Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Int J Mol Sci ; 24(9)2023 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-37175448

RESUMEN

Since aerobic glycolysis was first observed in tumors almost a century ago by Otto Warburg, the field of cancer cell metabolism has sparked the interest of scientists around the world as it might offer new avenues of treatment for malignant cells. Our current study claims the discovery of gnetin H (GH) as a novel glycolysis inhibitor that can decrease metabolic activity and lactic acid synthesis and displays a strong cytostatic effect in melanoma and glioblastoma cells. Compared to most of the other glycolysis inhibitors used in combination with the complex-1 mitochondrial inhibitor phenformin (Phen), GH more potently inhibited cell growth. RNA-Seq with the T98G glioblastoma cell line treated with GH showed more than an 80-fold reduction in thioredoxin interacting protein (TXNIP) expression, indicating that GH has a direct effect on regulating a key gene involved in the homeostasis of cellular glucose. GH in combination with phenformin also substantially enhances the levels of p-AMPK, a marker of metabolic catastrophe. These findings suggest that the concurrent use of the glycolytic inhibitor GH with a complex-1 mitochondrial inhibitor could be used as a powerful tool for inducing metabolic catastrophe in cancer cells and reducing their growth.


Asunto(s)
Antineoplásicos , Glioblastoma , Humanos , Fenformina , Glucólisis , Glucosa/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Línea Celular Tumoral
2.
Bioorg Med Chem ; 78: 117137, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36603398

RESUMEN

In cancer cells, glutaminolysis is the primary source of biosynthetic precursors. Recent efforts to develop amino acid analogues to inhibit glutamine metabolism in cancer have been extensive. Our lab recently discovered many L-γ-methyleneglutamic acid amides that were shown to be as efficacious as tamoxifen or olaparib in inhibiting the cell growth of MCF-7, SK-BR-3, and MDA-MB-231 breast cancer cells after 24 or 72 h of treatment. None of these compounds inhibited the cell growth of nonmalignant MCF-10A breast cells. These L-γ-methyleneglutamic acid amides hold promise as novel therapeutics for the treatment of multiple subtypes of breast cancer. Herein, we report our synthesis and evaluation of two series of tert-butyl ester and ethyl ester prodrugs of these L-γ-methyleneglutamic acid amides and the cyclic metabolite and its tert-butyl esters and ethyl esters on the three breast cancer cell lines MCF-7, SK-BR-3, and MDA-MB-231 and the nonmalignant MCF-10A breast cell line. These esters were found to suppress the growth of the breast cancer cells, but they were less potent compared to the L-γ-methyleneglutamic acid amides. Pharmacokinetic (PK) studies were carried out on the lead L-γ-methyleneglutamic acid amide to establish tissue-specific distribution and other PK parameters. Notably, this lead compound showed moderate exposure to the brain with a half-life of 0.74 h and good tissue distribution, such as in the kidney and liver. Therefore, the L-γ-methyleneglutamic acid amides were then tested on glioblastoma cell lines BNC3 and BNC6 and head and neck cancer cell lines HN30 and HN31. They were found to effectively suppress the growth of these cancer cell lines after 24 or 72 h of treatment in a concentration-dependent manner. These results suggest broad applications of the L-γ-methyleneglutamic acid amides in anticancer therapy.


Asunto(s)
Neoplasias de la Mama , Profármacos , Humanos , Femenino , Amidas/química , Profármacos/farmacología , Ésteres/farmacología , Ésteres/química , Aminoácidos , Neoplasias de la Mama/patología , Línea Celular Tumoral
3.
Eur J Med Chem ; 227: 113891, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34656042

RESUMEN

TumorSelect® is an anticancer technology that combines cytotoxics, nanotechnology, and knowledge of human physiology to develop innovative therapeutic interventions with minimal undesirable side effects commonly observed in conventional chemotherapy. Tumors have a voracious appetite for cholesterol which facilitates tumor growth and fuels their proliferation. We have transformed this need into a stealth delivery system to disguise and deliver anticancer drugs with the assistance of both the human body and the tumor cell. Several designer prodrugs are incorporated within pseudo-LDL nanoparticles, which carry them to tumor tissues, are taken up, internalized, transformed into active drugs, and inhibit cancer cell proliferation. Highly lipophilic prodrug conjugates of paclitaxel suitable for incorporation into the pseudo-LDL nanoparticles of the TumorSelect® delivery vehicle formulation were designed, synthesized, and evaluated in the panel of 24-h NCI-60 human tumor cell line screening to demonstrate the power of such an innovative approach. Taxane prodrugs, viz., ART-207 was synthesized by tethering paclitaxel to lipid moiety with the aid of a racemic solketal as a linker in cost-effective, simple, and straightforward synthetic transformations. In addition to the typical 24-h NCI screening protocol, these compounds were assessed for growth inhibition or killing of ovarian cell lines for 48 and 72h-time intervals and identified the long-lasting effectiveness of these lipophilic prodrugs. All possible, enantiomerically pure isomers of ART-207 were also synthesized, and cytotoxicities were biosimilar to racemic ART-207, suggesting that enantiopurity of linker has a negligible effect on cell proliferation. To substantiate further, ART-207 was evaluated for its in vivo tumor reduction efficacy by studying the xenograft model of ovarian cancer grown in SCID mice. Reduced weight loss (a measure of toxicity) in the ART-207 group was observed, even though it was dosed at 2.5x the paclitaxel equivalent of Abraxane®. As a result, our delineated approach is anticipated to improve patient quality of life, patient retention in treatment regimes, post-treatment rapid recovery, and overall patient compliance without compromising the efficacy of the cytotoxic promiscuous natural products.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Productos Biológicos/farmacología , Paclitaxel/farmacología , Profármacos/farmacología , Animales , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/química , Productos Biológicos/síntesis química , Productos Biológicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Congénicos , Ratones Endogámicos NOD , Ratones SCID , Conformación Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Paclitaxel/síntesis química , Paclitaxel/química , Profármacos/síntesis química , Profármacos/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
4.
Sci Rep ; 8(1): 1720, 2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29379063

RESUMEN

Human mesenchymal stem/stromal cells (hMSCs) provide support for cancer progression, partly through their secretome that includes extracellular vesicles (EVs). Based on deep-sequencing of small RNA from EVs of MSCs, we now report the characterization of novel small RNA, named n-miR-G665, which exhibits typical properties of miRNAs. n-miR-G665 sequence is conserved and expressed in most cell types. Knockdown studies using anti-agomirs and shRNA studies demonstrated that n-miR-G665 plays an important role in cell proliferation. Functional assays to reveal the targets of n-miR-G665 showed that polycomb protein Suz12 is regulated by n-miR-G665, which in turn regulates the expression of n-miR-G665 through feedback loop mechanism. These data shed light on a previously unknown novel feedback regulatory mechanism for controlling Suz12 expression regulated by previously not described miRNA, which may highlight a new therapeutic approach to control the polycomb repressor complex 2 activity in cancers.


Asunto(s)
Regulación de la Expresión Génica , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/fisiología , MicroARNs/metabolismo , Complejo Represivo Polycomb 2/biosíntesis , Línea Celular , Proliferación Celular , Vesículas Extracelulares/química , Técnicas de Silenciamiento del Gen , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , MicroARNs/genética , MicroARNs/aislamiento & purificación , Proteínas de Neoplasias , Factores de Transcripción
5.
Oncotarget ; 8(66): 109861-109876, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29299114

RESUMEN

Mesenchymal stromal cells (hMSCs) have been used to understand the stromal cell properties in solid tumors because of their ablity to differentiate into most cell types. We investigated the role of EVs from hMSCs (hMSC-EVs) in breast cancer metastasis using MDA-MB-231 parental cell line and organotropic sub-lines. We demonstrated that hMSC-EVs significantly suppressed the metastatic potential of the parental cell line when compared to their organotropic sublines. hMSC-EVs induce dormancy in the parental cell line but not in their organotropic sub-lines and miR-205 and miR-31 from EV cargo played a role. Further, Ubiquitin Conjugating Enzyme E2 N (UBE2N/Ubc13) - metastasis-regulating gene, is a target of these miRNAs and silencing of UBE2N/Ubc13 expression significantly suppressed migration, invasion, and proliferation of breast cancer cells. To summarize, hMSC-EVs support primary breast tumor progression but suppress the metastasis of breast cancer cells that are not organ-committed through the UBE2N/Ubc13 pathway and play a role in premetastic niche formation.

6.
Front Biosci (Elite Ed) ; 9(1): 162-173, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27814597

RESUMEN

It is well recognized that one of the major drawbacks of using traditional two dimensional cultures to model the living systems is inaccurately reflecting the physiological manner in which modulators, nutrients, oxygen, and metabolites are applied and removed. Moreover, the two dimensional culture system poorly reflects how different cell types interact with each other in the same microenvironment. Since the first global development of three dimensional (3D) cell culture techniques in the late 1960s, this last decade has seen an explosion of studies to promote 3D models in the fields of regenerative medicine and cancer. The recent surge of interest in 3D cell culture in cancer research is attributable to the interest in developing closer to real life models. The ability to include various cell types and extracellular components reflect more the physiological conditions of tumor microenvironment. In this short review, we will discuss different approaches of 3D culture system models and techniques with a focus on the 3D interactions of cancer cells with stromal cells in the goal to reevaluate old and develop new therapeutics.


Asunto(s)
Técnicas de Cultivo de Célula , Neoplasias/patología , Evaluación Preclínica de Medicamentos/métodos , Humanos , Dispositivos Laboratorio en un Chip , Neoplasias de Tejido Conjuntivo/patología , Neoplasias Glandulares y Epiteliales/patología , Proyectos de Investigación , Esferoides Celulares , Andamios del Tejido , Células Tumorales Cultivadas , Microambiente Tumoral
7.
Methods Mol Biol ; 1416: 159-69, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27236671

RESUMEN

Mesenchymal stem/stromal cells (MSCs) have been extensively investigated for their potential to regenerate tissue, to modulate the immune system, and their wound healing properties in over 350 clinical trials worldwide. MSCs from various tissues such as adipose, bone, and others are currently being studied in clinical trials in indications for ischemic, inflammatory, autoimmune, and degenerative disorders. As a result, numerous isolation protocols have been published. This chapter provides a simple protocol whereby a total of 80-100 million human MSCs, with an average viability greater than 90 %, can be produced from a relatively small (1-3 mL) bone marrow aspirate in 14-20 days using double stack culture chambers. MSCs were originally referred to as fibroblastoid colony forming cells because one of their characteristic features is adherence to tissue culture plastic and generation of colonies when plated at low densities. The efficiency with which they form colonies still remains an important assay for the quality of cell preparations. To assess the quality of cell preparations, two different colony forming unit (CFU) assays are also provided.


Asunto(s)
Ensayo de Unidades Formadoras de Colonias/métodos , Células Madre Mesenquimatosas/citología , Recuento de Células , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Humanos
8.
Stem Cells Int ; 2016: 1073140, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26649044

RESUMEN

Stem cells are proposed to continuously secrete trophic factors that potentially serve as mediators of autocrine and paracrine activities, associated with reprogramming of the tumor microenvironment, tissue regeneration, and repair. Hitherto, significant efforts have been made to understand the level of underlying paracrine activities influenced by stem cell secreted trophic factors, as little is known about these interactions. Recent findings, however, elucidate this role by reporting the effects of stem cell derived extracellular vesicles (EVs) that mimic the phenotypes of the cells from which they originate. Exchange of genetic information utilizing persistent bidirectional communication mediated by stem cell-EVs could regulate stemness, self-renewal, and differentiation in stem cells and their subpopulations. This review therefore discusses stem cell-EVs as evolving communication factors in stem cell biology, focusing on how they regulate cell fates by inducing persistent and prolonged genetic reprogramming of resident cells in a paracrine fashion. In addition, we address the role of stem cell-secreted vesicles in shaping the tumor microenvironment and immunomodulation and in their ability to stimulate endogenous repair processes during tissue damage. Collectively, these functions ensure an enormous potential for future therapies.

9.
Int J Cancer ; 138(1): 14-21, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25559768

RESUMEN

In recent years, the knowledge about the control of tumor microenvironment has increased and emerged as an important player in tumorigenesis. The role of normal stromal cells in the tumor initiation and progression has brought our vision in to the forefront of cell-to-cell communication. In this review, we focus on the mechanism of communication between stromal and tumor cells, which is based on the exchange of extracellular vesicles (EVs). We describe several, evergrowing, pieces of evidence that EVs transfer messages through their miRNA, lipid, protein and nucleic acid contents. A better understanding of this sophisticated method of communication between normal cancer cells may lead to developing novel approaches for personalized diagnostics and therapeutics.


Asunto(s)
Vesículas Extracelulares/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral , Animales , Transporte Biológico , Comunicación Celular , Vesículas Extracelulares/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Lípidos , Células Madre Mesenquimatosas/metabolismo , MicroARNs , Metástasis de la Neoplasia , Neoplasias/genética , Proteínas , Transducción de Señal
10.
Curr Stem Cell Res Ther ; 11(2): 141-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26423301

RESUMEN

Mesenchymal Stem/stromal cell (MSCs) transplantation procedures have been used since the 1960's to treat leukemia and other diseases, but due to the risks involved only patients with life threatening illnesses were typically subjected to the transplantation procedure until the last decade. Recent advancements in transplantation techniques have made it more feasible to use it for non-life-threatening diseases. However, the potential uses for stem cells are still limited by their rarity, and, in the case of allogeneic transplants, graft-vs.-host complications. An evolving alternative to conventional stem cell therapies is induced pluripotent stem-cell derived mesenchymal stem/stromal cells (iPSC- MSCs), which have a multi-lineage potential comparable to conventionally acquired MSCs with the added benefit of being less immunoreactive. However there are still many hurdles left to be overcome before they can be used regularly for personalized therapies. This review will focus on recent advancements that have been made regarding the role MSCs play in tumor development and the potential uses iPSC-MSCs may have in future cancer treatment.


Asunto(s)
Células Madre Pluripotentes Inducidas/trasplante , Trasplante de Células Madre Mesenquimatosas/tendencias , Células Madre Mesenquimatosas , Medicina de Precisión , Humanos
11.
Oncotarget ; 6(7): 4953-67, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25669974

RESUMEN

Human mesenchymal stem/stromal cells (hMSCs) have been shown to support breast cancer cell proliferation and metastasis, partly through their secretome. hMSCs have a remarkable ability to survive for long periods under stress, and their secretome is tumor supportive. In this study, we have characterized the cargo of extracellular vesicular (EV) fraction (that is in the size range of 40-150nm) of serum deprived hMSCs (SD-MSCs). Next Generation Sequencing assays were used to identify small RNA secreted in the EVs, which indicated presence of tumor supportive miRNA. Further assays demonstrated the role of miRNA-21 and 34a as tumor supportive miRNAs. Next, proteomic assays revealed the presence of ≈150 different proteins, most of which are known tumor supportive factors such as PDGFR-ß, TIMP-1, and TIMP-2. Lipidomic assays verified presence of bioactive lipids such as sphingomyelin. Furthermore, metabolite assays identified the presence of lactic acid and glutamic acid in EVs. The co-injection xenograft assays using MCF-7 breast cancer cells demonstrated the tumor supportive function of these EVs. To our knowledge this is the first comprehensive -omics based study that characterized the complex cargo of extracellular vesicles secreted by hMSCs and their role in supporting breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Vesículas Extracelulares/patología , Femenino , Xenoinjertos , Humanos , Metabolismo de los Lípidos , Células MCF-7 , Ratones , Ratones Desnudos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Proteoma/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Med ; 3(4): 796-811, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24802970

RESUMEN

Overall prognosis for osteosarcoma (OS) is poor despite aggressive treatment options. Limited access to primary tumors, technical challenges in processing OS tissues, and the lack of well-characterized primary cell cultures has hindered our ability to fully understand the properties of OS tumor initiation and progression. In this study, we have isolated and characterized cell cultures derived from four central high-grade human OS samples. Furthermore, we used the cell cultures to study the role of CD49f in OS progression. Recent studies have implicated CD49f in stemness and multipotency of both cancer stem cells and mesenchymal stem cells. Therefore, we investigated the role of CD49f in osteosarcomagenesis. First, single cell suspensions of tumor biopsies were subcultured and characterized for cell surface marker expression. Next, we characterized the growth and differentiation properties, sensitivity to chemotherapy drugs, and anchorage-independent growth. Xenograft assays showed that cell populations expressing CD49f(hi) /CD90(lo) cell phenotype produced an aggressive tumor. Multiple lines of evidence demonstrated that inhibiting CD49f decreased the tumor-forming ability. Furthermore, the CD49f(hi) /CD90(lo) cell population is generating more aggressive OS tumor growth and indicating this cell surface marker could be a potential candidate for the isolation of an aggressive cell type in OSs.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/metabolismo , Integrina alfa6/metabolismo , Osteosarcoma/metabolismo , Adolescente , Adulto , Animales , Antineoplásicos/farmacología , Neoplasias Óseas/patología , Movimiento Celular , Proliferación Celular , Niño , Cisplatino/farmacología , Progresión de la Enfermedad , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Femenino , Humanos , Masculino , Ratones Desnudos , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Osteosarcoma/patología , Cultivo Primario de Células , Células Tumorales Cultivadas
13.
Mol Pharm ; 11(2): 417-27, 2014 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-24380633

RESUMEN

This study examines the antitumor potential of curcumin and C6 ceramide (C6) against osteosarcoma (OS) cell lines when both are encapsulated in the bilayer of liposomal nanoparticles. Three liposomal formulations were prepared: curcumin liposomes, C6 liposomes and C6-curcumin liposomes. Curcumin in combination with C6 showed 1.5 times enhanced cytotoxic effect in the case of MG-63 and KHOS OS cell lines, in comparison with curcumin liposomes alone. Importantly, C6-curcumin liposomes were found to be less toxic on untransformed primary human cells (human mesenchymal stem cells) in comparison to OS cell lines. In addition, cell cycle assays on a KHOS cell line after treatment revealed that curcumin only liposomes induced G2/M arrest by upregulation of cyclin B1, while C6 only liposomes induced G1 arrest by downregulation of cyclin D1. C6-curcumin liposomes induced G2/M arrest and showed a combined effect in the expression levels of cyclin D1 and cyclin B1. The efficiency of the preparations was tested in vivo using a human osteosarcoma xenograft assay. Using pegylated liposomes to increase the plasma half-life and tagging with folate (FA) for targeted delivery in vivo, a significant reduction in tumor size was observed with C6-curcumin-FA liposomes. The encapsulation of two water insoluble drugs, curcumin and C6, in the lipid bilayer of liposomes enhances the cytotoxic effect and validates the potential of combined drug therapy.


Asunto(s)
Ceramidas/administración & dosificación , Curcumina/administración & dosificación , Liposomas/química , Nanopartículas/química , Osteosarcoma/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ceramidas/química , Ceramidas/farmacología , Curcumina/química , Curcumina/farmacología , Sistemas de Liberación de Medicamentos , Citometría de Flujo , Humanos , Concentración 50 Inhibidora , Ratones , Microscopía Electrónica de Transmisión , Estructura Molecular , Nanopartículas/uso terapéutico
14.
Nanomedicine ; 8(4): 440-51, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21839055

RESUMEN

The delivery of curcumin, a broad-spectrum anticancer drug, has been explored in the form of liposomal nanoparticles to treat osteosarcoma (OS). Curcumin is water insoluble and an effective delivery route is through encapsulation in cyclodextrins followed by a second encapsulation in liposomes. Liposomal curcumin's potential was evaluated against cancer models of mesenchymal (OS) and epithelial origin (breast cancer). The resulting 2-Hydroxypropyl-γ-cyclodextrin/curcumin - liposome complex shows promising anticancer potential both in vitro and in vivo against KHOS OS cell line and MCF-7 breast cancer cell line. An interesting aspect is that liposomal curcumin initiates the caspase cascade that leads to apoptotic cell death in vitro in comparison with DMSO-curcumin induced autophagic cell death. In addition, the efficiency of the liposomal curcumin formulation was confirmed in vivo using a xenograft OS model. Curcumin-loaded γ-cyclodextrin liposomes indicate significant potential as delivery vehicles for the treatment of cancers of different tissue origin. FROM THE CLINICAL EDITOR: Curcumin-loaded γ-cyclodextrin liposomes were demonstrated in vitro to have significant potential as delivery vehicles for the treatment of cancers of mesenchymal and epithelial origin. Differences between mechanisms of cell death were also evaluated.


Asunto(s)
Curcumina/farmacología , Osteosarcoma/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , gamma-Ciclodextrinas/farmacología , Animales , Antineoplásicos , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/ultraestructura , Caspasas/metabolismo , Línea Celular Tumoral , Curcumina/química , Femenino , Humanos , Liposomas , Ratones , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Osteosarcoma/ultraestructura , Trasplante Heterólogo , gamma-Ciclodextrinas/química
15.
Int J Biochem Cell Biol ; 43(11): 1563-72, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21794839

RESUMEN

Recently we demonstrated that the miRNA regulate human mesenchymal stem cells (hMSCs) differentiation. To determine the role of the miRNA pathway in hMSCs proliferation, Drosha and Dicer knockdown hMSCs were generated using a lentiviral based tetracycline inducible shRNA. hMSCs with reduced Drosha expression had a significantly reduced proliferation rate, while hMSCs with reduced Dicer expression displayed a proliferation rate similar to untransduced cells. Cell cycle analysis identified that unlike Dicer knockdown, Drosha knockdown hMSCs contained an increased number of G1 phase cells, with a reduced level of cells in S phase, compared to controls. ELISAs of hMSCs revealed decreased levels of pRB and stable levels of total RB with Drosha knockdown. Two key regulators of the G1/S phase transition, cyclin dependent kinase inhibitor 2A (p16) and cyclin dependent kinase inhibitor 2B (p15), were increased in Drosha knockdown cells but not in Dicer knockdown. Transcripts of 28S and 18S rRNA were significantly reduced in Drosha knockdown hMSCs, with no change in rRNA levels in Dicer knockdown hMSCs. 45S pre-rRNA transcripts were not significantly different in either knockdown model. The above results indicate that Drosha modifies hMSCs proliferation through a miRNA independent mechanism, potentially by regulating rRNA processing.


Asunto(s)
Ciclo Celular/genética , ARN Helicasas DEAD-box/deficiencia , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , ARN Ribosómico/metabolismo , Ribonucleasa III/deficiencia , Proliferación Celular , Células Cultivadas , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , ARN Helicasas DEAD-box/genética , Silenciador del Gen , Vectores Genéticos , Humanos , Lentivirus , Células Madre Mesenquimatosas/citología , MicroARNs/genética , ARN Mensajero/genética , ARN Ribosómico/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III/genética , Transducción Genética , Regulación hacia Arriba
16.
Methods Mol Biol ; 698: 11-21, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21431507

RESUMEN

In recent years, human mesenchymal stem cells (multipotential stromal cells) from bone marrow (hMSCs) have attracted enormous attention owing to their broad therapeutic potential. One of the problems in the overall therapeutic use of hMSCs has been the significant variability in the culture conditions used for their isolation and expansion. Since the seminal publications by Friedenstein and colleagues, the isolation and expansion of mesenchymal stromal cells (MSCs) from bone marrow have been of interest to several laboratories. As a result, numerous isolation protocols have been published. This chapter provides a simple protocol whereby a total of 80-100 million human MSCs, with an average viability greater than 90%, can be produced from a relatively small (1-3 mL) bone marrow aspirate in 14-20 days using double stacks culture chambers. MSCs were originally referred to as fibroblastoid colony forming cells because one of their characteristic features is adherence to tissue culture plastic and generation of colonies when plated at low densities. The efficiency with which they form colonies still remains an important assay for the quality of cell preparations. To assess the quality of cell preparations, two different colony forming unit (CFU) assays are also provided.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Ensayo de Unidades Formadoras de Colonias , Fibroblastos/citología , Humanos , Análisis de la Célula Individual
17.
Carcinogenesis ; 32(7): 964-72, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21317300

RESUMEN

Recent studies have implicated multipotential mesenchymal stem cells (MSCs) as an aid to breast cancer cell proliferation and metastasis, partly as a result of the MSCs secretome. As the tumor gets beyond 2 mm in diameter, the stromal cells could undergo starvation due to the lack of sufficient nutrients in solid tumor microenvironment. In this study, we investigated the survival mechanisms used by stressed stromal cells in breast cancers. We used serum-deprived mesenchymal stem cells (SD-MSCs) and MCF-7 breast cancer cells as model system with a hypothesis that stromal cells in the nutrient-deprived core utilize survival mechanisms for supporting surrounding cells. We tested this hypothesis using in vivo tumor xenografts in immunodeficient mice, which indicated that SD-MSCs supported MCF-7 tumor growth by protection from apoptosis. Histochemical assays showed that SD-MSCs-injected tumors exhibited higher cellularity, decreased apoptosis and decreased differentiation. Beclin-1 staining indicated autophagic areas surrounded by actively proliferating cells. Furthermore, in vitro studies demonstrate that SD-MSCs survive using autophagy and secrete paracrine factors that support tumor cells following nutrient/serum deprivation. Western blot and immunocytochemistry analysis of SD-MSCs demonstrated upregulation and perinuclear relocation of autophagy key regulators such as beclin-1, ATG10, ATG12, MAP-LC3 and lysosomes. Electron microscopic analysis detected a time-dependent increase in autophagosome formation and HDAC6 activity assays indicated the upregulation of autophagy. Taken together, these data suggest that under nutrient-deprived conditions that can occur in solid tumors, stromal cells utilize autophagy for survival and also secrete anti-apoptotic factors that can facilitate solid tumor survival and growth.


Asunto(s)
Autofagia , Células Madre Mesenquimatosas/inmunología , Neoplasias/patología , Células del Estroma/patología , Animales , Apoptosis/inmunología , Línea Celular Tumoral , Supervivencia Celular/inmunología , Células Cultivadas , Medio de Cultivo Libre de Suero , Femenino , Humanos , Ratones , Ratones SCID , Neoplasias/inmunología
18.
J Bone Joint Surg Am ; 92(5): 1231-40, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20439670

RESUMEN

BACKGROUND: Extra-axial chordomas are rare low-grade malignant tumors thought to arise from notochordal remnants in the extra-axial skeleton. Few studies have been done on this neoplasm because of its rarity. In addition, there is a lack of a good in vitro model on which to perform more characterization. METHODS: We describe a twenty-eight-year-old man with a mass in the right scapula. Cytomorphology and immunohistochemistry, including brachyury staining, were used to formulate the final diagnosis. A fragment of the tumor was placed in culture, and cells obtained were injected subcutaneously in an immunocompromised mouse. From the tumor developed in mice, a cell line has been derived and characterized by fluorescence-activated cell-sorting analysis, karyotyping, clonogenicity, and cell and tumor growth curves. RESULTS: Cytomorphology on the tumor showed nests of round cells with vacuoles and also physaliferous-like cells with uniform nuclei. Immunochemistry revealed a tumor positive for vimentin, moderately positive for S-100 and cytokeratin AE1/AE3, weakly positive for epithelial membrane antigen, and negative for p63 and cytokeratin (CK)-7. Further analysis revealed the tumor was diffusely and strongly positive for brachyury. The cell line derived from the tumor showed rapid doubling-time, a strong expression of mesenchymal cell surface markers, a karyotype of diploid or hypotetraploid clones with numerous chromosomal aberrations, and the ability to form colonies without attachment and to form tumors in immunocompromised mice. CONCLUSIONS: The diagnosis of the extra-axial chordoma is difficult but can be resolved by the detection of a strong brachyury expression. In addition, the derivation of a human extra-axial chordoma cell line could be a useful tool for the basic research of this rare neoplasm.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Cordoma/cirugía , Neoplasias de los Músculos/cirugía , Escápula/patología , Adulto , Animales , Cordoma/diagnóstico , Humanos , Masculino , Ratones , Neoplasias de los Músculos/diagnóstico , Trasplante de Neoplasias , Neoplasias Experimentales
19.
Breast Cancer Res Treat ; 121(2): 293-300, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19597705

RESUMEN

Adult human mesenchymal stem cells (hMSCs) have been shown to home to sites of breast cancer and integrate into the tumor stroma. We demonstrate here the effect of hMSCs on primary breast tumor growth and the progression of these tumors to hormone independence. Co-injection of bone marrow-derived hMSCs enhances primary tumor growth of the estrogen receptor-positive, hormone-dependent breast carcinoma cell line MCF-7 in the presence or absence of estrogen in SCID/beige mice. We also show hormone-independent growth of MCF-7 cells when co-injected with hMSCs. These effects were found in conjunction with increased immunohistochemical staining of the progesterone receptor in the MCF-7/hMSC tumors as compared to MCF-7 control tumors. This increase in PgR expression indicates a link between MCF-7 cells and MSCs through ER-mediated signaling. Taken together, our data reveal the relationship between tumor microenvironment and tumor growth and the progression to hormone independence. This tumor stroma-cell interaction may provide a novel target for the treatment of estrogen receptor-positive, hormone-independent, and endocrine-resistant breast carcinoma.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Células Madre Mesenquimatosas/patología , Animales , Antineoplásicos Hormonales/farmacología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Inmunohistoquímica , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Proc Natl Acad Sci U S A ; 105(47): 18372-7, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19011087

RESUMEN

We observed that microRNAs (miRNAs) that regulate differentiation in a variety of simpler systems also regulate differentiation of human multipotent stromal cells (hMSCs) from bone marrow. Differentiation of hMSCs into osteoblasts and adipocytes was inhibited by using lentiviruses expressing shRNAs to decrease expression of Dicer and Drosha, two enzymes that process early transcripts to miRNA. Expression analysis of miRNAs during hMSC differentiation identified 19 miRNAs that were up-regulated during osteogenic differentiation and 20 during adipogenic differentiation, 11 of which were commonly up-regulated in both osteogenic and adipogenic differentiation. In silico models predicted that five of the up-regulated miRNAs targeted leukemia inhibitory factor (LIF) expression. The prediction was confirmed for two of the miRNAs, hsa-mir 199a and hsa-mir346, in that over-expression of the miRNAs decreased LIF secretion by hMSCs. The results demonstrate that differentiation of hMSCs is regulated by miRNAs and that several of these miRNAs target LIF.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/fisiología , Factor Inhibidor de Leucemia/metabolismo , MicroARNs/fisiología , Células Madre Multipotentes/citología , Células del Estroma/citología , Tejido Adiposo/citología , Técnicas de Silenciamiento del Gen , Humanos , ARN Mensajero/genética , Ribonucleasa III/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA