Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Death Dis ; 14(8): 579, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37653021

RESUMEN

Eukaryotic initiation factor 5A2 (eIF5A2) is overexpressed in many types of cancer, and spermidine-mediated eIF5A hypusination (eIF5Ahpu) appears to be essential to most of eIF5A's biological functions, including its important role in regulating cancer cell proliferation, epithelial-mesenchymal transition (EMT), and cancer stem cell (CSC) properties as well as immune cell functions. Here we investigated the role of eIF5Ahpu in the growth of oral squamous cell carcinoma cells (OSCCs) and OSCC-induced polarization of M2-like tumor-associated macrophages (TAMs). TCGA dataset analysis revealed an overall upregulation in the mRNA expression of eIF5A2 and several key enzymes involved in polyamine (PA) metabolism in HNSCC, which was confirmed by Western blot and IHC studies. Blocking eIF5Ahpu by GC-7 but not the upstream key enzyme activities of PA metabolism, remarkably inhibited cell proliferation and the expression of EMT- and CSC-related genes in OSCC cells. In addition, blocking eIF5Ahpu robustly inhibited OSCC-induced M2-like TAM polarization in vitro. More Importantly, blocking eIF5Ahpu dramatically retarded tumor growth and infiltration/polarization of M2-like TAM in a syngeneic orthotopic murine tongue SCC model. Thus, eIF5Ahpu plays dual functions in regulating tumor cell growth and polarization of M2-TAMs in OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Boca , Neoplasias de la Lengua , Animales , Ratones , Neoplasias de la Boca/genética , Factores de Iniciación de Péptidos/genética , Neoplasias de la Lengua/genética , Macrófagos Asociados a Tumores , Humanos , Factor 5A Eucariótico de Iniciación de Traducción
2.
Stem Cell Res Ther ; 13(1): 263, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35725660

RESUMEN

BACKGROUND: Peripheral nerve injuries (PNIs) remain one of the great clinical challenges because of their considerable long-term disability potential. Postnatal neural crest-derived multipotent stem cells, including gingiva-derived mesenchymal stem cells (GMSCs), represent a promising source of seed cells for tissue engineering and regenerative therapy of various disorders, including PNIs. Here, we generated GMSC-repopulated nerve protectors and evaluated their therapeutic effects in a crush injury model of rat sciatic nerves. METHODS: GMSCs were mixed in methacrylated collagen and cultured for 48 h, allowing the conversion of GMSCs into Schwann-like cells (GiSCs). The phenotype of GiSCs was verified by fluorescence studies on the expression of Schwann cell markers. GMSCs encapsulated in the methacrylated 3D-collagen hydrogel were co-cultured with THP-1-derived macrophages, and the secretion of anti-inflammatory cytokine IL-10 or inflammatory cytokines TNF-α and IL-1ß in the supernatant was determined by ELISA. In addition, GMSCs mixed in the methacrylated collagen were filled into a nerve protector made from the decellularized small intestine submucosal extracellular matrix (SIS-ECM) and cultured for 24 h, allowing the generation of functionalized nerve protectors repopulated with GiSCs. We implanted the nerve protector to wrap the injury site of rat sciatic nerves and performed functional and histological assessments 4 weeks post-surgery. RESULTS: GMSCs encapsulated in the methacrylated 3D-collagen hydrogel were directly converted into Schwann-like cells (GiSCs) characterized by the expression of S-100ß, p75NTR, BDNF, and GDNF. In vitro, co-culture of GMSCs encapsulated in the 3D-collagen hydrogel with macrophages remarkably increased the secretion of IL-10, an anti-inflammatory cytokine characteristic of pro-regenerative (M2) macrophages, but robustly reduced LPS-stimulated secretion of TNF-1α and IL-1ß, two cytokines characteristic of pro-inflammatory (M1) macrophages. In addition, our results indicate that implantation of functionalized nerve protectors repopulated with GiSCs significantly accelerated functional recovery and axonal regeneration of crush-injured rat sciatic nerves accompanied by increased infiltration of pro-regenerative (M2) macrophages while a decreased infiltration of pro-inflammatory (M1) macrophages. CONCLUSIONS: Collectively, these findings suggest that Schwann-like cells converted from GMSCs represent a promising source of supportive cells for regenerative therapy of PNI through their dual functions, neurotrophic effects, and immunomodulation of pro-inflammatory (M1)/pro-regenerative (M2) macrophages.


Asunto(s)
Células Madre Mesenquimatosas , Traumatismos de los Nervios Periféricos , Animales , Colágeno/metabolismo , Humanos , Hidrogeles , Interleucina-10/metabolismo , Células Madre Mesenquimatosas/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de los Nervios Periféricos/patología , Traumatismos de los Nervios Periféricos/terapia , Ratas , Células de Schwann/metabolismo , Nervio Ciático/patología
3.
Materials (Basel) ; 15(10)2022 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-35629668

RESUMEN

The hybrid manufacturing method of laser cladding deposition (LCD) additive manufacturing and electrochemical machining (ECM) is a promising approach to advanced manufacturing technology for difficult machined materials. The anisotropic electrochemical performance of LCD-produced Ti6Al4V alloy was studied in 15 wt.% NaCl solution by polarization curve measurements and ECM tests. The horizontal-plane (X0Y plane) exhibits a more stable passive film in both static electrolyte and low current density ECM processes than the vertical-plane (X0Z plane). Additionally, the horizontal-plane exhibits a higher material removal rate and more consistent dissolved surface roughness in comparison with the vertical-plane during the high current density ECM process. The microstructure of the LCD-produced Ti6Al4V alloy on the horizontal-plane consisted of equiaxed-like prior-ß grains and slightly finer α-laths but was composed by columnar prior-ß grains and coarser α-laths on the vertical-plane. These differences in the microstructural characteristics produce the distinctions observed in the electrochemical dissolution behavior and electrochemical machinability on the horizontal- and vertical-planes.

4.
NPJ Regen Med ; 6(1): 59, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34593823

RESUMEN

Achieving a satisfactory functional recovery after severe peripheral nerve injuries (PNI) remains one of the major clinical challenges despite advances in microsurgical techniques. Nerve autografting is currently the gold standard for the treatment of PNI, but there exist several major limitations. Accumulating evidence has shown that various types of nerve guidance conduits (NGCs) combined with post-natal stem cells as the supportive cells may represent a promising alternative to nerve autografts. In this study, gingiva-derived mesenchymal stem cells (GMSCs) under 3D-culture in soft collagen hydrogel showed significantly increased expression of a panel of genes related to development/differentiation of neural crest stem-like cells (NCSC) and/or Schwann cell precursor-like (SCP) cells and associated with NOTCH3 signaling pathway activation as compared to their 2D-cultured counterparts. The upregulation of NCSC-related genes induced by 3D-collagen hydrogel was abrogated by the presence of a specific NOTCH inhibitor. Further study showed that GMSCs encapsulated in 3D-collagen hydrogel were capable of transmigrating into multilayered extracellular matrix (ECM) wall of natural NGCs and integrating well with the aligned matrix structure, thus leading to biofabrication of functionalized NGCs. In vivo, implantation of functionalized NGCs laden with GMSC-derived NCSC/SCP-like cells (designated as GiSCs), significantly improved the functional recovery and axonal regeneration in the segmental facial nerve defect model in rats. Together, our study has identified an approach for rapid biofabrication of functionalized NGCs through harnessing 3D collagen hydrogel-directed conversion of GMSCs into GiSCs.

5.
Cell Death Dis ; 11(5): 338, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32382005

RESUMEN

Ameloblastoma (AM) is a benign but locally aggressive tumor with high recurrences. Currently, underlying pathophysiology remains elusive, and radical surgery remains the most definitive treatment with severe morbidities. We have recently reported that AM harbors a subpopulation of tumor epithelial stem-like cells (AM-EpiSCs). Herein, we explored whether LGR5+ epithelial cells in AM possess stem-like cell properties and their potential contribution to pathogenesis and recurrence of AM. We found that LGR5 and stem cell-related genes were co-expressed in a subpopulation of AM epithelial cells both in vivo and in vitro, which were enriched under 3D-spheroid culture. As compared to LGR5- counterparts, LGR5+ AM epithelial cells showed increased expression of various EMT- and stemness-related genes, and functionally, exhibited increased capacity to form 3D-spheroids and generate human tumor 3D organoids, which recapitulated the histopathologic features of distinct subtypes of solid AM, thus, contributing a useful human tumor platform for targeted therapeutic screening. Treatment with a selective BRAFV600E inhibitor, vemurafenib, unexpectedly enriched the subpopulation of LGR5+ AM-EpiSCs in tumor 3D organoids, which may have explained therapeutic resistances and recurrences. These findings suggest that LGR5+ AM-EpiSCs play a pivotal role in pathogenesis and progression of AM and targeted inhibition of both BRAF and LGR5 potentially serves a novel nonsurgical adjuvant therapeutic approach for this aggressively benign jaw tumor.


Asunto(s)
Ameloblastoma/metabolismo , Ameloblastoma/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Madre Neoplásicas/patología , Organoides/patología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Autorrenovación de las Células , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Masculino , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Trombospondinas/metabolismo
6.
Tissue Eng Part A ; 25(11-12): 887-900, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30311853

RESUMEN

IMPACT STATEMENT: Peripheral nerve injuries (PNIs) are common and debilitating, usually resulting in considerable long-term disability and remaining an unmet clinical need. Even though the combination of mesenchymal stem cells (MSCs) and the state-of-the-art tissue engineering technologies has shown promising therapeutic potentials for PNI, there is still not a single licensed stem cell-based product for peripheral nerve repair/regeneration. Emerging evidence indicates that MSC-derived extracellular vesicles (EVs) are comparably effective as MSCs in the therapy of a variety of disease models or pathological conditions. This report shows that local delivery of gingiva-derived mesenchymal stem cell (GMSC)-derived EVs could obviously promote axonal regeneration and functional recovery of injured mice sciatic nerves. Importantly, the findings suggest that GMSC-derived EVs promoted the expression of Schwann cell dedifferentiation/repair phenotype-related genes in vitro, particularly c-JUN, a key transcription factor that drives the activation of repair phenotype of Schwann cells during PNI and regeneration.


Asunto(s)
Vesículas Extracelulares/trasplante , Encía/metabolismo , Células Madre Mesenquimatosas/metabolismo , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos , Células de Schwann/metabolismo , Nervio Ciático , Animales , Movimiento Celular , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patología , Femenino , Encía/patología , Células Madre Mesenquimatosas/patología , Ratones , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/patología , Traumatismos de los Nervios Periféricos/terapia , Ratas , Ratas Sprague-Dawley , Células de Schwann/patología , Nervio Ciático/lesiones , Nervio Ciático/fisiología
7.
Sci Rep ; 8(1): 6634, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29700345

RESUMEN

Despite the promising neuro-regenerative capacities of stem cells, there is currently no licensed stem cell-based product in the repair and regeneration of peripheral nerve injuries. Here, we explored the potential use of human gingiva-derived mesenchymal stem cells (GMSCs) as the only cellular component in 3D bio-printed scaffold-free neural constructs that were transplantable to bridge facial nerve defects in rats. We showed that GMSCs have the propensity to aggregate into compact 3D-spheroids that could produce their own matrix. When cultured under either 2D- or 3D-collagen scaffolds, GMSC spheroids were found to be more capable of differentiating into both neuronal and Schwann-like cells than their adherent counterparts. Using a scaffold-free 3D bio-printer system, nerve constructs were printed from GMSC spheroids in the absence of exogenous scaffolds and allowed to mature in a bioreactor. In vivo transplantation of the GMSC-laden nerve constructs promoted regeneration and functional recovery when used to bridge segmental defects in rat facial nerves. Our findings suggest that GMSCs represent an easily accessible source of MSCs for 3D bio-printing of scaffold-free nervous tissue constructs with promising potential application for repair and regeneration of peripheral nerve defects.


Asunto(s)
Nervio Facial , Encía/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Regeneración Nerviosa , Impresión Tridimensional , Andamios del Tejido , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Colágeno/metabolismo , Humanos , Inmunohistoquímica , Ratas , Andamios del Tejido/química
8.
Mol Neurobiol ; 55(8): 6965-6983, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29372546

RESUMEN

Non-genetic induction of somatic cells into neural crest stem-like cells (NCSCs) is promising for potential cell-based therapies for post-traumatic peripheral nerve regeneration. Here, we report that human gingiva-derived mesenchymal stem cells (GMSCs) could be reproducibly and readily induced into NCSCs via non-genetic approaches. Compared to parental GMSCs, induced NCSC population had increased expression in NCSC-related genes and displayed robust differentiation into neuronal and Schwann-like cells. Knockdown of the expression of Yes-associated protein 1 (YAP1), a critical mechanosensor and mechanotransducer, attenuated the expression of NCSC-related genes; specific blocking of RhoA/ROCK activity and non-muscle myosin II (NM II)-dependent contraction suppressed YAP1 and NCSC-related genes and concurrently abolished neural spheroid formation in NCSCs. Using a rat model of facial nerve defect, implantation of NCSC-laden nerve conduits promoted functional regeneration of the injured nerve. These promising findings demonstrate that induced NCSCs derived from GMSCs represent an easily accessible and promising source of neural stem-like cells for peripheral nerve regeneration.


Asunto(s)
Nervio Facial/fisiología , Encía/citología , Células Madre Mesenquimatosas/citología , Regeneración Nerviosa/fisiología , Cresta Neural/citología , Células-Madre Neurales/trasplante , Actomiosina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Medios de Cultivo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Nervio Facial/efectos de los fármacos , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Laminas/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Miosina Tipo II/metabolismo , Células-Madre Neurales/efectos de los fármacos , Péptidos/farmacología , Fosfoproteínas/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Factores de Transcripción , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Señalizadoras YAP , Proteína de Unión al GTP rhoA/metabolismo
9.
Stem Cells ; 35(9): 2083-2094, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28699252

RESUMEN

Epithelial-mesenchymal transition (EMT), a biological process associated with cancer stem-like or cancer-initiating cell formation, contributes to the invasiveness, metastasis, drug resistance, and recurrence of the malignant tumors; it remains to be determined whether similar processes contribute to the pathogenesis and progression of ameloblastoma (AM), a benign but locally invasive odontogenic neoplasm. Here, we demonstrated that EMT- and stem cell-related genes were expressed in the epithelial islands of the most common histologic variant subtype, the follicular AM. Our results revealed elevated interleukin (IL)-6 signals that were differentially expressed in the stromal compartment of the follicular AM. To explore the stromal effect on tumor pathogenesis, we isolated and characterized both mesenchymal stromal cells (AM-MSCs) and epithelial cells (AM-EpiCs) from follicular AM and demonstrated that, in in vitro culture, AM-MSCs secreted a significantly higher level of IL-6 as compared to the counterpart AM-EpiCs. Furthermore, both in vitro and in vivo studies revealed that exogenous and AM-MSC-derived IL-6 induced the expression of EMT- and stem cell-related genes in AM-EpiCs, whereas such effects were significantly abrogated either by a specific inhibitor of STAT3 or ERK1/2, or by knockdown of Slug gene expression. These findings suggest that AM-MSC-derived IL-6 promotes tumor-stem like cell formation by inducing EMT process in AM-EpiCs through STAT3 and ERK1/2-mediated signaling pathways, implying a role in the etiology and progression of the benign but locally invasive neoplasm. Stem Cells 2017;35:2083-2094.


Asunto(s)
Ameloblastoma/metabolismo , Ameloblastoma/patología , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Interleucina-6/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ameloblastoma/genética , Animales , Carcinogénesis/patología , Separación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Desnudos , Transducción de Señal
10.
Immunity ; 43(2): 251-63, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26275994

RESUMEN

Regulatory T (Treg) cells are essential for maintenance of immune homeostasis. Here we found that hydrogen sulfide (H2S) was required for Foxp3(+) Treg cell differentiation and function and that H2S deficiency led to systemic autoimmune disease. H2S maintained expression of methylcytosine dioxygenases Tet1 and Tet2 by sulfhydrating nuclear transcription factor Y subunit beta (NFYB) to facilitate its binding to Tet1 and Tet2 promoters. Transforming growth factor-ß (TGF-ß)-activated Smad3 and interleukin-2 (IL-2)-activated Stat5 facilitated Tet1 and Tet2 binding to Foxp3. Tet1 and Tet2 catalyzed conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in Foxp3 to establish a Treg-cell-specific hypomethylation pattern and stable Foxp3 expression. Consequently, Tet1 and Tet2 deletion led to Foxp3 hypermethylation, impaired Treg cell differentiation and function, and autoimmune disease. Thus, H2S promotes Tet1 and Tet2 expression, which are recruited to Foxp3 by TGF-ß and IL-2 signaling to maintain Foxp3 demethylation and Treg-cell-associated immune homeostasis.


Asunto(s)
Colitis/inmunología , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/metabolismo , Sulfuro de Hidrógeno/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Factor de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Colitis/genética , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Dioxigenasas , Factores de Transcripción Forkhead/genética , Homeostasis/genética , Homeostasis/inmunología , Humanos , Interleucina-2/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Factor de Transcripción STAT5/metabolismo , Proteína smad3/metabolismo , Linfocitos T Reguladores/trasplante , Factor de Crecimiento Transformador beta/inmunología
11.
Clin Cancer Res ; 19(12): 3176-88, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23616636

RESUMEN

PURPOSE: Osteonecrosis of the jaw (ONJ) is emerging as one of the important complications in cancer patients treated with antiresorptive agents. This study explored the potential role of interleukin (IL)-17-mediated M1/M2 macrophage alterations in the pathogenesis of bisphosphonate-related osteonecrosis of the jaw (BRONJ). EXPERIMENTAL DESIGN: The expression of IL-17 and M1 and M2 macrophage markers at the local mucosal site of human BRONJ lesions was examined by immunofluorescence studies. BRONJ-like disease was induced in C57BL/6 mice and multiple myeloma-burdened mice by intravenous injection of zoledronate to evaluate the correlation of elevated IL-17 levels with changes in M1 and M2 macrophage phenotypes and the therapeutic effects of blocking IL-17 on pathogenesis of BRONJ-like disease. RESULTS: Increased T-helper (TH)17 cells and IL-17 cytokine correlate with an increase in M1/M2 macrophages ratio at the local mucosal site of both murine and human BRONJ lesion. Convincingly, in mice burdened with multiple myeloma, a combination of elevated suprabasal level and drug-induced IL-17 activity augmented the incidence of BRONJ; both systemic increase of IL-17 and disease severity could be reversed by adoptive transfer of ex vivo expanded M2 macrophages. Targeting IL-17 via specific neutralizing antibodies or a small inhibitory molecule, laquinimod, significantly decreased M1/M2 ratio and concomitantly suppressed BRONJ-like condition in mice. Mechanistically, IL-17 enhanced IFN-γ-induced M1 polarization through augmenting STAT-1 phosphorylation while suppressing IL-4-mediated M2 conversion via inhibiting STAT-6 activation. CONCLUSIONS: These findings have established a compelling linkage between activated IL-17-mediated polarization of M1 macrophages and the development of BRONJ-like conditions in both human disease and murine models.


Asunto(s)
Osteonecrosis de los Maxilares Asociada a Difosfonatos/patología , Interleucina-17/biosíntesis , Maxilares/patología , Neoplasias/complicaciones , Animales , Osteonecrosis de los Maxilares Asociada a Difosfonatos/inmunología , Osteonecrosis de los Maxilares Asociada a Difosfonatos/metabolismo , Difosfonatos/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Imidazoles/administración & dosificación , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Interleucina-17/metabolismo , Maxilares/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Ácido Zoledrónico
12.
Stem Cells Dev ; 21(6): 937-47, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-21689066

RESUMEN

Mesenchymal stem cells (MSCs) are capable of regenerative and immunomodulatory functions in cell-based therapies in a variety of human diseases and injuries; however, their therapeutic efficacy and potential side effects remain major obstacles in clinical applications. We report here a 3D spheroid culture approach to optimize stem cell properties and therapeutic effects of human gingiva-derived mesenchymal stem cells (GMSCs) in mitigation of experimental oral mucositis. Under growth condition of ultra-low attachment, GMSCs spontaneously aggregated into 3D spheroids and exhibited distinct early stem cell phenotype characterized by elevated expression Stro-1 and CXC chemokine receptor 4 (CXCR-4) as well as OCT-4 and Nanog, 2 important transcriptional factors relevant to stem cell properties, and decreased expression of MSC-associated markers, including CD29, CD90, and CD105. Functionally, spheroid GMSCs are capable of enhanced multipotency and augmented secretion of several chemokines and cytokines relevant to cell migration, survival, and angiogenesis. More importantly, spheroid GMSCs expressed increased levels of reactive oxygen species, hypoxia-inducible factor (HIF)-1 and -2α, and manganese superoxide dismutase, which correlated with improved resistance to oxidative stress-induced apoptosis. Using an in vivo murine model of chemotherapy-induced oral mucositis, we demonstrated that spheroid-derived GMSCs possessed better therapeutic efficacy than their adherent cells in reversing body weight loss and promoting the regeneration of disrupted epithelial lining of the mucositic tongues. These findings suggest that 3D spheroid culture allows early stemness preservation and potentially precondition GMSCs for enhanced mitigation of oral mucositis.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Encía/patología , Células Madre Mesenquimatosas/citología , Esferoides Celulares/patología , Estomatitis/terapia , Animales , Técnicas de Cultivo de Célula , Trasplante de Células Madre Mesenquimatosas , Ratones , Estomatitis/patología , Estomatitis/prevención & control , Resultado del Tratamiento
13.
Stem Cells ; 29(11): 1849-60, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21987520

RESUMEN

The immunomodulatory and anti-inflammatory functions of mesenchymal stromal cells (MSCs) have been demonstrated in several autoimmune/inflammatory disease models, but their contribution to the mitigation of contact hypersensitivity (CHS) remains unclear. Here, we report a new immunological approach using human gingiva-derived MSCs (GMSCs) to desensitize and suppress CHS and the underlying mechanisms. Our results showed that systemic infusion of GMSCs before the sensitization and challenge phase dramatically suppress CHS, manifested as a decreased infiltration of dendritic cells (DCs), CD8(+) T cells, T(H)-17 and mast cells (MCs), a suppression of a variety of inflammatory cytokines, and a reciprocal increased infiltration of regulatory T cells and expression of IL-10 at the regional lymph nodes and the allergic contact areas. The GMSC-mediated immunosuppressive effects and mitigation of CHS were significantly abrogated on pretreatment with indomethacin, an inhibitor of cyclooxygenases. Under coculture condition of direct cell-cell contact or via transwell system, GMSCs were capable of direct suppression of differentiation of DCs and phorbol 12-myristate 13-acetate-stimulated activation of MCs, whereas the inhibitory effects were attenuated by indomethacin. Mechanistically, GMSC-induced blockage of de novo synthesis of proinflammatory cytokines by MCs is mediated partly by the tumor necrosis factor-alpha/prostaglandin E(2) (PGE(2)) feedback axis. These results demonstrate that GMSCs are capable of desensitizing allergic contact dermatitis via PGE(2)-dependent mechanisms.


Asunto(s)
Dermatitis por Contacto/metabolismo , Dinoprostona/metabolismo , Encía/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Células Cultivadas , Dermatitis por Contacto/genética , Dinoprostona/genética , Citometría de Flujo , Humanos , Ratones
14.
Stem Cells ; 28(10): 1856-68, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20734355

RESUMEN

Increasing evidence has supported the important role of mesenchymal stem cells (MSCs) in wound healing, however, the underlying mechanism remains unclear. Recently, we have isolated a unique population of MSCs from human gingiva (GMSCs) with similar stem cell-like properties, immunosuppressive, and anti-inflammatory functions as human bone marrow-derived MSCs (BMSCs). We describe here the interplay between GMSCs and macrophages and the potential relevance in skin wound healing. When cocultured with GMSCs, macrophages acquired an anti-inflammatory M2 phenotype characterized by an increased expression of mannose receptor (MR; CD206) and secretory cytokines interleukin (IL)-10 and IL-6, a suppressed production of tumor necrosis factor (TNF)-α, and decreased ability to induce Th-17 cell expansion. In vivo, we demonstrated that systemically infused GMSCs could home to the wound site in a tight spatial interaction with host macrophages, promoted them toward M2 polarization, and significantly enhanced wound repair. Mechanistically, GMSC treatment mitigated local inflammation mediated by a suppressed infiltration of inflammatory cells and production of IL-6 and TNF-α, and an increased expression of IL-10. The GMSC-induced suppression of TNF-α secretion by macrophages appears to correlate with impaired activation of NFκB p50. These findings provide first evidence that GMSCs are capable to elicit M2 polarization of macrophages, which might contribute to a marked acceleration of wound healing.


Asunto(s)
Encía/citología , Macrófagos/citología , Macrófagos/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Cicatrización de Heridas/inmunología , Animales , Western Blotting , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Cancer Lett ; 289(2): 151-60, 2010 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-19748175

RESUMEN

The potential role of cancer stem-like cells (CSCs) in chemoresistance of human oral squamous cell carcinoma (OSCC) was examined. A small subpopulation (1-2%) of CD133(+) CSCs was identified in OSCC cell lines and tissues. These CD133(+) CSCs possess higher clonogenicity, invasiveness, and increased in vivo tumorigenicity as compared to CD133(-) counterparts. Meanwhile, CD133(+) CSCs were substantially resistant to standard chemotherapy, wherein both in vitro and in vivo treatment with paclitaxel resulted in a marked enrichment for CD133(+) CSCs. Our data suggest that CD133(+) cells represent a small subpopulation of CSCs that may contribute to chemoresistance in human OSCC.


Asunto(s)
Antígenos CD/metabolismo , Antineoplásicos/efectos adversos , Carcinoma de Células Escamosas/patología , Resistencia a Antineoplásicos , Glicoproteínas/metabolismo , Neoplasias de la Boca/patología , Células Madre Neoplásicas/patología , Péptidos/metabolismo , Antígeno AC133 , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Ensayo de Unidades Formadoras de Colonias , Femenino , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones Desnudos , Neoplasias de la Boca/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Int J Cancer ; 126(1): 90-103, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19569175

RESUMEN

Adjunctive chemotherapy with bisphosphonates has been reported to delay bone metastasis and improve overall survival in breast cancer. Aside from its antiresorptive effect, bisphosphonates exhibit antitumor activities, in vitro and in vivo, via several mechanisms, including antiangiogenesis. In this study, we investigated the potential molecular mechanisms underlying the antiangiogenic effect of non-nitrogen-containing and nitrogen-containing bisphosphonates, clodronate and pamidronate, respectively, in insulin-like growth factor (IGF)-1 responsive human breast cancer cells. We tested whether bisphosphonates had any effects on hypoxia-inducible factor (HIF)-1alpha/vascular endothelial growth factor (VEGF) axis that plays a pivotal role in tumor angiogenesis, and our results showed that both pamidronate and clodronate significantly suppressed IGF-1-induced HIF-1alpha protein accumulation and VEGF expression in MCF-7 cells. Mechanistically, we found that either pamidronate or clodronate did not affect mRNA expression of HIF-1alpha, but they apparently promoted the degradation of IGF-1-induced HIF-1alpha protein. Meanwhile, we found that the presence of pamidronate and clodronate led to a dose-dependent decease in the newly-synthesized HIF-1alpha protein induced by IGF-1 in breast cancer cells after proteasomal inhibition, thus, indirectly reflecting the inhibition of protein synthesis. In addition, our results indicated that the inhibitory effects of bisphosphonates on the HIF-1alpha/VEGF axis are associated with the inhibition of the phosphoinositide 3-kinase/AKT/mammalian target of rapamycin signaling pathways. Consistently, we demonstrated that pamidronate and clodronate functionally abrogated both in vitro and in vivo tumor angiogenesis induced by IGF-1-stimulated MCF-7 cells. These findings have highlighted an important mechanism of the pharmacological action of bisphosphonates in the inhibition of tumor angiogenesis in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/irrigación sanguínea , Difosfonatos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Neovascularización Patológica/prevención & control , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR
17.
PLoS One ; 4(11): e7798, 2009 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-19907660

RESUMEN

BACKGROUND: Alterations in the stem cell niche are likely to contribute to tumorigenesis; however, the concept of niche promoted benign tumor growth remains to be explored. Here we use keloid, an exuberant fibroproliferative dermal growth unique to human skin, as a model to characterize benign tumor-like stem cells and delineate the role of their "pathological" niche in the development of the benign tumor. METHODS AND FINDINGS: Subclonal assay, flow cytometric and multipotent differentiation analyses demonstrate that keloid contains a new population of stem cells, named keloid derived precursor cells (KPCs), which exhibit clonogenicity, self-renewal, distinct embryonic and mesenchymal stem cell surface markers, and multipotent differentiation. KPCs display elevated telomerase activity and an inherently upregulated proliferation capability as compared to their peripheral normal skin counterparts. A robust elevation of IL-6 and IL-17 expression in keloid is confirmed by cytokine array, western blot and ELISA analyses. The altered biological functions are tightly regulated by the inflammatory niche mediated by an autocrine/paracrine cytokine IL-17/IL-6 axis. Utilizing KPCs transplanted subcutaneously in immunocompromised mice we generate for the first time a human keloid-like tumor model that is driven by the in vivo inflammatory niche and allows testing of the anti-tumor therapeutic effect of antibodies targeting distinct niche components, specifically IL-6 and IL-17. CONCLUSIONS/SIGNIFICANCE: These findings support our hypothesis that the altered niche in keloids, predominantly inflammatory, contributes to the acquirement of a benign tumor-like stem cell phenotype of KPCs characterized by the uncontrolled self-renewal and increased proliferation, supporting the rationale for in vivo modification of the "pathological" stem cell niche as a novel therapy for keloid and other mesenchymal benign tumors.


Asunto(s)
Interleucina-17/metabolismo , Interleucina-6/metabolismo , Queloide/metabolismo , Adulto , Animales , Proliferación Celular , Separación Celular , Citocinas/metabolismo , Fibroblastos/metabolismo , Humanos , Inflamación , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
18.
J Immunol ; 183(12): 7787-98, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19923445

RESUMEN

Aside from the well-established self-renewal and multipotent differentiation properties, mesenchymal stem cells exhibit both immunomodulatory and anti-inflammatory roles in several experimental autoimmune and inflammatory diseases. In this study, we isolated a new population of stem cells from human gingiva, a tissue source easily accessible from the oral cavity, namely, gingiva-derived mesenchymal stem cells (GMSCs), which exhibited clonogenicity, self-renewal, and multipotent differentiation capacities. Most importantly, GMSCs were capable of immunomodulatory functions, specifically suppressed peripheral blood lymphocyte proliferation, induced expression of a wide panel of immunosuppressive factors including IL-10, IDO, inducible NO synthase (iNOS), and cyclooxygenase 2 (COX-2) in response to the inflammatory cytokine, IFN-gamma. Cell-based therapy using systemic infusion of GMSCs in experimental colitis significantly ameliorated both clinical and histopathological severity of the colonic inflammation, restored the injured gastrointestinal mucosal tissues, reversed diarrhea and weight loss, and suppressed the overall disease activity in mice. The therapeutic effect of GMSCs was mediated, in part, by the suppression of inflammatory infiltrates and inflammatory cytokines/mediators and the increased infiltration of regulatory T cells and the expression of anti-inflammatory cytokine IL-10 at the colonic sites. Taken together, GMSCs can function as an immunomodulatory and anti-inflammatory component of the immune system in vivo and is a promising cell source for cell-based treatment in experimental inflammatory diseases.


Asunto(s)
Colitis/inmunología , Colitis/prevención & control , Encía/citología , Encía/inmunología , Inmunomodulación/fisiología , Mediadores de Inflamación/fisiología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Adulto , Animales , Células Cultivadas , Técnicas de Cocultivo , Colitis/metabolismo , Femenino , Encía/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Distribución Aleatoria
19.
Clin Cancer Res ; 13(16): 4686-94, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699846

RESUMEN

PURPOSE: Nicotine, the major component in cigarette smoke, can promote tumor growth and angiogenesis in various cancers, including lung cancer. Hypoxia-inducible factor-1alpha (HIF-1alpha) is overexpressed in human lung cancers, particularly in non-small cell lung cancers (NSCLC), and is closely associated with an advanced tumor grade, increased angiogenesis, and resistance to chemotherapy and radiotherapy. The purpose of this study was to investigate the effects of nicotine on the expression of HIF-1alpha and its downstream target gene, vascular endothelial growth factor (VEGF), in human lung cancer cells. EXPERIMENTAL DESIGN: Human NSCLC cell lines A549 and H157 were treated with nicotine and examined for expression of HIF-1alpha and VEGF using Western blot or ELISA. Loss of HIF-1alpha function using specific small interfering RNA was used to determine whether HIF-1alpha is directly involved in nicotine-induced tumor angiogenic activities, including VEGF expression, cancer cell migration, and invasion. RESULTS: Nicotine increased HIF-1alpha and VEGF expression in NSCLC cells. Pharmacologically blocking nicotinic acetylcholine receptor-mediated signaling cascades, including the Ca2+/calmodulin, c-Src, protein kinase C, phosphatidylinositol 3-kinase, mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2, and the mammalian target of rapamycin pathways, significantly attenuated nicotine-induced up-regulation of HIF-1alpha protein. Functionally, nicotine potently stimulated in vitro tumor angiogenesis by promoting tumor cell migration and invasion. These proangiogenic and invasive effects were partially abrogated by treatment with small interfering RNA specific for HIF-1alpha. CONCLUSION: These findings identify novel mechanisms by which nicotine promotes tumor angiogenesis and metastasis and provide further evidences that HIF-1alpha is a potential anticancer target in nicotine-associated lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Neoplasias Pulmonares/metabolismo , Nicotina/farmacología , Receptores Nicotínicos/fisiología , Transducción de Señal/fisiología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias Pulmonares/patología , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/fisiología , ARN Interferente Pequeño/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis
20.
Clin Cancer Res ; 13(9): 2568-76, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17473185

RESUMEN

PURPOSE: Human papillomavirus (HPV)-16 oncoproteins, E6 and E7, are associated with enhanced tumor angiogenesis in human cervical cancers. The purpose of this study was (a) to investigate whether expression of HPV-16 E6 and E7 oncoproteins induces hypoxia-inducible factor 1 alpha (HIF-1 alpha) and vascular endothelial growth factor expression in cervical cancer cells; and (b) to assess the effect of resveratrol on 16 E6- and E7-induced HIF-1 alpha and VEGF gene expression. EXPERIMENTAL DESIGN: Human cervical cancer cell lines C-33A and HeLa were transiently cotransfected with pSG5-HPV-16 E6 or 16 E7 constructs along with HIF-1 alpha small interfering RNA (siRNA) or nonspecific siRNA. The expression of HIF-1 alpha/VEGF was measured using real-time PCR, Western blot analysis, or ELISA. The in vitro angiogenic activity induced by 16 E6- and E7-transfected cells was examined. The effect of resveratrol on oncoprotein-induced HIF-1 alpha/VEGF expression and in vitro angiogenesis was investigated. RESULTS: HPV-16 E6- and E7-transfected cervical cancer cells express increased HIF-1 alpha protein and VEGF expression. These stimulatory effects were abrogated by cotransfection with either HIF-1 alpha siRNA or treatment with resveratrol. Blocking extracellular signal-regulated kinase 1/2 (ERK 1/2) and phosphoinositide-3-kinase by PD98059 and LY294002, respectively, abolished 16 E6- and E7-induced HIF-1 alpha and VEGF expression. Functionally, we showed that HPV-16 E6- and E7-transfected cervical cancer cells stimulated in vitro capillary or tubule formation, and these angiogenic effects could be abolished either by cotransfection with HIF-1 alpha siRNA or by treatment with resveratrol. CONCLUSION: HPV-16 oncoproteins contribute to enhanced angiogenesis in cervical cancer cells via HIF-1 alpha-dependent VEGF expression. Resveratrol suppresses 16 E6- and E7-induced HIF-1 alpha-mediated angiogenic activity and, thus, is a promising chemotherapeutic agent for human cervical cancer.


Asunto(s)
Carcinoma/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas Represoras/genética , Neoplasias del Cuello Uterino/metabolismo , Anticarcinógenos/farmacología , Carcinoma/genética , Cromonas/farmacología , Femenino , Flavonoides/farmacología , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/análisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Morfolinas/farmacología , Neovascularización Patológica/genética , Proteínas E7 de Papillomavirus , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Resveratrol , Estilbenos/farmacología , Activación Transcripcional , Transfección , Neoplasias del Cuello Uterino/genética , Factor A de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...