Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Stem Cell Res Ther ; 15(1): 58, 2024 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-38433223

RESUMO

OBJECTIVES: Currently, no approved stem cell-based therapies for preserving ovarian function during aging. To solve this problem, we developed a long-term treatment for human embryonic stem cell-derived mesenchymal progenitor cells (hESC-MPCs). We investigated whether the cells retained their ability to resist ovarian aging, which leads to delayed reproductive senescence. MATERIALS AND METHODS: In a middle-aged female model undergoing natural aging, we analyzed whether hESC-MPCs benefit the long-term maintenance of reproductive fecundity and ovarian reservoirs and how their transplantation regulates ovarian function. RESULTS: The number of primordial follicles and mice with regular estrous cycles were increased in perimenopausal mice who underwent multiple introductions of hESC-MPCs compared to age-matched controls. The estradiol levels in the hESC-MPCs group were restored to those in the young and adult groups. Embryonic development and live birth rates were higher in the hESC-MPC group than in the control group, suggesting that hESC-MPCs delayed ovarian senescence. In addition to their direct effects on the ovary, multiple-treatments with hESC-MPCs reduced ovarian fibrosis by downregulating inflammation and fibrosis-related genes via the suppression of myeloid-derived suppressor cells (MDSCs) produced in the bone marrow. CONCLUSIONS: Multiple introductions of hESC-MPCs could be a useful approach to prevent female reproductive senescence and that these cells are promising sources for cell therapy to postpone the ovarian aging and retain fecundity in perimenopausal women.


Assuntos
Células-Tronco Embrionárias Humanas , Células-Tronco Mesenquimais , Adulto , Gravidez , Pessoa de Meia-Idade , Feminino , Humanos , Animais , Camundongos , Perimenopausa , Fertilidade , Envelhecimento , Fibrose
2.
BMB Rep ; 56(8): 439-444, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37357536

RESUMO

Emphysema is a chronic obstructive lung disease characterized by inflammation and enlargement of the air spaces. Regorafenib, a potential senomorphic drug, exhibited a therapeutic effect in porcine pancreatic elastase (PPE)-induced emphysema in mice. In the current study we examined the preventive role of regorafenib in development of emphysema. Lung function tests and morphometry showed that oral administration of regorafenib (5 mg/kg/day) for seven days after instillation of PPE resulted in attenuation of emphysema. Mechanistically, regorafenib reduced the recruitment of inflammatory cells, particularly macrophages and neutrophils, in bronchoalveolar lavage fluid. In agreement with these findings, measurements using a cytokine array and ELISA showed that expression of inflammatory mediators including interleukin (IL)-1ß, IL-6, and CXCL1/KC, and tissue inhibitor of matrix metalloprotease-1 (TIMP-1), was downregulated. The results of immunohistochemical analysis confirmed that expression of IL-6, CXCL1/KC, and TIMP-1 was reduced in the lung parenchyma. Collectively, the results support the preventive role of regorafenib in development of emphysema in mice and provide mechanistic insights into prevention strategies. [BMB Reports 2023; 56(8): 439-444].


Assuntos
Enfisema , Enfisema Pulmonar , Animais , Camundongos , Modelos Animais de Doenças , Enfisema/tratamento farmacológico , Interleucina-6 , Pulmão/metabolismo , Camundongos Endogâmicos C57BL , Elastase Pancreática , Enfisema Pulmonar/induzido quimicamente , Enfisema Pulmonar/tratamento farmacológico , Enfisema Pulmonar/metabolismo , Suínos , Inibidor Tecidual de Metaloproteinase-1/farmacologia , Inibidor Tecidual de Metaloproteinase-1/uso terapêutico
3.
Virchows Arch ; 480(2): 449-457, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34510267

RESUMO

Slug is a transcription factor belonging to the slug/snail superfamily. The protein is involved in embryonic development and epithelial-mesenchymal transition of tumors. Slug is also under temporal regulation during cell cycle. Here, we examined relationship between pSlugS158 (site-specific phosphorylation) and the cell cycle, and checked whether its phosphorylation level reflects mitotic activity in tissue specimens. Cell cycle analysis was performed after cell synchronization. To evaluate pSlugS158 identifying mitotic figures, we performed immunohistochemistry (IHC) for pSlugS158 in various formalin-fixed paraffin-embedded tissues; in addition, mitotic counts were compared with those in sections stained with hematoxylin and eosin (HE) and IHC for PHH3, a mitotic marker. We found that the level of pSlugS158 protein increased specifically at M phase and decreased at the G1/S phases in vitro. In almost all tested tissues, nuclear stain of pSlugS158 was identified in the cell with mitotic figures. There was no significant difference in mitotic counts between HE- and pSlugS158-stained sections. In conclusion, pSlugS158 may be a novel and practical immunohistochemical marker for detecting mitotic figures in human tissues.


Assuntos
Biomarcadores Tumorais , Mitose , Fatores de Transcrição da Família Snail , Biomarcadores Tumorais/análise , Amarelo de Eosina-(YS) , Hematoxilina , Humanos , Imuno-Histoquímica , Índice Mitótico , Fosforilação , Projetos Piloto
4.
Stem Cell Res Ther ; 12(1): 431, 2021 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-34332643

RESUMO

BACKGROUND: Premature ovarian insufficiency (POI) is one of the most serious side effects of chemotherapy in young cancer survivors. It may not only reduce fecundity but also affect lifelong health. There is no standard therapy for preserving ovarian health after chemotherapy. Recently, administration of embryonic stem cell-derived mesenchymal progenitor cells (ESC-MPCs) has been considered a new therapeutic option for preventing POI. However, the previous method of directly injecting cells into the veins of patients exhibits low efficacy and safety. This study aimed to develop safe and effective local delivery methods for the prevention of POI using two types of bioinspired scaffolds. METHODS: Female mice received intraperitoneal cisplatin for 10 days. On day 11, human ESC-MPCs were delivered through systemic administration using intravenous injection or local administration using intradermal injection and intradermal transplantation with a PLGA/MH sponge or hyaluronic acid (HA) gel (GEL) type of scaffold. PBS was injected intravenously as a negative control. Ovarian function and fertility were evaluated 4 weeks after transplantation. Follicle development was observed using hematoxylin and eosin staining. The plasma levels of sex hormones were measured using ELISA. Expression levels of anti-Müllerian hormone (AMH) and ki-67 were detected using immunostaining, and the quality of oocytes and embryos was evaluated after in vitro fertilization. The estrous cycles were observed at 2 months after transplantation. RESULTS: The local administration of human ESC-MPCs using the bioinspired scaffold to the backs of mice effectively prolonged the cell survival rate in vivo. The HA GEL group exhibited the best recovered ovarian functions, including a significantly increased number of ovarian reserves, estrogen levels, and AMH levels and decreased apoptotic levels. Furthermore, the HA GEL group showed improved quality of oocytes and embryos and estrous cycle regularity. CONCLUSIONS: HA GEL scaffolds can be used as new delivery platforms for ESC-MPC therapy, and this method may provide a novel option for the clinical treatment of chemotherapy-induced POI.


Assuntos
Antineoplásicos , Células-Tronco Embrionárias Humanas , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Insuficiência Ovariana Primária , Animais , Feminino , Humanos , Camundongos , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/prevenção & controle
5.
Tissue Eng Regen Med ; 18(4): 651-662, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34165777

RESUMO

BACKGROUND: Leydig cells (LCs) are testicular somatic cells that are the major producers of testosterone in males. Testosterone is essential for male physiology and reproduction. Reduced testosterone levels lead to hypogonadism and are associated with diverse pathologies, such as neuronal dysfunction, cardiovascular disease, and metabolic syndrome. LC transplantation is a promising therapy for hypogonadism; however, the number of LCs in the testis is very rare and they do not proliferate in vitro. Therefore, there is a need for an alternative source of LCs. METHODS: To develop a safer, simple, and rapid strategy to generate human LC-like cells (LLCs) from stem cells, we first performed preliminary tests under different conditions for the induction of LLCs from human CD34/CD73 double positive-testis-derived stem cells (HTSCs). Based on the embryological sequence of events, we suggested a 3-step strategy for the differentiation of human ESCs into LLCs. We generated the mesendoderm in the first stage and intermediate mesoderm (IM) in the second stage and optimized the conditions for differentiation of IM into LLCs by comparing the secreted testosterone levels of each group. RESULTS: HTSCs and human embryonic stem cells can be directly differentiated into LLCs by defined molecular compounds within a short period. Human ESC-derived LLCs can secrete testosterone and express steroidogenic markers. CONCLUSION: We developed a rapid and efficient protocol for the production of LLCs from stem cells using defined molecular compounds. These findings provide a new therapeutic cell source for male hypogonadism.


Assuntos
Células-Tronco Embrionárias Humanas , Hipogonadismo , Diferenciação Celular , Humanos , Células Intersticiais do Testículo , Masculino , Testosterona
6.
BMB Rep ; 54(7): 380-385, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34154701

RESUMO

Proper targeting of the ßPAK-interacting exchange factor (ßPIX)/G protein-coupled receptor kinase-interacting target protein (GIT) complex into distinct cellular compartments is essential for its diverse functions including neurite extension and synaptogenesis. However, the mechanism for translocation of this complex is still unknown. In the present study, we reported that the conventional kinesin, called kinesin-1, can transport the ßPIX/GIT complex. Additionally, ßPIX bind to KIF5A, a neuronal isoform of kinesin-1 heavy chain, but not KIF1 and KIF3. Mapping analysis revealed that the tail of KIF5s and LZ domain of ßPIX were the respective binding domains. Silencing KIF5A or the expression of a variety of mutant forms of KIF5A inhibited ßPIX targeting the neurite tips in PC12 cells. Furthermore, truncated mutants of ßPIX without LZ domain did not interact with KIF5A, and were unable to target the neurite tips in PC12 cells. These results defined kinesin-1 as a motor protein of ßPIX, and may provide new insights into ßPIX/GIT complex-dependent neuronal pathophysiology. [BMB Reports 2021; 54(7): 380-385].


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cinesinas/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Animais , Proteínas de Ciclo Celular/fisiologia , Neurônios/metabolismo , Células PC12 , Isoformas de Proteínas/metabolismo , Ratos , Fatores de Troca de Nucleotídeo Guanina Rho/fisiologia
7.
Cells ; 10(6)2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34073789

RESUMO

Mesenchymal progenitor cells (MPCs) are a promising cell source for regenerative medicine because of their immunomodulatory properties, anti-inflammatory molecule secretion, and replacement of damaged cells. Despite these advantages, heterogeneity in functional potential and limited proliferation capacity of MPCs, as well as the lack of suitable markers for product potency, hamper the development of large-scale manufacturing processes of MPCs. Therefore, there is a sustained need to develop highly proliferative and standardized MPCs in vitro and find suitable functional markers for measuring product potency. In this study, three lines of pluripotent stem cell (PSC)-derived MPCs with high proliferative ability were established and compared with bone-marrow-derived MPCs using proliferation assays and microarrays. A total of six genes were significantly overexpressed (>10-fold) in the highest proliferative MPC line (CHA-hNT5-MPCs) and validated by qRT-PCR. However, only two of the genes (MYOCD and ODZ2) demonstrated a significant correlation with MPC senescence in vitro. Our study provides new gene markers for predicting replicative senescence and the available quantity of MPCs but may also help to guide the development of new standard criteria for manufacturing.


Assuntos
Antígenos de Diferenciação/biossíntese , Proliferação de Células , Senescência Celular , Células-Tronco Mesenquimais/metabolismo , Antígenos de Diferenciação/genética , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos
8.
Investig Clin Urol ; 62(4): 399-407, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34085786

RESUMO

PURPOSE: P21-activated kinase 4 (PAK4), a serine/threonine kinase that regulates a number of fundamental cellular processes, has been suggested as a prognostic factor for various human tumors. The aim of the present study was to evaluate the clinical implications of phospho-Ser474 PAK4 (pPAK4S474), an activated form of PAK4, in surgically treated renal cell carcinoma (RCC). MATERIALS AND METHODS: Samples from 131 patients with surgically treated RCC were immunostained to detect PAK4 and pPAK4S474. Expression of PAK4 and pPAK4S474 was compared with clinicopathological characteristics and survival after nephrectomy. RESULTS: PAK4 and pPAK4S474 were expressed predominantly in the nucleus. Overall, 57.3% (75/131) and 24.4% (29/119) of specimens exhibited high expression of pPAK4S474 and PAK4, respectively. High expression of pPAK4S474 was associated with adverse pathologic characteristics, including advanced tumor stage and grade (p=0.036 and p=0.002, respectively), whereas this association was not significant for PAK4 expression (each p>0.05). Kaplan-Meier estimates showed that high expression of pPAK4S474 was associated with shorter recurrence-free survival in a subgroup with localized RCC and with cancer-specific survival in the total RCC cohort (log-rank test: p=0.001 and p=0.005, respectively), whereas PAK4 expression was not. Multivariate Cox regression analysis identified that high pPAK4S474 expression was an independent predictor of recurrence in the subgroup with localized RCC. CONCLUSIONS: pPAK4S474 may be a more accurate prognostic factor than total PAK4 in RCC patients. This marker would be useful for identifying patients with pathologically localized disease who may require further interventions.


Assuntos
Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Recidiva Local de Neoplasia/metabolismo , Quinases Ativadas por p21/metabolismo , Idoso , Carcinoma de Células Renais/cirurgia , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Feminino , Humanos , Estimativa de Kaplan-Meier , Neoplasias Renais/cirurgia , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Fosforilação , Prognóstico , Modelos de Riscos Proporcionais , Taxa de Sobrevida
9.
Ageing Res Rev ; 68: 101332, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33753287

RESUMO

Cellular senescence occurs in response to diverse stresses (e.g., telomere shortening, DNA damage, oxidative stress, oncogene activation). A growing body of evidence indicates that alterations in multiple components of endocytic pathways contribute to cellular senescence. Clathrin-mediated endocytosis (CME) and caveolae-mediated endocytosis (CavME) represent major types of endocytosis that are implicated in senescence. More recent research has also identified a chromatin modifier and tumor suppressor that contributes to the induction of senescence via altered endocytosis. Here, molecular regulators of aberrant endocytosis-induced senescence are reviewed and discussed in the context of their capacity to serve as senescence-inducing stressors or modifiers.


Assuntos
Senescência Celular , Endocitose , Dano ao DNA , Humanos , Estresse Oxidativo
10.
J Periodontal Implant Sci ; 50(6): 379-391, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33350178

RESUMO

PURPOSE: The aims of this study were to evaluate the 5-year cumulative survival rate (CSR) of implants placed with guided bone regeneration (GBR) compared to implants placed in native bone, and to identify factors contributing to implant failure in regenerated bone. METHODS: This retrospective cohort study included 240 patients who had implant placement either with a GBR procedure (regenerated bone group) or with pristine bone (native bone group). Data on demographic features (age, sex, smoking, and medical history), location of the implant, implant-specific features, and grafting procedures and materials were collected. The 5-year CSRs in both groups were estimated using Kaplan-Meier analysis. Risk factors for implant failure were analyzed with a Cox proportional hazards model. RESULTS: In total, 264 implants in the native bone group and 133 implants in the regenerated bone group were analyzed. The 5-year CSRs were 96.4% in the regenerated bone group and 97.5% in the native bone group, which was not a significant difference. The multivariable analysis confirmed that bone status was not an independent risk factor for implant failure. However, smoking significantly increased the failure rate (hazard ratio, 10.7; P=0.002). CONCLUSIONS: The 5-year CSR of implants placed in regenerated bone using GBR was comparable to that of implants placed in native bone. Smoking significantly increased the risk of implant failure in both groups.

11.
Stem Cell Res Ther ; 11(1): 255, 2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32586410

RESUMO

BACKGROUND: Clinical use of mesenchymal stem cells (MSCs) requires a uniform cell population, and their harvesting is invasive and produces a limited number of cells. Human embryonic stem cell-derived MSCs (hESC-MSCs) can differentiate into three germ layers and possess immunosuppressive effects in vitro. Anticancer treatment is a well-known risk factor for premature ovarian failure (POF). In this study, we investigated the effect of hESC-MSC on recovery of ovarian function in cisplatin-induced POF in mice. METHODS: Female mice received intraperitoneal cisplatin for 10 days. On day 12, CHA15-derived hESC-MSCs were transplanted into the mice by tail vein injection. An injection of PBS served as the negative control. Ovaries were removed 28 days after transplantation for assessment of ovarian histology, immunostaining, and fertility testing by superovulation and in vitro fertilization. hESC-MSC transplantation into mice with cisplatin-induced damage restored body weight and ovary size. RESULTS: Mean primary and primordial follicle counts in the hESC-MSC group were significantly improved compared to the PBS group (P < 0.05), and counts of zona pellucida remnants, an apoptotic sign in ovarian follicles, were significantly reduced (P < 0.05). TUNEL assays and cleaved PARP immunostaining indicated apoptosis, which led to loss of ovarian stromal cells in negative control mice, while Ki-67 was higher in the hESC-MSC group and in non-cisplatin-treated controls than in the PBS group. Ovulation was reduced in the PBS group but recovered significantly in the hESC-MSC group. Rates of blastocyst formation from ovulated eggs and live births per mouse also recovered significantly in the hESC-MSC group. CONCLUSIONS: hESC-MSC restored structure and function in the cisplatin-damaged ovary. Our study provides new insights into the great clinical potential of human hESC-MSC in treating POF.


Assuntos
Células-Tronco Embrionárias Humanas , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Insuficiência Ovariana Primária , Animais , Cisplatino/toxicidade , Feminino , Humanos , Camundongos , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/terapia
12.
Oncol Lett ; 18(3): 2661-2669, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31452748

RESUMO

The aim of the present study was to evaluate programmed death-1 (PD-1) and programmed death-1 ligand-1 (PD-L1) expression in gastric carcinoma and to assess their effect on survival rate. A total of 170 surgically resected specimens were obtained from patients diagnosed with gastric carcinoma at St. Vincents Hospital, The Catholic University of Korea. Paraffin tissue sections from tissue microarray blocks were subjected to immunohistochemical analysis of PD-1 and PD-L1. In addition, PD-1 expression on CD4+ and CD8+ T cells isolated from peripheral blood mononuclear cells and gastric cancer tissues was evaluated by multicolor flow cytometry. PD-1 and PD-L1 were expressed in 30.0 and 60.5% of the gastric cancer tissues, respectively. The expression of PD-L1 was higher in patients with advanced T (P=0.035) and Tumor, Node and Metastasis stage (P=0.05). The patients with positive PD-L1 expression had shorter disease-free survival time than those without PD-L1 expression (P=0.005). Additionally, PD-L1 expression was significantly associated with poor prognosis (P=0.015). PD-1 and PD-L1 expression levels were significantly higher on CD8+ T cells than on CD4+ T cells (P<0.001). The data of the present study suggested that PD-L1 expression may be an independent indicator of poor prognosis in patients with gastric cancer. Furthermore, PD-L1 expression may play a role in immune evasion of gastric cancer.

13.
J Ocul Pharmacol Ther ; 35(4): 254-261, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31033390

RESUMO

Purpose: To investigate the anti-(lymph)angiogenic and anti-inflammatory effects of albendazole and to study whether these effects are additive with bevacizumab therapy in a murine corneal suture model. Methods: Corneal neovascularization (NV) and lymphangiogenesis (LY) were compared in a corneal suture model after administration of a subconjunctival injection of albendazole, bevacizumab, dexamethasone, or phosphate-buffered saline (PBS). Immunohistochemical staining and analysis were performed in each group. Real-time polymerase chain reaction (RT-PCR) was performed to quantify the expression of inflammatory cytokines (tumor necrosis factor [TNF]-alpha and interleukin-6), vascular endothelial growth factor (VEGF)-A, VEGF-C, vascular endothelial growth factor receptor (VEGFR)-2, and VEGFR-3. To evaluate the additive effect of albendazole, corneal NV and LY were also analyzed in a combined group of albendazole and bevacizumab therapy and the additive effect was compared with that in the group of double dose of bevacizumab. Results: The albendazole group showed less NV and less LY compared with the PBS control group (P < 0.01). When albendazole was combined with bevacizumab therapy, a significant decrease in NV and LY was seen compared with bevacizumab treatment alone, and with albendazole alone (all P values <0.05). The combination group showed better antilymphangiogenesis effect than the group of double dose bevacizumab. The albendazole-treated group showed reduced expression of VEGF-A, VEGF-C, TNF-alpha, and VEGFR-2 compared with corneas from the PBS group (P value <0.05 in all respective comparisons). Conclusion: Albendazole significantly decreased NV and LY in the cornea. This beneficial effect is additively enhanced when combined with bevacizumab treatment.


Assuntos
Albendazol/uso terapêutico , Inibidores da Angiogênese/farmacologia , Córnea/efeitos dos fármacos , Neovascularização da Córnea/tratamento farmacológico , Albendazol/administração & dosagem , Inibidores da Angiogênese/administração & dosagem , Animais , Bevacizumab/administração & dosagem , Bevacizumab/farmacologia , Córnea/patologia , Córnea/cirurgia , Neovascularização da Córnea/patologia , Neovascularização da Córnea/cirurgia , Modelos Animais de Doenças , Injeções Intraoculares , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência
14.
Exp Mol Med ; 51(2): 1-9, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755582

RESUMO

p21-Activated kinase 4 (PAK4), a member of the PAK family, regulates a wide range of cellular functions, including cell adhesion, migration, proliferation, and survival. Dysregulation of its expression and activity thus contributes to the development of diverse pathological conditions. PAK4 plays a pivotal role in cancer progression by accelerating the epithelial-mesenchymal transition, invasion, and metastasis. Therefore, PAK4 is regarded as an attractive therapeutic target in diverse types of cancers, prompting the development of PAK4-specific inhibitors as anticancer drugs; however, these drugs have not yet been successful. PAK4 is essential for embryonic brain development and has a neuroprotective function. A long list of PAK4 effectors has been reported. Recently, the transcription factor CREB has emerged as a novel effector of PAK4. This finding has broad implications for the role of PAK4 in health and disease because CREB-mediated transcriptional reprogramming involves a wide range of genes. In this article, we review the PAK4 signaling pathways involved in prostate cancer, Parkinson's disease, and melanogenesis, focusing in particular on the PAK4-CREB axis.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Suscetibilidade a Doenças , Transdução de Sinais , Quinases Ativadas por p21/metabolismo , Animais , Humanos , Melaninas/biossíntese , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Quinases Ativadas por p21/química
15.
Cell Transplant ; 27(10): 1523-1534, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30215278

RESUMO

Sertoli cells (SCs) in the mammalian testes are well known as supporting cells of spermatogenesis, but have recently become an attractive source of cell therapy because of their capacity for immune modulation and trophic effects. In order to increase their applicable efficacy, we demonstrate a novel differentiation method for mouse embryonic stem cell (ESC)-derived Sertoli-like cells (SLCs) via the intermediate mesoderm (IM). We show that IM derived from an induction of 6 days expressed markers such as Wt1, Lhx1, Pax2 and Osr1, and that a sequential induction of 6 days resulted in ESC-SLCs. The SLCs expressed their marker genes ( Sf1, Sox9, Gata4, Wt1, Fshr and Scf), but the pluripotency-marker gene Oct4 was decreased. After sorting by FSHR expression, high-purity (> 90%) SLCs were collected that showed distinct characteristics of SCs such as high phagocytic and immune modulation activities as well as the expression of immune-related genes. In addition, when transplanted into the seminiferous tubule of busulfan-treated mice, SLCs re-located and were maintained in the basal region of the tubule. These results demonstrated that our robust sequential differentiation system produced functional SLCs from mouse ESCs in vitro.


Assuntos
Mesoderma/citologia , Células-Tronco Embrionárias Murinas/citologia , Células de Sertoli/citologia , Animais , Diferenciação Celular , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose
16.
Oncogene ; 37(38): 5147-5159, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29849120

RESUMO

Epithelial-mesenchymal transition (EMT) facilitates cancer invasion and metastasis and thus accelerates cancer progression. p21-activated kinase 4 (PAK4) is a critical regulator of prostate cancer (PC) progression. Here, we report that PAK4 activation promotes PC progression through the EMT regulator Slug. We find that phosphorylated PAK4S474 (pPAK4) levels, an index of PAK4 activation, were tightly associated with Gleason score (p < 0.001), a clinical indicator of PC progression, but not with prostate serum antigen levels or tumor stage. Stable silencing of PAK4 in PC cells reduced their potential for EMT, cellular invasion, and metastasis in vivo. PAK4 bound and directly phosphorylated Slug at two previously unknown sites, S158 and S254, which resulted in its stabilization. The non-phosphorylatable form SlugS158A/S254A upregulated transcription of CDH1, which encodes E-cadherin, and thus suppressed EMT and invasion, to a greater extent than did wild-type Slug. The strong EMT inducer TGF-ß elevated pPAK4 and pSlugS158 levels; PAK4 knockdown or introduction of a dominant-negative form of PAK4 inhibited both TGF-ß-stimulated EMT and an increase in pSlugS158 levels. Finally, immunohistochemistry revealed a positive correlation between pPAK4 and pSlugS158 but an inverse correlation between pSlugS158 and E-cadherin. The results suggest that the PAK4-Slug axis represents a novel pathway that promotes PC progression.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias da Próstata/patologia , Fatores de Transcrição da Família Snail/metabolismo , Quinases Ativadas por p21/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Progressão da Doença , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Metástase Neoplásica , Fosforilação , Prognóstico , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/genética , Fatores de Transcrição da Família Snail/química , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Quinases Ativadas por p21/deficiência , Quinases Ativadas por p21/genética
17.
Exp Mol Med ; 50(5): 1-10, 2018 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29780159

RESUMO

Calcium mobilization is necessary for cell movement during embryonic development, lymphocyte synapse formation, wound healing, and cancer cell metastasis. Depletion of calcium in the lumen of the endoplasmic reticulum using inositol triphosphate (IP3) or thapsigargin (TG) is known to induce oligomerization and cytoskeleton-mediated translocation of stromal interaction molecule 1 (STIM1) to the plasma membrane, where it interacts with the calcium release-activated calcium channel Orai1 to mediate calcium influx; this process is referred to as store-operated calcium entry (SOCE). Furthermore, aberrant STIM1 or SOCE regulation is associated with cancer cell motility and metastasis. The p21-activated kinases (PAKs), which are downstream effectors of GTPases, reportedly regulate cytoskeletal organization, protrusive activity, and cell migration. Although cytoskeletal remodeling apparently contributes to calcium mobilization via SOCE, and vice versa, the mechanisms by which they regulate each other remain unclear. In this study, we aimed to characterize whether PAK1 modulates calcium mobilization and STIM1 localization. Our data demonstrate that PAK1 interacts with STIM1 in vitro and that this interaction was enhanced by treatment with a nascent adhesion inducer, such as phorbol 12,13-dibutyrate (PDBu). Under basal conditions, both proteins appeared to primarily colocalize in the cytosol, whereas treatment with PDBu induced their colocalization to vinculin-positive peripheral adhesions. Downregulation of PAK1 activity via chemical inhibitors or by PAK1 shDNA expression impaired STIM1-mediated calcium mobilization via SOCE. Based on these findings, we propose that PAK1 interacts with STIM1 to regulate calcium mobilization and the formation of cellular adhesions.


Assuntos
Cálcio/metabolismo , Quinases Ativadas por p21/metabolismo , Adesão Celular , Extensões da Superfície Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Espaço Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Fosforilação , Ligação Proteica , Molécula 1 de Interação Estromal/metabolismo , Vinculina/metabolismo
18.
Int J Ophthalmol ; 10(11): 1647-1654, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29181306

RESUMO

AIM: To investigate the anti-(lymph)angiogenic and/or anti-inflammatory effect of itraconazole in a corneal suture model and penetrating keratoplasty (PK) model. METHODS: Graft survival, corneal neovascularization, and corneal lymphangiogenesis were compared among itraconazole, amphotericin B, dexamethasone, phosphate buffered saline (PBS) and surgery-only groups following subconjunctival injection in mice that underwent PK and corneal suture. Immunohistochemical staining and analysis were performed in each group. Real-time polymerase chain reaction (RT-PCR) was performed to quantify the expression of inflammatory cytokines (TNF-alpha, IL-6) and vascular endothelial growth factor (VEGF)-A, VEGF-C, VEGFR-2, and VEGFR-3. RESULTS: In the suture model, the itraconazole group showed less angiogenesis, less lymphangiogenesis, and less inflammatory infiltration than the PBS group (all P<0.05). The itraconazole group showed reduced expression of VEGF-A, VEGFR-2, TNF-alpha, IL-6 than the PBS group (all P<0.05). In PK model, the two-month graft survival rate was 28.57% in itraconazole group, 62.50% in dexamethasone group, 12.50% in PBS group, 0 in amphotericin B group and 0 in surgery-only group. Graft survival in the itraconazole group was higher than that in the amphotericin, PBS and surgery-only group (P=0.057, 0.096, 0.012, respectively). The itraconazole group showed less total angiogenesis and lymphangiogenesis than PBS group (all P<0.05). CONCLUSION: Itraconazole decrease neovascularization, lymphangiogenesis, and inflammation in both a corneal suture model and PK model. Itraconazole has anti-(lymph)-angiogenic and anti-inflammatory effects in addition to its intrinsic antifungal effect and is therefore an alternative treatment option in cases where steroids cannot be used.

19.
Anticancer Res ; 37(11): 6269-6280, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29061810

RESUMO

Colchicine is an alkaloid widely used for the treatment of inflammatory diseases, such as gout. It suppresses cell division by inhibiting mitosis. We investigated the anticancer effects of colchicine on human hypopharyngeal cancer cells and the mechanisms underlying its anticancer effects. XTT cell proliferation assay showed that colchicine inhibited the growth and proliferation of human hypopharyngeal cancer cells (FaDu and SNU1041) in a dose- and time-dependent manner. Colchicine also inhibited the migration, invasion, and adhesion of hypopharyngeal cancer cells in a dose-dependent manner. The levels of mRNA expression and activity of matrix metalloproteinase-9 (MMP9) and urokinase-type plasminogen activator (uPA) decreased after treatment with colchicine. Further investigation revealed that colchicine inhibited the phosphorylation of the FAK/SRC complex and paxillin. Tumor volume ratios in colchicine-treated mice (0.1 mg/kg, every 2 days for 14 days) increased less than in control mice. To our knowledge, this is the first report showing that colchicine can suppress cell invasion, migration, and adhesion through reduced expression of MMP9, the uPA system, and the FAK/SRC complex. Colchicine has the potential to prevent disease progression in hypopharyngeal cancer and may have application as an adjunctive treatment.


Assuntos
Antineoplásicos/administração & dosagem , Colchicina/administração & dosagem , Neoplasias Hipofaríngeas/tratamento farmacológico , Metaloproteinase 9 da Matriz/genética , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colchicina/farmacologia , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Int J Ophthalmol ; 10(6): 834-839, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28730071

RESUMO

AIM: To evaluate the effect of sorafenib in murine high risk keratoplasty model. METHODS: Graft survival, corneal neovascularization, and corneal lymphangiogenesis were compared among the sorafenib, dexamethasone, dimethyl sulfoxide (DMSO), and phosphate buffered saline (PBS) groups following subconjunctival injection in mice that underwent high risk penetrating keratoplasty (HRPK). Real-time polymerase chain reaction was performed to quantify the expression of inflammatory cytokines and vascular endothelial growth factor (VEGF)-A, VEGF-C, vascular endothelial growth factor receptor (VEGFR)-2, VEGFR-3. RESULTS: The two-month graft survival rate for HRPK was 42.86% in sorafenib group, 37.50% in dexamethasone group, 0 in DMSO group, and 0 in PBS group. Sorafenib significantly increased graft survival compared to the DMSO and PBS group (P<0.05). The sorafenib didn't show significant effect in decreasing neovascularization compared with dexamethsone, DMSO, and PBS group. The sorafenib showed less total lymphangiogenesis than the dexamethasone, DMSO, and PBS group (P=0.011, P<0.001, P<0.001, respectively). The sorafenib group showed reduced expression of VEGF-C, tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, VEGFR-2 and VEGFR-3 compared with DMSO group and PBS group (all P<0.05). The sorafenib group didn't show difference in the expression of VEGF-A compared with DMSO, neither with PBS. The sorafenib group showed reduced expression of VEGFR-3 compared with dexamethasone (P=0.051). CONCLUSION: The subconjunctivally administered sorafenib shows significant anti-lymphangiogenic effect, resulting in increased transplant survival in a murine high risk keratoplasty model. We suggest that a close linkage between decreased VEGF-C/VEGFR-2 and -3 signaling and increased corneal graft survival by sorafenib seems to exist.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA