Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetes Metab J ; 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38772544

RESUMO

Background: Islet transplantation holds promise for treating selected type 1 diabetes mellitus patients, yet the scarcity of human donor organs impedes widespread adoption. Porcine islets, deemed a viable alternative, recently demonstrated successful longterm survival without zoonotic risks in a clinically relevant pig-to-non-human primate islet transplantation model. This success prompted the development of a clinical trial protocol for porcine islet xenotransplantation in humans. Methods: A single-center, open-label clinical trial initiated by the sponsor will assess the safety and efficacy of porcine islet transplantation for diabetes patients at Gachon Hospital. The protocol received approval from the Gachon Hospital Institutional Review Board (IRB) and the Korean Ministry of Food and Drug Safety (MFDS) under the Investigational New Drug (IND) process. Two diabetic patients, experiencing inadequate glycemic control despite intensive insulin treatment and frequent hypoglycemic unawareness, will be enrolled. Participants and their family members will engage in deliberation before xenotransplantation during the screening period. Each patient will receive islets isolated from designated pathogen-free pigs. Immunosuppressants and systemic infection prophylaxis will follow the program schedule. The primary endpoint is to confirm the safety of porcine islets in patients, and the secondary endpoint is to assess whether porcine islets can reduce insulin dose and the frequency of hypoglycemic unawareness. Conclusion: A clinical trial protocol adhering to global consensus guidelines for porcine islet xenotransplantation is presented, facilitating streamlined implementation of comparable human trials worldwide.

2.
Stem Cells Int ; 2021: 8873383, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093711

RESUMO

Although human induced pluripotent stem cells (iPSCs) can serve as a universal cell source for regenerative medicine, the use of iPSCs in clinical applications is limited by prohibitive costs and prolonged generation time. Moreover, allogeneic iPSC transplantation requires preclusion of mismatches between the donor and recipient human leukocyte antigen (HLA). We, therefore, generated universally compatible immune nonresponsive human iPSCs by gene editing. Transcription activator-like effector nucleases (TALENs) were designed for selective elimination of HLA DR expression. The engineered nucleases completely disrupted the expression of HLA DR on human dermal fibroblast cells (HDF) that did not express HLA DR even after stimulation with IFN-γ. Teratomas formed by HLA DR knockout iPSCs did not express HLA DR, and dendritic cells differentiated from HLA DR knockout iPSCs reduced CD4+ T cell activation. These engineered iPSCs might provide a novel translational approach to treat multiple recipients from a limited number of cell donors.

3.
Sci Rep ; 11(1): 8617, 2021 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-33883656

RESUMO

Many groups are working to improve the results of clinical allogeneic islet transplantation in a primate model. However, few studies have focused on the optimal islet dose for achieving normal glycemia without exogenous insulin after transplantation in primate models or on the relationship between rejection and islet amyloid polypeptide (IAPP) expression. We evaluated the dose (10,000, 20,000, and > 25,000 islet equivalents (IEQ)/kg) needed to achieve normal glycemia without exogenous insulin after transplantation using eleven cynomolgus monkeys, and we analyzed the characteristics exhibited in the islets after transplantation. 10,000 IEQ/kg (N = 2) failed to control blood glucose level, despite injection with the highest dose of exogenous insulin, and 20,000 IEQ/kg group (N = 5) achieved unstable control, with a high insulin requirement. However, 25,000 IEQ/kg (N = 4) achieved normal glycemia without exogenous insulin and maintained it for more than 60 days. Immunohistochemistry results from staining islets found in liver biopsies indicated that as the number of transplanted islets decreased, the amount of IAPP accumulation within the islets increased, which accelerated CD3+ T cell infiltration. In conclusion, the optimal transplantation dose for achieving a normal glycemia without exogenous insulin in our cynomolgus monkey model was > 25,000 IEQ/kg, and the accumulation of IAPP early after transplantation, which depends on the transplanted islet dose, can be considered one factor in rejection.


Assuntos
Diabetes Mellitus Experimental/imunologia , Insulina/imunologia , Ilhotas Pancreáticas/imunologia , Macaca fascicularis/imunologia , Animais , Complexo CD3/imunologia , Teste de Tolerância a Glucose/métodos , Imuno-Histoquímica/métodos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Transplante Heterólogo/métodos
4.
Tissue Eng Regen Med ; 18(2): 305-313, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33591557

RESUMO

BACKGROUND: Since primates have more biological similarities to humans than do other animals, they are a valuable resource in various field of research, including biomedicine, regenerative medicine, and drug discovery. However, there remain limitations to maintenance and expansion of primary hepatocytes derived from nonhuman primates. To overcome these limitations, we developed a novel culture system for primate cells. METHODS: Primary hepatocytes from Macaca fascicularis (mf-PHs) were isolated from hepatectomized liver. To generate chemically derived hepatic progenitor cells (mf-CdHs), mf-PHs were cultured with reprogramming medium containing A83-01, CHIR99021, and hepatocyte growth factor (HGF). The bi-potent differentiation capacity of mf-CdHs into hepatocytes and biliary epithelial cells was confirmed by treatment with hepatic differentiation medium (HDM) and cholangiocytic differentiation medium (CDM), respectively. RESULTS: mf-PHs cultured with reprogramming medium showed rapid proliferation capacity in vitro and expressed progenitor-specific markers. Moreover, when cultured in HDM, these progenitor cells stably differentiated into hepatocyte-like cells expressing the mature hepatic markers. On the other hand, when cultured in CDM, the differentiated biliary epithelial cells expressed mature cholangiocyte characteristics. CONCLUSION: The results of the present study demonstrate that we successfully induced the formation of hepatic progenitor cells from mf-PHs by culturing them with a combination of small molecules, including growth factors. These results offer a means of expanding nonhuman primate hepatocytes without genetic manipulation for cellular resource, preclinical applications and regenerative medicine for the liver.


Assuntos
Hepatócitos , Regeneração Hepática , Células-Tronco , Animais , Diferenciação Celular , Células Cultivadas , Macaca fascicularis
5.
Stem Cell Rev Rep ; 17(3): 1053-1067, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33423156

RESUMO

Human embryonic stem cells (hESCs) hold promise in regenerative medicine but allogeneic immune rejections caused by highly polymorphic human leukocyte antigens (HLAs) remain a barrier to their clinical applications. Here, we used a CRISPR/Cas9-mediated HLA-editing strategy to generate a variety of HLA homozygous-like hESC lines from pre-established hESC lines. We edited four pre-established HLA-heterozygous hESC lines and created a mini library of 14 HLA-edited hESC lines in which single HLA-A and HLA-B alleles and both HLA-DR alleles are disrupted. The HLA-edited hESC derivatives elicited both low T cell- and low NK cell-mediated immune responses. Our library would cover about 40% of the Asian-Pacific population. We estimate that HLA-editing of only 19 pre-established hESC lines would give rise to 46 different hESC lines to cover 90% of the Asian-Pacific population. This study offers an opportunity to generate an off-the-shelf HLA-compatible hESC bank, available for immune-compatible cell transplantation, without embryo destruction. Graphical Abstract.


Assuntos
Edição de Genes , Células-Tronco Embrionárias Humanas , Embrião de Mamíferos , Transplante de Células-Tronco Hematopoéticas , Humanos , Medicina Regenerativa
6.
Curr Eye Res ; 45(11): 1352-1358, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32223337

RESUMO

Purpose: This study aimed to determine the effect of thymosin beta 4 (Tß4) on human corneal epithelial cell migration and the downstream signaling pathways. Tß4 has a role in tissue development, cell migration, inflammation, and wound healing. A previous study showed that Tß4 directly binds to F0-F1 ATP synthase. Other studies reported the role of extracellular ATP and purinergic receptors in cell migration with several cell types. Despite advancing to the clinical stage for treatment of eye disorders, the effect of Tß4 on human corneal epithelial cell (HCEC) migration and proliferation and the precise downstream signaling pathway(s) have not been identified. Methods: Various concentrations of Tß4 were tested in vitro on human corneal epithelial cell proliferation using the CCK-8 Kit and on cell migration using the gap closure migration assay. Additionally, ATP levels at various time points were determined using the ATP Lite One-Step Kit. The Fluo 8 NO Wash Calcium Assay Kit was used to measure the intracellular Ca2+ concentration after treatment with various concentrations of Tß4. P2X7 inhibitors were tested on ATP signaling and migration. Total- and phospho-ERK1/2 levels were determined in western blot. Results: Tß4 enhanced HCEC proliferation and migration in a dose- and time-dependent manner. Moreover, these functions were related to increased extracellular ATP levels, intracellular Ca2+ influx, and ERK1/2 phosphorylation. Tß4-mediated HCEC migration was inhibited by specific P2X7 purinergic receptor antagonists suggesting the role of this receptor in Tß4-mediated human corneal epithelial cell migration. Conclusions: These results suggest that Tß4-mediated HCEC proliferation and migration are associated with increased ATP levels, P2X7 R-mediated Ca2+ influx, and the ERK1/2 signaling pathway. This study begins to describe the mechanisms for Tß4-mediated corneal healing and regeneration.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Epitélio Corneano/efeitos dos fármacos , Receptores Purinérgicos P2X7/metabolismo , Transdução de Sinais/fisiologia , Timosina/farmacologia , Trifosfato de Adenosina/metabolismo , Western Blotting , Cálcio/metabolismo , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Epitélio Corneano/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Sistema de Sinalização das MAP Quinases/fisiologia
7.
Sci Rep ; 10(1): 793, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31964980

RESUMO

The most obvious method to observe transplanted islets in the liver is direct biopsy, but the distribution and location of the best biopsy site in the recipient's liver are poorly understood. Islets transplanted into the whole liver of five diabetic cynomolgus monkeys that underwent insulin-independent survival for an extended period of time after allo-islet transplantation were analyzed for characteristics and distribution tendency. The liver was divided into segments (S1-S8), and immunohistochemistry analysis was performed to estimate the diameter, beta cell area, and islet location. Islets were more distributed in S2 depending on tissue size; however, the number of islets per tissue size was high in S1 and S8. Statistical analysis revealed that the characteristics of islets in S1 and S8 were relatively similar to other segments despite various transplanted islet dosages and survival times. In conclusion, S1, which exhibited high islet density and reflected the overall characteristics of transplanted islets, can be considered to be a reasonable candidate for a liver biopsy site in this monkey model. The findings obtained from the five monkey livers with similar anatomical features to human liver can be used as a reference for monitoring transplanted islets after clinical islet transplantation.


Assuntos
Transplante das Ilhotas Pancreáticas/métodos , Ilhotas Pancreáticas , Fígado/citologia , Aloenxertos , Animais , Biópsia , Diabetes Mellitus Experimental/patologia , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Insulina/metabolismo , Macaca fascicularis , Masculino
8.
J Invest Dermatol ; 139(3): 692-701, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30393080

RESUMO

Much of our understanding of human biology and the function of mammalian cells in tissue regeneration have been derived from mechanistically and genetically manipulated rodent models. However, current models examining epidermal wound repair fail to address both the cross-species mechanistic and immunogenic differences simultaneously. Herein, we describe a multifaceted approach intended to better recapitulate human skin recovery in rodent models. First, immunodeficient NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ mice were intravenously inoculated with human hematopoietic stem cells to become, in essence, humanized, and capable of initiating an adaptive immune response. Next, a chimney-shaped mechanical device was implanted onto the excisional wound site to prevent healing by primary intention (contraction) and expedite cell transplantation. Subsequently, cell therapy was administered by transplanting cord blood-derived endothelial progenitor cells or human pluripotent stem cell-derived endothelial cells into the wound site to examine the regeneration process at a histological level. This study demonstrates human cutaneous repair in a murine model by addressing both the mechanistic and immunogenic differences in the epidermis. We further show human leukocyte recruitment in damaged tissue and improved healing by secondary intention in the transplanted groups, highlighting the need for useful preclinical animal models to better understand leukocyte function in human (tissue repair and) regeneration.


Assuntos
Imunidade Adaptativa/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Pele/lesões , Cicatrização/fisiologia , Ferimentos e Lesões/terapia , Animais , Biópsia por Agulha , Modelos Animais de Doenças , Células Endoteliais/transplante , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Distribuição Aleatória , Regeneração/fisiologia , Ferimentos e Lesões/imunologia
9.
Biochem Biophys Res Commun ; 504(1): 302-308, 2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30190122

RESUMO

Mesenchymal stromal cells (MSCs) isolated from numerous tissues including human fetal tissue are currently used in cell therapy and regenerative medicine. Among fetal tissues, the umbilical cord (UC) is one of the sources for both MSCs and endothelial cells (ECs). To establish ectopic vascularized bone tissue formation, UC-derived MSCs and ECs were isolated. UC-MSCs expressing human BMP-2 (hBMP-2-MSCs) were generated using an adenoviral system to promote bone formation. These cells were then transplanted with Matrigel into the subcutaneous tissue of an immune deficient NSG mouse, and bone tissue was analyzed after several weeks. The osteogenic differentiation ability of MSCs was elevated by transduction of the hBMP-2 expressing adenoviral system, and vascularization of bone tissue was enhanced by human umbilical vein endothelial cells (HUVEC). In this study, our results provide evidence that MSCs and HUVECs from human umbilical cord are suitable cells to investigate bone tissue engineering. The results also suggest that the co-transplantation of hBMP2-MSCs and HUVECs may be a simple and efficient strategy for improving tissue generation and angiogenesis in bone tissue engineering using stem cells.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Células-Tronco Mesenquimais/citologia , Osteogênese , Engenharia Tecidual/métodos , Cordão Umbilical/citologia , Animais , Regeneração Óssea , Diferenciação Celular , Transplante de Células , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Neovascularização Fisiológica
10.
Tissue Eng Part A ; 24(7-8): 576-583, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28726547

RESUMO

Three-dimensional (3D) bioprinting technology is a promising new technology in the field of bioartificial organ generation with regard to overcoming the limitations of organ supply. The cell source for bioprinting is very important. Here, we generated 3D hepatic scaffold with mouse-induced hepatocyte-like cells (miHeps), and investigated whether their function was improved after transplantation in vivo. To generate miHeps, mouse embryonic fibroblasts (MEFs) were transformed with pMX retroviruses individually expressing hepatic transcription factors Hnf4a and Foxa3. After 8-10 days, MEFs formed rapidly growing hepatocyte-like colonies. For 3D bioprinting, miHeps were mixed with a 3% alginate hydrogel and extruded by nozzle pressure. After 7 days, they were transplanted into the omentum of Jo2-treated NOD Scid gamma (NSG) mice as a liver damage model. Real-time polymerase chain reaction and immunofluorescence analyses were conducted to evaluate hepatic function. The 3D bioprinted hepatic scaffold (25 × 25 mm) expressed Albumin, and ASGR1 and HNF4a expression gradually increased for 28 days in vitro. When transplanted in vivo, the cells in the hepatic scaffold grew more and exhibited higher Albumin expression than in vitro scaffold. Therefore, combining 3D bioprinting with direct conversion technology appears to be an effective option for liver therapy.


Assuntos
Bioimpressão/métodos , Hepatócitos/metabolismo , Animais , Receptor de Asialoglicoproteína/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Camundongos , Impressão Tridimensional , Reação em Cadeia da Polimerase em Tempo Real , Engenharia Tecidual/métodos
11.
Xenotransplantation ; 25(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29135052

RESUMO

BACKGROUND: Porcine islet xenotransplantation is considered an attractive alternative treatment for type 1 diabetes mellitus. However, it is largely limited because of initial rejection due to Instant Blood-Mediated Inflammatory Reaction (IBMIR), oxidative stress, and inflammatory responses. Recently, soluble tumor necrosis factor-ɑ receptor type I (sTNF-αR) and heme oxygenase (HO)-1 genes (HO-1/sTNF-αR) have been shown to improve the viability and functionality of porcine islets after transplantation. METHODS: In this study, genetically modified mesenchymal stem cells (MSCs) expressing the HO-1/sTNF-αR genes (HO-1/sTNF-αR-MSC) were developed using an adenoviral system, and porcine islet viability and function were confirmed by in vitro tests such as GSIS, AO/PI, and the ADP/ATP ratio after coculturing with HO-1/sTNF-αR-MSCs. Subsequently, isolated porcine islets were transplanted underneath the kidney capsule of diabetic humanized mice without MSCs, with MSCs or with HO-1/sTNF-αR-MSCs. RESULTS: According to the results, the HO-1/sTNF-αR-MSC-treated group exhibited improved survival of porcine islets and could reverse hyperglycemia more than porcine islets not treated with MSCs or islets cotransplanted with MSCs. Moreover, the HO-1/sTNF-αR-MSC group maintained its morphological characteristics and the insulin secretion pattern of transplanted porcine islets similar to endogenous islets in immunocompetent humanized mice. CONCLUSIONS: Our results suggest that HO-1/sTNF-αR-MSCs are efficient tools for porcine islet xenotransplantation, and this study may provide basic information for pre-clinical animal models and future clinical trials of porcine islet xenotransplantation.


Assuntos
Sobrevivência de Enxerto , Heme Oxigenase-1/genética , Xenoenxertos/imunologia , Proteínas de Membrana/genética , Células-Tronco Mesenquimais/citologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Técnicas de Cocultura , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/terapia , Sobrevivência de Enxerto/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Ilhotas Pancreáticas/imunologia , Transplante das Ilhotas Pancreáticas/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos Transgênicos , Transplante Heterólogo/métodos
12.
Cytotherapy ; 19(9): 1035-1047, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28760351

RESUMO

BACKGROUND AIMS: Major challenges in de-differentiated liposarcoma (DDLPS) therapy are the high rate of sequential recurrence (>80%) and metastasis (20-30%) following surgical removal. However, well-defined therapeutic strategies for this rare malignancy are lacking and are critically needed. METHODS: We investigated a new approach to DDLPS therapy with mesenchymal stromal cells expressing herpes simplex virus-thymidine kinase (MSC-TK). In an effort to evaluate this efficacy, in vitro cytotoxicity of MSC-TK against DDLPS cells was analyzed using an apoptosis assay. For pre-clinical study, the MSC-TK-induced reduction in recurrence and metastasis was validated in a recurrent DDLPS model after the macroscopic complete resection and lung metastasis DDLPS model. RESULTS: MSC-TK induced apoptosis in DDLPS cells by bystander effects via gap junction intracellular communication (GJIC) of toxic ganciclovir (GCV). Recurrent DDLPS models following no residual tumor/microscopic tumor resection and lung metastasis DDLPS models were established, which suggested clinical relevance. MSC-TK markedly reduced locoregional recurrence rates and prolonged recurrence-free survival, thus increasing overall survival in the recurrent DDLPS model. MSC-TK followed by GCV treatment yielded a statistically significant reduction in early- and advanced-stage lung metastasis. DISCUSSION: This therapeutic strategy may serve as an alternative or additional strategy by applying MSC-TK to target residual tumors following surgical resection, thus reducing local relapse and metastasis in these patients.


Assuntos
Lipossarcoma/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Simplexvirus/genética , Timidina Quinase/genética , Administração Intravenosa , Animais , Apoptose/efeitos dos fármacos , Efeito Espectador , Comunicação Celular/efeitos dos fármacos , Ganciclovir/farmacologia , Terapia Genética/métodos , Humanos , Lipossarcoma/patologia , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/terapia , Camundongos , Recidiva Local de Neoplasia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cytotherapy ; 19(2): 170-180, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28024875

RESUMO

BACKGROUND: There are various types of adipose tissue in the human body, and their morphology is known to be closely related to cell function and metabolism. However, the functional differences among the mesenchymal stromal cells (MSCs) of different abdominal adipose tissues have not been clearly elucidated. METHODS: MSCs were isolated from different abdominal adipose tissues according to their regional distribution and included superficial subcutaneous, deep subcutaneous, omentum, mesentery and retroperitoneal MSCs. The immunophenotype, proliferative ability and angiogenic function of these MSCs were compared based on flow cytometry analysis, CCK-8 proliferation, in vitro differentiation, tubule formation and in vivo plug assay. RESULTS: The plastic adherence, cell morphology and general immunophenotype are similar among the MSCs. However, subcutaneous adipose tissue-derived MSCs have a faster growth rate and a higher level of CD146 expression than the other MSCs. Moreover, according to the fluorescence-activated cell sorting (FACS) enrichment procedure, the expression level of CD146 is positively related to the growth rate and angiogenic capability of MSCs. DISCUSSION: MSCs in adipose tissue showed slightly different characteristics depending on their location of origin, and they possessed different angiogenic abilities that were mediated by the expression of CD146. This study provides evidence that subcutaneous adipose tissue is the most appropriate source of MSCs for therapeutic cell transplantation in vascular disease.


Assuntos
Gordura Abdominal/citologia , Separação Celular/métodos , Citometria de Fluxo/métodos , Células-Tronco Mesenquimais/classificação , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica , Animais , Antígeno CD146/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunofenotipagem , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos
14.
Wound Repair Regen ; 24(4): 686-94, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27237949

RESUMO

As wound contraction in the cutaneous layer occurs rapidly in mice, mechanical means are typically used to deliberately expose the wound to properly investigate healing by secondary intention. Previously, silicon rings and splinting models were attempted to analyze histological recovery but prevention of surrounding epidermal cell migration and subsequent closure was minimal. Here, we developed an ideal chimney wound model to evaluate epidermal regeneration in murine under hESC-EC transplantation through histological analysis encompassing the three phases of regeneration: migration, proliferation, and remodeling. Human embryonic stem cell derived endothelial cells (hESC-EC) were transplanted due to possessing a well-known therapeutic effect in angiogenesis which also enhances epidermal repair to depict the process of regeneration. Following a standard 1 mm biopsy punch, a chimney manufactured by modifying a 1.7 mL microtube was simply inserted into the excisional wound to complete the modeling process. Under this model, the excisional wound remained fully exposed for 14 days and even after 4 weeks, only a thin transparent layer of epidermal tissue covered the wound site. This approach is able to more accurately depict epidermal repair in relation to histology while also being a user-friendly and cost-effective way to mimic human recovery in rodents and evaluate epithelial repair induced by a form of therapy.


Assuntos
Células Endoteliais/metabolismo , Células-Tronco Embrionárias Humanas/transplante , Regeneração/fisiologia , Transplante de Células-Tronco/métodos , Cicatrização/fisiologia , Ferimentos Penetrantes/fisiopatologia , Animais , Colágeno Tipo VIII/metabolismo , Análise Custo-Benefício , Modelos Animais de Doenças , Células Endoteliais/citologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ferimentos Penetrantes/terapia
15.
Biochem Biophys Res Commun ; 456(1): 110-5, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25446107

RESUMO

RNA leukemia viruses induce T-cell lymphoblastic lymphomas or myeloid leukemias. Infection of cells with Moloney murine leukemia virus (M-MuLV) up-regulates the expression of a number of cellular genes, including those involved in T-lymphocyte activation. Previously, we demonstrated that this up-regulation occurs via the trans-activation activity of the M-MuLV long terminal repeat (LTR) sequences which produce an LTR-encoded transcript. Sequence analysis of the LTR revealed a potential transcription unit for RNA polymerase III (Pol III) within the U3 region that is actively occupied by Pol II factors. Here, we provide the direct evidence of involvement of Pol III in the trans-activation process and demonstrate the precise localization of the intragenic control elements for accurate and active Pol III transcription. Deletions of a copy of the directed repeats and further immediate upstream sequences significantly abrogated the generation of LTR-encoded transcript and abolished the trans-activational activity, whereas the deletion of a copy of directed repeats alone proportionally reduced the transcript size, but still retained moderately high trans-activational activity. In electrophoretic mobility shift assay, the fraction containing a multiple transcription factor TFIIIC complex strongly bound to the LTR-U3 probe containing the essential control elements. The specificity of the DNA-TFIIIC interaction was confirmed by conducting competition assays with DNA fragments containing a genuine Pol III-transcribed gene, VA1, and by vaccinia virus infection which stimulates the expression of Pol III factors. However, a deletion mutant lacking an essential control element bound to the TFIIIC complex poorly, consequently resulting in weak Pol III transcription as assessed by an IRES-GFP reporter system. This correlation strongly supports the possibility that the generation of LTR-encoded transcript is directed by Pol III. Therefore, this finding suggests the involvement of Pol III transcription in the retrovirus-induced activation of cellular genes, potentially contributing to leukemogenesis.


Assuntos
RNA Polimerase III/metabolismo , Retroviridae/genética , Sequências Repetidas Terminais , Células 3T3 , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Citometria de Fluxo , Deleção de Genes , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Leucemia Murina de Moloney , Mutação , Plasmídeos/metabolismo , Transcrição Gênica , Ativação Transcricional , Fator de Necrose Tumoral alfa/metabolismo
16.
Exp Mol Pathol ; 97(3): 440-4, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25281918

RESUMO

Angiomyolipomas (AMLs) are relatively rare hamartomatous or benign tumors that occasionally occur as part of tuberous sclerosis complex (TSC). Mutations in either of the two genes, TSC1 and TSC2, have been attributed to the development of TSC. Between 1994 and January 2009, 83 patients were diagnosed with AML at the Samsung Medical Center. In that group of patients, 5 (6%) had AML with TSC (AML-TSC). Mutational analysis of the TSC2 gene was performed using 7 samples from the 5 AML-TSC patients and 14 samples from 14 patients with sporadic AML without TSC (AML-non-TSC). From this analysis, mutations in TSC genes were identified in 5 samples from the AML-TSC patients (mutation detection rate=71%) and 3 samples from AML-non-TSC patients (mutation detection rate=21%). In the case of AML-TSC, 6 mutations were found including 3 recurrent mutations and 3 novel mutations, while in the case of AML-non-TSC, 4 mutations were identified once, including 1 novel mutation. Also MLPA analysis of the TSC2 gene showed that TSC2 exon deletion is more frequently observed in AML-TSC patients than in AML-non-TSC patients. This is the first mutation and multiplex ligation-dependent probe amplification (MLPA) analyses of TSC2 in Korean AMLs that focus on TSC. This study provides data that are representative of the distribution of mutations and exon deletions at TSC genes in clinically diagnosed AML-TSC cases of the Korean population.


Assuntos
Angiomiolipoma/genética , Mutação , Esclerose Tuberosa/genética , Proteínas Supressoras de Tumor/genética , Adulto , Idoso , Animais , Análise Mutacional de DNA , Éxons , Feminino , Deleção de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Multiplex , República da Coreia , Proteína 2 do Complexo Esclerose Tuberosa , Adulto Jovem
17.
Oncotarget ; 5(19): 9065-78, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25238053

RESUMO

Liposarcoma is one of the most common histologic types of soft tissue sarcoma and is frequently an aggressive cancer with poor outcome. Hence, alternative approaches other than surgical excision are necessary to improve treatment of well-differentiated/dedifferentiated liposarcoma (WDLPS/DDLPS). For this reason, we performed a two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization-time of flight mass spectrometry/mass spectrometry (MALDI-TOF/MS) analysis to identify new factors for WDLPS and DDLPS. Among the selected candidate proteins, gankyrin, known to be an oncoprotein, showed a significantly high level of expression pattern and inversely low expression of p53/p21 in WDLPS and DDLPS tissues, suggesting possible utility as a new predictive factor. Moreover, inhibition of gankyrin not only led to reduction of in vitro cell growth ability including cell proliferation, colony-formation, and migration, but also in vivo DDLPS cell tumorigenesis, perhaps via downregulation of the p53 tumor suppressor gene and its p21 target and also reduction of AKT/mTOR signal activation. This study identifies gankyrin, for the first time, as new potential predictive and oncogenic factor of WDLPS and DDLPS, suggesting the potential for service as a future LPS therapeutic approach.


Assuntos
Biomarcadores Tumorais/genética , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Lipossarcoma/patologia , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Proto-Oncogênicas/genética , Proteína Supressora de Tumor p53/biossíntese , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação para Baixo , Células HEK293 , Humanos , Lipossarcoma/tratamento farmacológico , Camundongos , Prognóstico , Complexo de Endopeptidases do Proteassoma/biossíntese , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/biossíntese , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Serina-Treonina Quinases TOR/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Biochem Biophys Res Commun ; 450(2): 984-90, 2014 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-24971534

RESUMO

The epithelium-specific ETS transcription factor-1 (ESE-1) is physiologically important in the pathogenesis of various diseases. Recently, OCT4, a transcription factor involved in stem cell pluripotency, has been implicated in tumorigenesis. In this study, we invested the molecular mechanism by which ESE-1 regulates transcription of OCT4 in NCCIT human embryonic carcinoma cells. Real-time PCR analysis revealed that OCT4 levels were high in undifferentiated NCCIT cells but significantly decreased upon retinoic acid-mediated differentiation, concomitant with up-regulation of ESE-1 expression. OCT4 mRNA level rose following shRNA-mediated knockdown of ESE-1, but declined when ESE-1 was overexpressed, suggesting that the expression levels of OCT4 and ESE-1 may be coordinated in an opposite manner. Promoter-reporter assays revealed that induced OCT4 promoter activity in NCCIT cells was significantly down-regulated by ESE-1 overexpression in a dose-dependent manner. The inhibitory effect of ESE-1 on OCT4 promoter activity was relieved by co-expression of an ESE-1 mutant lacking the transactivation domain, but not by mutants lacking other domains. Serial deletion and site-directed mutagenesis of the OCT4 promoter revealed that a potential ETS binding site (EBS) is present in the conserved region 2 (CR2). ESE-1 interacted with the EBS element in CR2 and enrichment of CR2 significantly increased upon RA-mediated differentiation of NCCIT cells, suggesting that this binding is likely to be involved in ESE-1-mediated repression of OCT4 promoter activity upon differentiation. Taken together, the results of this study reveal the molecular details of the mechanism by which the oncogenic factor ESE-1 regulates expression of the stem cell transcription factor OCT4 in pluripotent NCCIT cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco de Carcinoma Embrionário/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fatores de Transcrição/metabolismo , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Células-Tronco de Carcinoma Embrionário/citologia , Técnicas de Silenciamento de Genes , Humanos , Mutação , Células-Tronco Pluripotentes/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ets/genética , Fatores de Transcrição/genética , Transcrição Gênica , Ativação Transcricional
19.
Int J Gynecol Cancer ; 23(9): 1552-60, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24172092

RESUMO

BACKGROUND AND OBJECTIVE: The aim of this study was to characterize primary cells from extrauterine carcinosarcoma (CS) and to establish a primary CS xenograft mouse model. METHODS: Primary cells were isolated from a patient with CS and cultured in vitro. Primary CS cells were verified for their ability to consecutively generate tumorigenesis in NOD/SCID mice. The properties of xenograft tumor and explants cells were investigated by immunohistochemistry, cytogenetic, and FACS analysis. Anticancer drug susceptibility of primary CS was analyzed using CCK-8. RESULTS: Primary CS cells greater than 27 passages in vitro showed an ability of a series of xenograft tumorigenesis in vivo having the same marker expression and cytogenetic character as that of original tumor. In addition, explants of xenograft tumors retained their original characteristics in the in vitro culture system. Finally, the analysis of the susceptibility to anticancer drug revealed that primary CS cells were susceptible to both doxorubicin and nilotinib, which are tyrosine kinase inhibitors. CONCLUSIONS: The primary CS cells and the primary CS xenograft tumorigenesis introduce a new therapeutic model for targeting cancer and also explore a deeper understanding of generation of the tumor itself.


Assuntos
Neoplasias Abdominais/patologia , Carcinossarcoma/patologia , Xenoenxertos , Transplante de Neoplasias/métodos , Parede Abdominal/patologia , Animais , Endometriose/complicações , Endometriose/patologia , Evolução Fatal , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Cultura Primária de Células , Células Tumorais Cultivadas
20.
Arterioscler Thromb Vasc Biol ; 33(12): 2839-49, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24092748

RESUMO

OBJECTIVE: Allogeneic transplantation of human embryonic stem cell (hESC) derivatives has the potential to elicit the patient's immune response and lead to graft rejection. Although hESCs and their derivatives have been shown to have advantageous immune properties in vitro, such observations could not be determined experimentally in vivo because of ethical and technical constraints. However, the generation of humanized mice (hu-mice) harboring a human immune system has provided a tool to perform in vivo immunologic studies of human cells and tissues. Using this model, we sought to examine the therapeutic potential of hESC-derived endothelial cells, human embryonic fibroblasts, and cord blood-derived endothelial progenitor cells in a human immune system environment. APPROACH AND RESULTS: All cell types transplanted in hu-mice showed significantly reduced cell survival during the first 14 days post-transplantation compared with that observed in immunodeficient mice. During this period, no observable therapeutic effects were detected in the hindlimb ischemic mouse models. After this point, the cells demonstrated improved survival and contributed to a long-term improvement in blood perfusion. All cell types showed reduced therapeutic efficacy in hu-mice compared with NOD scid IL2 receptor gamma chain knockout mice. Interestingly, the eventual improvement in blood flow caused by the hESC-derived endothelial cells in hu-mice was not much lower than that observed in NOD scid IL2 receptor gamma chain knockout mice. CONCLUSIONS: These findings suggest that hESC derivatives may be considered a good source for cell therapy and that hu-mice could be used as a preclinical in vivo animal model for the evaluation of therapeutic efficacy to predict the outcomes of human clinical trials.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Células-Tronco Embrionárias/transplante , Células Endoteliais/transplante , Sangue Fetal/imunologia , Isquemia/cirurgia , Músculo Esquelético/irrigação sanguínea , Animais , Biomarcadores/sangue , Linhagem Celular , Sobrevivência Celular , Modelos Animais de Doenças , Células-Tronco Embrionárias/imunologia , Células Endoteliais/imunologia , Fibroblastos/imunologia , Fibroblastos/transplante , Sobrevivência de Enxerto , Membro Posterior , Humanos , Imunocompetência , Hospedeiro Imunocomprometido , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Isquemia/imunologia , Isquemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Neovascularização Fisiológica , Recuperação de Função Fisiológica , Fluxo Sanguíneo Regional , Especificidade da Espécie , Fatores de Tempo , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...