic lymph nodes. Implanted India ink also demonstrated connection between the lymphatic vessels of the renal capsule and the parathymic lymph nodes. The metastatic potential was independent of the primary tumor growth rate. CONCLUSION: The renal capsule-parathymic lymph node complex seems to be suitable for the isolated in vivo examination of metastatic development and for the detailed analysis of secondary tumors.


Subject(s)
Carcinoma, Renal Cell/secondary , Disease Models, Animal , Kidney Neoplasms/secondary , Liver Neoplasms/pathology , Lymph Nodes/pathology , Thymus Gland/pathology , Wilms Tumor/secondary , Animals , Carcinoma, Renal Cell/pathology , Female , Kidney Neoplasms/pathology , Lymphatic Metastasis , Male , Rats , Rats, Inbred F344 , Subrenal Capsule Assay , Tumor Cells, Cultured , Wilms Tumor/pathology
14.
Zhonghua Wai Ke Za Zhi ; 47(16): 1249-52, 2009 Aug 15.
Article in Zh | MEDLINE | ID: mdl-19781175

ABSTRACT

OBJECTIVE: To study the protective effect of islet xenograft and its possible mechanism of high expression of heme oxygenase-1 (HO-1) in donor pancreas islet induced by cobalt protoporphyrin (CoPP). METHODS: Male SD rats and C57BL/6 mouse were used as donors and recipients respectively. Donors were divided into 3 groups according to different pretreatment 24 hours before donation: control group (injected intraperitoneally with NaCl), induce group [injected intraperitoneally with cobalt-protoporphyrin (CoPP)], block group (injected intraperitoneally with CoPP and zinc protoporphyrin simultaneously). A modified approach was used for islet isolation.Recipients were rendered diabetic by intraperitoneal injection of streptozotocin. Islets were transplanted into mouse subrenal capsule. Postoperative mouse glycemia were monitored daily and normoglycemia time was compared among each group. The receptor mouse serum IL-10 was detected by ELISA approach, and real-time PCR was used to check the expression of IL-10 mRNA in islet graft tissues. The graft tissues were observed for the lymphocyte infiltration after HE staining. RESULTS: Diabetes mice accepted islets untreated, induced or blocked maintained the euglycemia for (9.3 +/- 1.4), (16.3 +/- 1.5) and (9.7 +/- 1.0) d respectively. The xeno-islets presented HO-1 over-expression survived much longer than that absent (P < 0.05), it was no significance between control group and block group (P > 0.05). The mouse islet serum IL-10 content after induction was (73.0 +/- 9.7) pg/ml, significantly higher than (30.6 +/- 3.9) pg/ml of the untreated group and (32.1 +/- 5.9) pg/ml of the blocked group (P < 0.05), there was no difference between control group and block group (P > 0.05). Moreover, the IL-10 mRNA expression up-regulated statistic significantly in HO-1 induced islet xeno-graft. Pathological examination showed that the graft lymphocyte infiltration of the induced group was obviously less serious than the other two groups. CONCLUSIONS: The higher expression of HO-1 induced by CoPP in vivo would significantly prolong graft survival time and its mechanism could be related to immune modulation of IL-10.


Subject(s)
Heme Oxygenase-1/metabolism , Pancreas Transplantation , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/surgery , Graft Survival , Heme Oxygenase-1/drug effects , Interleukin-10/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Protoporphyrins/pharmacology , Rats , Rats, Sprague-Dawley , Subrenal Capsule Assay , Transplantation, Heterologous
15.
Methods Mol Biol ; 1922: 49-55, 2019.
Article in English | MEDLINE | ID: mdl-30838564

ABSTRACT

Tissue interactions are crucial during the development of organs. Among the most studied tissue interactions are those that take place between the epithelial cells and the underlying mesenchymal cells, known as epithelial-mesenchymal interactions. Tissue recombination assay is a valuable model to study the mechanisms involved in the regulation of such interactions. Here, we describe how to dissociate and recombine the epithelial and mesenchymal components of the embryonic tooth. In addition, we explain how to transplant the recombined tissues under the kidney capsule of immunocompromised mice in order to allow their further development into a mature tooth.


Subject(s)
Embryonic Development , Epithelial Cells/physiology , Mesenchymal Stem Cells/physiology , Subrenal Capsule Assay/methods , Tooth/cytology , Animals , Embryo, Mammalian , Mice , Tooth/embryology
16.
Transplantation ; 85(4): 640-4, 2008 Feb 27.
Article in English | MEDLINE | ID: mdl-18347545

ABSTRACT

Xenothymus transplantation under the kidney capsule in athymic rodents frequently leads to multiorgan autoimmunity. Herein, we explore whether this is an intrinsic risk of xenothymus grafting or whether it depends on the transplant technique. We developed a new technique of "venous pouch" thymus grafting (heart-xenothymus) and compared this with the conventional kidney subcapsular technique (kidney-xenothymus) in a rat-into-nude-mouse model. Whereas lethal autoimmunity developed in 90% of kidney-xenothymus recipients, all heart-xenothymus grafted mice remained completely healthy. Autoimmunity in heart-xenothymus recipients was absent despite a significantly improved T-cell generation and was associated with significantly higher CD4+CD25+ T-cell frequencies and CD4+CD25+ cell Foxp3 mRNA levels than those observed in kidney-xenothymus recipients. In conclusion, we describe a novel vascular pouch technique of xenothymus transplantation that prevents the development of autoimmunity in nude mice. Our data further suggest that prevention of autoimmunity is related to a superior development of regulatory T-cells.


Subject(s)
Autoimmunity , Lymphocyte Depletion , T-Lymphocytes/immunology , Thymus Gland/transplantation , Transplantation, Heterologous/immunology , Animals , Autoimmune Diseases/immunology , Mice , Rats , Subrenal Capsule Assay/methods , Transplantation, Heterologous/adverse effects
17.
Transplant Proc ; 40(2): 455-6, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374099

ABSTRACT

Immunomodulatory properties have been recognized for human alpha-1 antitrypsin (hAAT). However, production of anti-hAAT antibodies in mice may inactivate the protein. In this study, we evaluated the effects of chronic hAAT administration on allogeneic islet graft survival. Chemically diabetic mice lacking an efficient humoral response due to the targeted disruption of the Ig mu-chain (muMT mice) or wild-type (WT) C57BL/6 mice received DBA/2 mouse islets under the kidney capsule. hAAT (Prolastin or Aralast) was given intraperitoneally on day 0 and every 3 days thereafter. Control animals received no treatment. hAAT administration in WT mice resulted in prolongation of islet allograft survival in a dose-dependent fashion in both hAAT-treated groups. Lack of Ig response (muMT mice) per se conferred a beneficial effect on graft survival that worsened in the Prolastin-treated groups but improved in the Aralast-treated group. Our data indicate that systemic administration of hAAT results in prolongation of islet allograft survival. Absence of mature B cells and Ig mu-chain resulted in improved graft survival, pointing to a role for B cells in the rejection process in this model. Treatment with Prolastin worsened graft survival in muMT mice, whereas Aralast did improve it, suggesting a different efficacy and possible actions of the two drug formulations.


Subject(s)
Antibody Formation/physiology , Diabetes Mellitus, Experimental/surgery , Graft Survival/drug effects , Islets of Langerhans Transplantation/physiology , alpha 1-Antitrypsin/therapeutic use , Animals , Antibody Formation/drug effects , Graft Survival/immunology , Islets of Langerhans Transplantation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Subrenal Capsule Assay , Transplantation, Homologous
18.
Transplant Proc ; 40(2): 529-32, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18374121

ABSTRACT

In a previous study, we observed that combination therapy of nonobese diabetic (NOD) mice with epidermal growth factor (EGF) and gastrin partially restored pancreatic islet beta-cell mass and reversed hyperglycemia without the use of immunotherapy. Herein we have studied the effects of EGF plus gastrin on recurrent autoimmune responses in diabetic NOD mice transplanted with syngeneic islets. EGF (10 microg/kg) plus gastrin (30 microg/kg) given intraperitoneally (i.p.) once daily to diabetic NOD mice (blood glucose, 23 +/- 2 mmol/L) significantly prolonged the median survival time of NOD islet grafts to 60 days (n = 10 mice) measured as the days until hyperglycemia recurrence (blood glucose > or =12 mmol/L; versus EGF alone to 36 days (n = 10), or gastrin alone, 19 days (n = 10), or vehicle, 11 days (n = 9). At 7-14 days after transplantation insulin-stained beta-cells were much more numerous in islet grafts of EGF plus gastrin-treated mice (13.0 +/- 0.9 x 10(5) cells) versus grafts in vehicle-treated mice (1.0 +/- 0.3 x 10(5) cells). CD45+ leukocytes were significantly reduced in number and surrounded but did not destroy the beta cells in the islets of EGF plus gastrin-treated mice (29 +/- 2 x 10(5) cells) versus those in vehicle-treated mice (57 +/- 3 x 10(5) cells). We concluded that the EGF plus gastrin combination therapy inhibited the recurrent autoimmune response and delayed rejection of syngeneic islet grafts, suggesting a therapeutic role for these peptides in islet transplantation.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Epidermal Growth Factor/therapeutic use , Gastrins/therapeutic use , Islets of Langerhans Transplantation , Mice, Inbred NOD , Animals , Combined Modality Therapy , Drug Therapy, Combination , Female , Immunohistochemistry , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation/pathology , Leukocyte Count , Mice , Subrenal Capsule Assay , Transplantation, Isogeneic
19.
Transplant Proc ; 40(5): 1650-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18589167

ABSTRACT

Current pancreatic islet transplantation protocols achieve remarkable short-term success, but long-term insulin independence remains elusive. Hypoxic and inflammatory insults cause substantial early posttransplant graft loss while allo/autoimmunity and immunosuppressive drug toxicity threaten long-term graft mass and function. Exendin-4 (Ex4) is a GLP-1 receptor agonist that promotes beta-cell proliferation, survival, and differentiation. To determine whether Ex-4 displays potential as a graft-supportive agent, we transplanted 500 murine islets under the kidney capsule of syngeneic or allogeneic streptozocin-treated recipient mice and immediately initiated daily treatment with vehicle or Ex4. Graft beta-cell proliferation, death, and vascularity were assessed at 1, 3, and 10 days after syngeneic islet transplantation. For allogeneic recipients, blood glucose and body weight were assessed until glycemic deterioration. Ex-4 did not promote graft beta-cell proliferation, reduce beta-cell death, or enhance graft vascularity over the first 10 days after syngeneic islet transplantation. A trend toward prolongation of posttransplant euglycemia was observed with Ex4 treatment in nonimmune-suppressed allograft recipients, but its use in this setting was associated with frequent, severe hypoglycemia over the first 2 posttransplant days. Our findings do not support a beneficial effect of Ex-4 on islet grafts during the critical early posttransplant period, further, they demonstrate a significant hypoglycemic potential of Ex-4 in the first days after islet transplantation in mice. Optimal application of GLP-1 receptor agonists for long-term proliferative and survival benefits in transplantation may require earlier intervention prior to and/or during islet isolation for peri-transplant cytoprotection and administration beyond the period of engraftment.


Subject(s)
Cell Survival/drug effects , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/cytology , Islets of Langerhans Transplantation/physiology , Peptides/pharmacology , Venoms/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/surgery , Exenatide , Hyperglycemia/physiopathology , Insulin-Secreting Cells/drug effects , Islets of Langerhans Transplantation/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Subrenal Capsule Assay , Transplantation, Homologous , Transplantation, Isogeneic
20.
Diabetes ; 55(11): 2931-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17065328

ABSTRACT

We have developed a magnetic resonance imaging (MRI) technique for imaging Feridex (superparamagnetic iron oxide [SPIO])-labeled islets of Langerhans using a standard clinical 1.5-Tesla (T) scanner and employing steady-state acquisition imaging sequence (3DFIESTA). Both porcine and rat islets were labeled with SPIO by a transfection technique using a combination of poly-l-lysine and electroporation. Electron microscopy demonstrated presence of SPIO particles within the individual islet cells, including beta-cells and particles trapped between cell membranes. Our labeling method produced a transfection rate of 860 pg to 3.4 ng iron per islet, dependent on the size of the islet. The labeling procedure did not disrupt either the function or viability of the islets. In vitro 3DFIESTA magnetic resonance images of single-labeled islets corresponded with their optical images. In vivo T2*-weighted scan using 1.5 T detected as few as 200 SPIO-labeled islets transplanted under rat kidney capsule, which correlated with immunohistochemistry of the transplant for insulin and iron. Ex vivo 3DFIESTA images of kidneys containing 200, 800 or 2,000 SPIO-labeled islet isografts showed good correlation between signal loss and increasing numbers of islets. These data provide evidence that islets can be labeled with SPIO and imaged using clinically available 1.5- T MRI.


Subject(s)
Islets of Langerhans Transplantation/physiology , Islets of Langerhans/cytology , Magnetic Resonance Imaging/methods , Animals , Animals, Newborn , Cell Survival , Electroporation , Iron-Dextran Complex , Islets of Langerhans/ultrastructure , Male , Models, Animal , Rats , Rats, Inbred Lew , Sensitivity and Specificity , Subrenal Capsule Assay , Swine , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL