Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Rep ; 49(6): 4307-4319, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35525887

RESUMO

BACKGROUND: Shikonin is a natural multipotent anti-tumorigenic compound. We investigated potential synergy between shikonin and anti-diabetic metformin against tumorigenic properties of breast cancer cell line MCF-7. METHODS AND RESULTS: The IC50 of shikonin and metformin was determined after a single treatment of two cell lines MCF-7 and MDA-MB-231. We then measured optimal doses of each drug, used in combination, in MCF-7 cells. These sub-IC50 doses were co-applied for all subsequent combined treatments to evaluate their synergistic effects on MCF-7 tumorigenic properties. Next, we examined expression levels of the genes crucial for apoptosis, cell growth, and EMT using RT-PCR or real-time PCR and monitored CD44/CD24 ratios using flow cytometry. Binding energies between shikonin and growth molecules were measured by in silico simulation. Shikonin caused significantly reduced cell survival that was accelerated by the synergizing presence of metformin. Drug combination induced apoptosis and ROS levels while fully blocking cell migration and reverting EMT. RT-PCR showed strong suppression of BCL-2 but induction of BAX and PTEN. Prolonged shikonin treatment caused a total loss of the nuclear membrane, whereas metformin prevented this damage while promoting apoptotic morphologies. Our real-time PCR detected reduced levels of EMT genes but increases in the anti-EMT gene CDH1. Combined treatment also reduced CD44/CD24 ratios in favor of chemosensitivity. Binding energies strongly favored shikonin interactions with growth-signaling molecules. CONCLUSIONS: Shikonin and metformin synergize in inhibiting the tumorigenic activities of MCF-7 cells including their proliferation, invasiveness, and EMT with a potential to inhibit multidrug resistance.


Assuntos
Neoplasias da Mama , Metformina , Apoptose , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Humanos , Células MCF-7 , Metformina/farmacologia , Naftoquinonas
2.
J Cell Biochem ; 121(1): 200-212, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31310388

RESUMO

Human teratocarcinoma cell line Ntera2 (NT2) expresses dopamine signals and has shown its safe profile for clinical applications. Attempts to restore complete dopaminergic (DAergic) phenotype enabling these cells to secrete dopamine have not been fully successful so far. We applied a blend of gene transfer techniques and a defined medium to convert NT2 cells to fully DAergic. The cells were primarily engineered to overexpress the Pitx3 gene product and then cultured in a growth medium supplemented with knockout serum and retinoic acid to form embroid bodies (EBs). Trypsinization of EB colonies produced single cells ready for differentiation. Neuronal/DAergic induction was promoted by applying conditioned medium taken from engineered human astrocytomas over-secreting glial cell-derived neurotrophic factor (GDNF). Immunocytochemistry, reverse-transcription and real-time polymerase chain reaction analyses confirmed significantly induced expression of molecules involved in dopamine signaling and metabolism including tyrosine hydroxylase, Nurr1, dopamine transporter, and aromatic acid decarboxylase. High-performance liquid chromatography analysis indicated release of dopamine only from a class of fully differentiated cells expressing Pitx3 and exposed to GDNF. In addition, Pitx3 and GDNF additively promoted in vitro neuroprotection against Parkinsonian toxin. One month after transplantation to the striatum of 6-OHDA-leasioned rats, differentiated NT2 cells survived and induced significant increase in striatal volume. Besides, cell implantation improved motor coordination in Parkinson's disease (PD) rat models. Our findings highlight the importance of Pitx3-GDNF interplay in dopamine signaling and indicate that our strategy might be useful for the restoration of DAergic fate of NT2 cells to make them clinically applicable toward cell replacement therapy of PD.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Astrocitoma/metabolismo , Comportamento Animal , Diferenciação Celular , Linhagem Celular Tumoral , Meios de Cultivo Condicionados , Dopamina/metabolismo , Técnicas de Transferência de Genes , Teste de Complementação Genética , Células HEK293 , Humanos , Oxidopamina/farmacologia , Doença de Parkinson/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Tretinoína/metabolismo
3.
Mol Biotechnol ; 60(3): 185-193, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29322302

RESUMO

Viral gene delivery is hailed as a great milestone in gene-based therapeutic approaches. The human immunodeficiency virus-derived lentiviral vectors (LVs) are advantageous in infecting both dividing and non-dividing cells leading to continuous expression of transgenes. A variety of protocols are available for concentration of LVs. We primarily generated our internal ribosome entry site (IRES)-based LVs. Virus titration and transduction efficiency were compared between various strategies that included sucrose cushion centrifugation (SCC), protein column ultrafiltration and polyethylene glycol precipitation. Among these approaches, SCC resulted in concentration of high-titer EGFP-expressing lentivirus (1.4 ± 0.3 × 109 TU/ml) with the lowest protein impurities. Further, we examined transduction strengths of our three methods on two challenging stem cells. Both human NT2 and mouse bone marrow-derived mesenchymal stem cells demonstrated high transduction using SCC of 65 ± 2.8 and 49 ± 0.8%, respectively. Finally, lentivirus particles harboring IRES-based transfer vectors of specific genes, concentrated by SCC, integrated into host genome. Taken together, development of cost-effective and efficient concentration strategies such as our SCC method is yet highly demanded to broaden the horizons of lentivirus application in clinical and translational research.


Assuntos
Centrifugação/métodos , Lentivirus/metabolismo , Polietilenoglicóis/química , Células-Tronco/metabolismo , Sacarose/química , Transdução Genética , Ultrafiltração/métodos , Carga Viral , Animais , Linhagem Celular , Vetores Genéticos/metabolismo , Humanos , Camundongos , Recombinação Genética/genética , Transgenes
4.
Neural Regen Res ; 12(7): 1186-1192, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28852404

RESUMO

Cell replacement therapy utilizing mesenchymal stem cells as its main resource holds great promise for ultimate treatment of human neurological disorders. Parkinson's disease (PD) is a common, chronic neurodegenerative disorder hallmarked by localized degeneration of a specific set of dopaminergic neurons within a midbrain sub-region. The specific cell type and confined location of degenerating neurons make cell replacement therapy ideal for PD treatment since it mainly requires replenishment of lost dopaminergic neurons with fresh and functional ones. Endogenous as well as exogenous cell sources have been identified as candidate targets for cell replacement therapy in PD. In this review, umbilical cord mesenchymal stem cells (UCMSCs) are discussed as they provide an inexpensive unlimited reservoir differentiable towards functional dopaminergic neurons that potentially lead to long-lasting behavioral recovery in PD patients. We also present miRNAs-mediated neuronal differentiation of UCMSCs. The UCMSCs bear a number of outstanding characteristics including their non-tumorigenic, low-immunogenic properties that make them ideal for cell replacement therapy purposes. Nevertheless, more investigations as well as controlled clinical trials are required to thoroughly confirm the efficacy of UCMSCs for therapeutic medical-grade applications in PD.

5.
Rejuvenation Res ; 20(4): 309-319, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28437187

RESUMO

Human adipose-derived stem cells (hADSCs) have great capacity to differentiate into mesodermal origins as well as nonmesodermal lineages, including neural cells. This valuable feature paves the way for the therapeutic application of hADSCs for neurodegenerative maladies such as Parkinson's disease (PD). We tested the capacity of choroid plexus epithelial cell-conditioned medium (CPEC-CM) alone or cocktailed with knockout serum (KS) to induce dopaminergic (DAergic) differentiation of hADSCs. To this end, hADSCs from lipoaspirate were phenotypically characterized and shown to maintain mesodermal multipotency so that selected media easily differentiated them into osteoblasts, chondrocytes, and adipocytes. To begin inducing hADSC neuronal differentiation, we isolated CPECs from rat brain and expanded them in culture to obtain CPEC-CM. We then treated hADSCs with optimized quantities of collected CPEC-CM, KS, or both. The ADSCs treated with either CPEC-CM or CPEC-CM and KS displayed morphological changes typical of neuron-like phenotypes. As revealed by reverse transcription polymerase chain reaction (RT-PCR), quantitative real-time PCR (qPCR), and immunostaining analyses, hADSCs cotreated with CPEC-CM and KS expressed significantly higher levels of neuronal and DAergic markers in comparison with single-treated groups. Moreover, the hADSCs began expressing dopamine-biosynthesizing enzymes mainly after cotreatment with CPEC-CM and KS. Consequently, only cotreated hADSCs were capable of synthesizing and releasing dopamine detectable by high-performance liquid chromatography (HPLC). Finally, hADSCs growing in an ordinary medium were found positive for astrocytic marker glial fibrillary acidic protein (GFAP), but stopped GFAP expression on either single or cotreatments. These combined results suggest that CPEC-CM and KS can synergize to remarkably augment DAergic induction of hADSCs, an effect that has implications for cell replacement therapy for PD and related disorders.


Assuntos
Tecido Adiposo/citologia , Plexo Corióideo/citologia , Meios de Cultivo Condicionados/farmacologia , Dopamina/metabolismo , Células Epiteliais/citologia , Neurônios/metabolismo , Soro/metabolismo , Células-Tronco/citologia , Adipogenia/efeitos dos fármacos , Adulto , Animais , Biomarcadores/metabolismo , Linhagem da Célula/efeitos dos fármacos , Separação Celular , Forma Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Pessoa de Meia-Idade , Neurônios/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Ratos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Adulto Jovem
6.
Breast Cancer Res Treat ; 162(2): 231-241, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28116540

RESUMO

We investigated the role of glial cell line-derived neurotrophic factor (GDNF) in compensating trastuzumab (TZMB)-induced apoptosis in HER2+ breast cancer (BC) cells using xenograft tumors. We generated BC xenografts in nude mice using samples from three patients selected based on their HER2 status and response to TZMB therapy. TZMB treatment resulted in shrinkage of the HER2+ TZMB-sensitive xenograft tumor but not the HER2- or HER2+ TZMB-resistant ones. GDNF neutralized TZMB activity and induced growth in all tumors. Three distinct cell lines were derived from these tumors and named, respectively, TZMB-sensitive (TSTC), HER2- (HNTC), and TZMB-resistant (TRTC). Over 50% of TRTC but 1% of TSTC cells expressed CD44, whereas 84% of TSTC were CD24+ compared to only 1% of TRTC, despite comparable levels of HER2 detected in both. TZMB induced profound morphological changes toward apoptosis in TSTC but not in TRTC or HNTC. However, GDNF significantly compensated TZMB-mediated TSTC cell loss and promoted growth by 37 and 50%, respectively, in TSTC and TRTC. Inhibition of SRC by Saracatinib (SARC) blocked GDNF function and accelerated TZMB-mediated cell death in TSTC, but GDNF continued promoting TRTC growth. These changes paralleled with expression levels of the key molecules involved in growth and apoptosis. Collectively, we found in our xenograft samples that firstly SRC mediates GDNF pro-survival functions by bridging RET-HER2 crosstalk in TZMB-responsive BC tumors. Secondly, SARC-TZMB interactions can synergistically eradicate such tumor cells; and thirdly, GDNF can support antibody resistance by acting independent from SRC in tumors with poor HER2 response to TZMB therapy.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Receptor ErbB-2/metabolismo , Trastuzumab/farmacologia , Quinases da Família src/metabolismo , Animais , Apoptose , Benzodioxóis/farmacologia , Biomarcadores , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Antígeno CD24/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Expressão Gênica , Humanos , Receptores de Hialuronatos/metabolismo , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-ret/genética , Quinazolinas/farmacologia , Receptor ErbB-2/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/genética
7.
Arch Iran Med ; 19(8): 561-70, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27544365

RESUMO

BACKGROUND/OBJECTIVE: Degeneration of dopaminergic neurons in Parkinson's disease (PD) implies cell replacement using potentially differentiable sources as a promising therapeutic solution. We tested the capacity of conditioned medium from choroid plexus epithelial cells (CPECs-CM) to induce the dopaminergic potential of umbilical cord matrix mesenchymal stem cells (UCMSCs). METHODS: We isolated UCMSCs from human umbilical cord and CPECs from rat brain. Following expansion and characterization, CPECs-CM were collected, tested for expression of various growth factors, and applied to UCMSCs. Differentiation was examined and UCMSCs were injected into 6-OHDA-leasioned striatum to test their survival and function. RESULTS: RT-PCR and immuno-staining demonstrated neuronal/dopaminergic signaling in UCMSCs induced by CPECs-CM and accelerated by addition of retinoic acid (RA) and fibroblast growth factor-2. Expression of ß-tubulin-3, Nestin and MAP2 confirmed neuronal differentiation whereas tyrosine hydroxylase, aromatic acid decarboxylase and dopamine transporter were expressed as signs of dopaminergic differentiation. Post-transplantation, the UCMSCs survived, showed reduced rate of apoptosis and led to animals' recovery from apomorphine-induced rotations. CONCLUSION: The combination of neurotrophic factors present in CPECs-CM and RA can synergize to maximize dopaminergic differentiation of potential cell sources including UCMSCs. Our study may have implications for PD cell replacement therapy.


Assuntos
Apomorfina/farmacologia , Meios de Cultivo Condicionados/farmacologia , Agonistas de Dopamina/farmacologia , Neurônios Dopaminérgicos/metabolismo , Células Epiteliais/metabolismo , Células-Tronco Mesenquimais/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Plexo Corióideo/citologia , Modelos Animais de Doenças , Humanos , Doença de Parkinson Secundária/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Rotação , Cordão Umbilical/citologia
8.
Cancer Microenviron ; 9(1): 71-4, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26412079

RESUMO

Presence of tumor initiating cells and a proper niche is essential for metastatic colonization. SLUG and SOX9 transcription factors play essential roles in induction and maintenance of tumor initiating capacity in breast cancer cells. On the other hand, Tenascin-C and Periostin are crucial factors in metastatic niche that support tumor initiating capability in breast cancer. In this study, regulatory effect of SLUG and SOX9 transcription factors on the expression of Tenascin-C and Periostin was examined. SLUG and SOX9 were overexpressed and knocked-down in MCF7 and MDA-MB-231 cells, respectively. The cells as little and highly invasive breast cancer-derived cells were infected by inducing and shRNA lentivirus constructs. Then, Tenascin-C and Periostin as well as SLUG and SOX9 expression levels were measured in the cells via Real-Time PCR. Simultaneous overexpression of SLUG and SOX9 significantly induced Tenascin-C and Periostin expression. SLUG and SOX9 knock-down also significantly reduced the expression of Tenascin-C and Periostin. In this analysis Periostin showed the most deviation in both up- and down-regulation levels. This regulatory effect might shed light to a crosstalk between factors involved in the tumor initiating capacity and metastatic niche of the breast cancer.

9.
Cell J ; 15(3): 206-11, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24027660

RESUMO

OBJECTIVE: 8-Methoxypsoralen (8-MOP) is a photoactive compound widely used in the treatment of proliferate disorders. The present study investigates the effects of 8-MOP on ovary function and pituitary-gonad axis in mice. MATERIALS AND METHODS: : In this experimental analytical study, 45 female Balb/C mice were divided into three groups (n=15), control, sham (olive oil injection) and experimental. The experimental group were received an intraperitoneal (i.p.) injection of the LD50 dose of 60 mg/kg 8-MOP. At 30 days after injection, the animals were sacrificed while in the proestrus stage and examined for morphological and histological changes their ovaries. Blood samples were collected and estrogen, luteinizing hormone (LH) and follicle stimulating hormone (FSH) levels were assessed by radioimmunoassay. Data were analyzed using one-way ANOVA and the t test. RESULTS: The mean levels of estrogen and progesterone in the experimental group significantly decreased (p< 0.001). However, there was a significant increase in LH and FSH levels in this group compared to the control groups (p< 0.001). The mean number and diameter of the corpus luteum (CL) and the number of growing follicles in the experimental group significantly reduced compared to the control and sham groups (p< 0.001). The mean granulosa thickness in the experimental group also significantly decreased compared to the control and sham groups (p< 0.001). CONCLUSION: Our data indicated that 8-MOP can affect the levels of LH, FSH, estrogen and progesterone. Our findings further suggest that consecutive doses of 8-MOP may impair the female reproductive tract (or development).

10.
Cytotherapy ; 15(8): 951-60, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23732047

RESUMO

BACKGROUND AIMS: Therapeutic promises of adult stem cells have been overshadowed by an elicited immune response, low maintenance of implanted cells or concerns regarding their migration to non-target sites. These problems might be lessened by the use of immune privilege cells and tissues for implantation. METHODS: In this study, human adipose-derived mesenchymal stromal cells (hADMSCs) were stably transfected with a vector containing Turbo green fluorescent protein (GFP) and JRed, which allows tracing the cells after transplantation. Labeled hADMSCs were transplanted into the adult rat brain followed by assessment of their survival and migration during 6 months after transplantation. RESULTS: Results indicate that there were no postsurgical complications, and the animals thrived after transplantation. The lesions of the surgical process were remarkable at the first weeks, and a high number of transplanted cells were accumulated around them. Cell populations declined over time as they partly migrated away from the injection sites; nonetheless, they were detectable at each examination time point. Although the cells could survive and remain at the injection site for up to 6 months, some of them drifted to spleen, which is an indication of their ability to cross the blood-brain barrier. CONCLUSIONS: Despite the high survival rate of hADMSCs in the xenogenic condition, which is an ideal criterion in cell therapy, irregular migration tendency must be handled with caution.


Assuntos
Barreira Hematoencefálica/citologia , Sobrevivência de Enxerto , Transplante de Células-Tronco Mesenquimais , Tecido Adiposo/citologia , Animais , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Humanos , Células-Tronco Mesenquimais , Ratos , Ratos Wistar
11.
Xenotransplantation ; 20(3): 165-76, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23679842

RESUMO

BACKGROUND: Novel threads of discovery provide the basis for optimism for the development of a stem-cell-based strategy for the treatment of retinal blindness. Accordingly, achievement to suitable cell source with potential-to-long-term survival and appropriate differentiation can be an effective step in this direction. METHODS: After derivation of human adipose-derived mesenchymal stem cells (HAD-MSCs), they were stably transfected with a vector containing Turbo-green fluorescent protein (GFP) and JRed to be able to trace them after transplantation. Labeled HAD-MSCs were transplanted into the intact adult rat eye and their survival, integration, and migration during 6 months post-transplantation were assessed. RESULTS: The transplanted cells were traceable in the rat vitreous humor (VH) up until 90 days after transplantation, with gradual reduction in numbers, their adhesion and expansion capacity after recovery. These cells were also integrated into the ocular tissues. Nonetheless, some of the implanted cells succeeded to cross the blood-retina barrier (BRB) and accumulate in the spleen with time. CONCLUSIONS: The survival of the HAD-MSCs for a period of 90 days in VH and even longer period of up to 6 months in other eye tissues makes them a promising source to be considered in regenerative medicine of eye diseases. However, the potency of crossing the BRB by the implanted cells suggests that use of HAD-MSCs must be handled with extreme caution.


Assuntos
Olho/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Tecido Adiposo/citologia , Animais , Cegueira/patologia , Cegueira/cirurgia , Barreira Hematorretiniana , Diferenciação Celular , Sobrevivência Celular , Expressão Gênica , Xenoenxertos , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Procedimentos Cirúrgicos Oftalmológicos , Ratos , Ratos Wistar , Doenças Retinianas/patologia , Doenças Retinianas/cirurgia , Fatores de Tempo , Corpo Vítreo/citologia
12.
Neurochem Res ; 38(8): 1590-604, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23636806

RESUMO

We have investigated the mechanism of shikonin function on protection of dopaminergic neurons against 6-OHDA-induced neurotoxicity. Treatment of rat pheochromocytoma cell line PC12 by serial dilutions of shikonin determined 10 µM of the compound as its optimum concentration for protection saving nearly 70 % of the cells against toxicity. Reverse transcription-PCR analysis of shikonin-treated cells showed threefold increase in mRNA levels of glutathione peroxidase-1 (GPX-1) as a representative component of the intracellular anti-oxidant defense system. To elucidate shikonin-GPX1 relationships and maximize protection, we transduced PC12 cells using recombinant lentivirus vectors that harbored GPX-1 coding sequence. This change upregulated GPX-1 expression, increased peroxidase activity and made neuronal cells resistant to 6-OHDA-mediated toxicity. More importantly, addition of shikonin to GPX1-overexpressing PC12 cells augmented GPX-1 protein content by eightfold leading to fivefold increase of enzymatic activity, 91 % cell survival against neurotoxicity and concomitant increases in intracellular glutathione (GSH) levels. Depletion of intracellular GSH rendered all cell groups highly susceptible to toxicity; however, shikonin was capable of partially saving them. Subsequently, GSH-independent superoxide dismutase mRNA was found upregulated by shikonin. As signs of apoptosis inhibition, the compound upregulated Bcl-2, downregulated Bax, and prevented cell nuclei from undergoing morphological changes typical of apoptosis. Also, a co-staining method demonstrated GPX-1 overexpression significantly increases the percent of live cells that is maximized by shikonin treatment. Our data indicate that shikonin as an antioxidant compound protects dopaminergic neurons against 6-OHDA toxicity and enhances their survival via both glutathione-dependent and direct anti-apoptotic pathways.


Assuntos
Apoptose/efeitos dos fármacos , Dopamina/metabolismo , Glutationa/metabolismo , Naftoquinonas/farmacologia , Oxidopamina/farmacologia , Animais , Sequência de Bases , Primers do DNA , Glutationa Peroxidase/metabolismo , Células HEK293 , Humanos , Células PC12 , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Tumour Biol ; 34(4): 2019-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23686802

RESUMO

Metastatic colonization represents the final step of metastasis, and is the major cause of cancer mortality. Metastasis as an "inefficient" process requires the right population of tumor cells in a suitable microenvironment to form secondary tumors. Cancer stem cells are the only capable population of tumor cells to progress to overt metastasis. On the other hand, the occurrence of appropriate microenvironmental conditions within the target tissue would be critical for metastasis formation. Metastatic niche seems to be the specialized microenvironment to support tumor initiating cells at the distant organ. Master regulators not only determine cancer stem cell state, but also may have regulatory roles in metastatic niche elements. Meanwhile, both cancer stem cell and metastatic niche may function like two sides of the metastatic coin. Hypoxia inducible factors have multiple roles in regulation of both sides of this coin. TGF-ß superfamily, also, have been considered as master regulators of epithelial to mesenchymal transition and metastasis and may play crucial roles in regulation of metastatic niche as well. In this regard, we hypothesize the presence of a possible emerging molecular pathway in the biological process of breast cancer metastasis. In this process, non-Smad TGF-ß-induced metastasis connects cancer stem cell and metastatic niche formation through a central path, "Metastasis Pathway".


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal , Metástase Neoplásica , Células-Tronco Neoplásicas/metabolismo , Matriz Extracelular , Feminino , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral
14.
Rejuvenation Res ; 16(3): 185-99, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23472740

RESUMO

We examined the impact of strong co-presence and function of glutathione peroxidase-1 (GPX-1) and glial cell line-derived neurotrophic factor (GDNF) on protecting the rat dopaminergic pheochromocytoma cell line PC12 against 6-hydroxydopamine (6-OHDA) and hydrogen peroxide (H2O2) toxicities. Primarily, GPX-1 over-expression by PC12 cells infected with pLV-GPX1 lentivirus vectors significantly increased cell survival against 6-OHDA toxicity (p<0.01). Addition of conditioned medium collected from growing wild-type astrocytes (Control astro-CM) increased survival rate of pLV-GPX1 infectants by 10% compared to their un-treated counterparts (p<0.05) and 20% compared to their treated empty vector control (p<0.01). Treatment of pLV-GPX1 cells with astro-CM of GDNF-over-secreting astrocytes (Test astro-CM) significantly induced GPX-1 expression, peroxidase enzymatic activity, and intra-cellular glutathione (GSH) levels. These changes paralleled with protection of 90% of GDNF⁺/GPX1⁺ PC12 cells against toxicity, a rate that was 37% up from their un-infected un-treated (GDNF⁻/GPX1⁻) controls (p<0.001), and 12% up from pLV-GPX1 cells that received only Control astro-CM (GPX⁺/GDNF⁻) (p<0.01). GPX-1 over-expression per se suppressed intra-cellular H2O2 elevation upon 6-OHDA exposure, and addition of GDNF medium significantly accelerated this suppression (p<0.01). Substitution of 6-OHDA with H2O2 induced similar intra-cellular changes and comparable protection levels. In all cell groups, increased cell survival against either compound was further confirmed by increased live cell counts measured by double staining. Following depletion of intra-cellular GSH, only 46% of pLV-GPX1 cells survived 6-OHDA toxicity, whereas over 70% of them were saved upon GDNF treatment (p<0.001). Moreover, capase-3 activation was reduced in pLV-GPX1 cells and maximized by addition of GDNF. Comparison analyses established correlations between GPX-1-GDNF co-presence and both enhanced cell protection and diminished levels of activated caspase-3. Our data collectively indicate that GDNF is capable of inducing anti-oxidant activities of intra-cellular GPX-1 and that growth-promoting potential of GDNF and anti-oxidant properties of GPX-1 can, in concert, maximize survival of dopaminergic neurons.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/toxicidade , Neuroglia/metabolismo , Oxidopamina/metabolismo , Regulação para Cima , Animais , Sequência de Bases , Primers do DNA , Células HEK293 , Humanos , Neuroglia/efeitos dos fármacos , Neuroglia/enzimologia , Células PC12 , Reação em Cadeia da Polimerase , Ratos , Glutationa Peroxidase GPX1
15.
Mol Cell Biochem ; 371(1-2): 9-22, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22899171

RESUMO

Oxidative stress is a major component of harmful cascades activated in neurodegenerative disorders. Here, we tried to elucidate the possible neuroprotective effect of Salvigenin, a natural polyphenolic compound, on oxidative stress-induced apoptosis and autophagy in human neuroblastoma SH-SY5Y cells. We measured cell viability by MTT test and found that 25 µM is the best protective concentration of Salvigenin. GSH and SOD assays suggested that Salvigenin activates antioxidant factors. At the same time, measurement of ER stress-associated proteins including calpain and caspase-12 showed the ability of Salvigenin to decrease ER stress. We found that Salvigenin could decrease the apoptotic factors. Salvigenin inhibited H(2)O(2)-induced caspase-3 which is a hallmark of apoptosis in addition to reducing Bax\Bcl-2 ratio by 1.45 fold. Additionally, Salvigenin increased the levels of autophagic factors. Our results showed an increase in LC3-II/LC3-I ratio, Atg7, and Atg12 in the presence of 25 µM of Salvigenin by about 1.28, 1.25, and 1.54 folds, respectively, compared to H(2)O(2)-treated cells. So it seems that H(2)O(2) cytotoxicity mainly results from apoptosis. Besides, Salvigenin helps cells to survive by inhibiting apoptosis and enhancing autophagy that opens a new horizon for the future experiments.


Assuntos
Apoptose , Autofagia , Flavonas/farmacologia , Peróxido de Hidrogênio/farmacologia , Fármacos Neuroprotetores/farmacologia , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Relação Dose-Resposta a Droga , Humanos , Neuroblastoma/metabolismo , Estresse Oxidativo , Proteína X Associada a bcl-2/metabolismo
16.
Int J Breast Cancer ; 2012: 761917, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22482061

RESUMO

Epidermal growth factor (EGF) family of receptors is involved in cell growth and differentiation. The human EGF2 (HER2) lacks natural ligands, and correlation between HER2 levels and carcinogenesis makes the receptor an ideal candidate for targeted therapy in breast cancer. Trastuzumab is a humanized antibody applied against HER2-positive breast tumors in clinic. Metastatic tumors respond well to trastuzumab therapy for the first year, but development of antibody resistance helps the tumors to regrow allowing the disease to progress. Trastuzumab resistance is shaped via a range of intracellular signaling pathways that are interconnected and share in key effector molecules. Identification of a common node central to these resistance pathways could provide an ultimate solution for trastuzumab resistance in breast and other cancers.

17.
J Mol Neurosci ; 46(3): 654-65, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21969113

RESUMO

Optimized levels of glial cell line-derived neurotrophic factor (GDNF) are critical for protection of dopaminergic neurons against parkinsonian cell death. Recombinant lentiviruses harboring GDNF coding sequence were constructed and used to infect astrocytoma cell line 1321N1. The infected astrocytes overexpressed GDNF mRNA and secreted an average of 2.2 ng/mL recombinant protein as tested in both 2 and 16 weeks post-infection. Serial dilutions of GDNF-enriched conditioned medium from infected astrocytes added to growing neuroblastoma cell line SK-N-MC resulted in commensurate resistance against 6-OHDA toxicity. SK-N-MC cell survival rate rose from 51% in control group to 84% in the cells grown with astro-CM containing 453 pg secreted GDNF, an increase that was highly significant (P < 0.0001). However, larger volumes of the GDNF-enriched conditioned medium failed to improve cell survival and addition of volumes that contained 1,600 pg or more GDNF further reduced survival rate to below 70%. Changes in cell survival paralleled to changes in the percent of apoptotic cell morphologies. These data demonstrate the feasibility of using astrocytes as minipumps to stably oversecrete neurotrophic factors and further indicate that GDNF can be applied to neuroprotection studies in PD pending the optimization of its concentrations.


Assuntos
Astrócitos/patologia , Engenharia Genética/métodos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Neuroblastoma/patologia , Oxidopamina/toxicidade , Animais , Apoptose/efeitos dos fármacos , Astrocitoma/genética , Astrocitoma/patologia , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Linhagem Celular Tumoral , Citoproteção/genética , Estudos de Viabilidade , Vetores Genéticos/genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Simpatolíticos/toxicidade
18.
Iran J Reprod Med ; 10(5): 419-24, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25246906

RESUMO

BACKGROUND: Carbaryl is a carbamate insecticide widely used to control pests in agriculture and farm. Carbaryl adversely affect the reproductive endocrine systems in animals. OBJECTIVE: The aim of this study was to evaluate Carbaryl effects on the pituitary-gonad axis in rats. MATERIALS AND METHODS: In this experimental - analytical study, 60 adult male rats were divided into four equal groups: control, sham and experimental (1 and 2) groups that received 10 and 30 mg/kg Carbaryl via intraperitoneally injection. The sham group was subjected to intraperitoneally injection with olive oil while the control group did not receive any injection. Animals were sacrificed 35 days after the last treatment. Tissue sections were prepared from testes to investigate possible changes occurring in spermatogenic and Leydig cells. Blood samples were collected in which the levels of testosterone, luteinizing hormones (LH) and follicle stimulating hormone (FSH) were measured. RESULTS: The results showed significant reduction in testes weight (p=0.042) and seminiferous diameters (p<0.001) within the experimental groups compared with control group. Also, the number of germ cells, spermatocyts, spermatids and Leydig cells on the testes of the experimental groups was significantly decreased (p<0.001). Accordingly, significant decline in the testosterone levels (p<0.001) and increase in LH and FSH levels were observed (p<0.05). CONCLUSION: These results demonstrated that Carbaryl has capacity to exert adverse effects on fertility. Therefore, have to be taken to account in applying Carbaryl for any studies and or commercial use.

19.
Rejuvenation Res ; 14(2): 195-204, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21222532

RESUMO

The degeneration of dopaminergic neurons in the course of Parkinson disease is largely blamed on oxidative damage in the brain. This study examined the potency of glutathione peroxidase-1 (GPX-1) to protect dopaminergic neurons against toxicity induced by the parkinsonian neurotoxin 6-hydroxydopamine (6-OHDA). We generated pLV-GPX1, a recombinant lentivirus vector carrying the coding sequence for human GPX-1, into the SK-N-MC neuroblastoma cell line. The pLV-GPX1-infected neurons showed an over 3-fold increase in enzyme expression and a 2.6-fold increase in enzyme activity compared to the pLV-EGFP-infected control cells. In the pLV-GPX1-infected cells, we also detected significantly increased neuronal survival and resistance to 6-OHDA-mediated toxicity compared to our controls (75 ± 4% versus 51 ± 7%, p < 0.001). To maximize this protection, the neurons were treated with conditioned medium taken from growing primary astrocytes (astro-CM). We found the treated pLV-GPX1-infected neurons even more significantly resistant to 6-OHDA toxicity compared to their untreated counterparts (86 ± 5% versus 75 ± 4%, p < 0.001). Concomitant with increased neuroprotection, co-presence of overexpressed GPX-1 and astro-CM significantly increased glutathione (GSH) levels compared to when either of the two was present (p < 0.001). Further analysis showed nearly 2.7-fold reduction, in the presence of astro-CM, of hydrogen peroxide (H(2)O(2)) levels released from the pLV-GPX1-infected neurons compared to control groups (p < 0.001). Finally, regression analysis between H(2)O(2) levels and cell viability showed that co-presence of GPX-1 and astro-CM reduced 33% of cell death rate (p < 0.05). These data highlight the antioxidant properties of GPX-1 in protecting dopaminergic neurons and further emphasize the capacity of astrocytes in pumping growth-inducing factors that may synergize with GPX-1 to accelerate neuroprotection.


Assuntos
Astrócitos/metabolismo , Dopamina/metabolismo , Radicais Livres/metabolismo , Glutationa Peroxidase/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Astrócitos/citologia , Sobrevivência Celular , Células Cultivadas , Humanos , Hidroxidopaminas/toxicidade , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/metabolismo , Glutationa Peroxidase GPX1
20.
Biotechnol Lett ; 32(11): 1615-21, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20640481

RESUMO

We have developed an integrase-defective lentiviral (LV) vector in combination with a gene-targeting approach for gene therapy of ß-thalassemia. The ß-globin gene-targeting construct has two homologous stems including sequence upstream and downstream of the ß-globin gene, a ß-globin gene positioned between hygromycin and neomycin resistant genes and a herpes simplex virus type 1 thymidine kinase (HSVtk) suicide gene. Utilization of integrase-defective LV as a vector for the ß-globin gene increased the number of selected clones relative to non-viral methods. This method represents an important step toward the ultimate goal of a clinical gene therapy for ß-thalassemia.


Assuntos
Marcação de Genes/métodos , Vetores Genéticos , Lentivirus/genética , Globinas beta/genética , Antibacterianos/farmacologia , Cinamatos/farmacologia , Terapia Genética/métodos , Herpesvirus Humano 1/enzimologia , Higromicina B/análogos & derivados , Higromicina B/farmacologia , Integrases/deficiência , Neomicina/farmacologia , Recombinação Genética , Seleção Genética , Homologia de Sequência , Timidina Quinase/metabolismo , Talassemia beta/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...