Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
ChemMedChem ; : e202300728, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38757641

RESUMEN

Synthesis of non-platinum transition metal complexes with N,O donor chelating  ligand for application  against cancer with higher efficacy and selectivity is currently an important research field. We assessed The anti-cancer effects of a mixed ligand Ni(II) complex on human breast and lung cancer cell lines in the current investigation. Mononuclear mixed ligand octahedral Ni(II) complex [NiIIL(NO3)(MeOH)] complex(1), with tri-dentate phenol-based ligand 2,4-dichloro-6-((4-methylpiperazin-1-yl)methyl)phenol (HL) along with  methanol  and nitrate as ancillary ligand was prepared.  Piperazine moiety of the ligand exists as boat conformation in this complex as revealed from single crystal X-ray study. UV -visible spectrum of complex (1) exhibits three distinct d-d bands due to spin-allowed 3A2g→3T1g(P), 3A2g→3T1g(F) and 3A2g→3T2g(F) transitions as expected octahedral d8 system. Complex (1) through transactivation of p53 and its pro-apoptotic downstream targets induce apoptotic cell death in mouse and human cancer cells such as mcf-7, A549 and MDA-MB-231 in dose dependent manner. Furthermore, complex (1) was able to slow the migratory rate of MDA-MB-231 cells' in vitro as well as epithelia -mesenchymal transition (EMT), the key step for metastatic transition and malignancy. Over all our results suggest complex (1) as potential agent in anti-tumor treatment regimen both as cytotoxic and anti-metastatic agent against malignant neoplasia.

2.
J Leukoc Biol ; 115(5): 958-984, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38236200

RESUMEN

Myeloid-derived suppressor cells are heterogenous immature myeloid lineage cells that can differentiate into neutrophils, monocytes, and dendritic cells as well. These cells have been characterized to have potent immunosuppressive capacity in neoplasia and a neoplastic chronic inflammatory microenvironment. Increased accumulation of myeloid-derived suppressor cells was reported with poor clinical outcomes in patients. They support neoplastic progression by abrogating antitumor immunity through inhibition of lymphocyte functions and directly by facilitating tumor development. Yet the shifting genetic signatures of this myeloid lineage cell toward immunosuppressive functionality in progressive tumor development remain elusive. We have attempted to identify the gene expression profile using lineage-specific markers of these unique myeloid lineage cells in a tumor microenvironment and bone marrow using a liquid transplantable mice tumor model to trace the changing influence of the tumor microenvironment on myeloid-derived suppressor cells. We analyzed the phenotype, functional shift, suppressive activity, differentiation status, and microarray-based gene expression profile of CD11b+Gr1+ lineage-specific cells isolated from the tumor microenvironment and bone marrow of 4 stages of tumor-bearing mice and compared them with control counterparts. Our analysis of differentially expressed genes of myeloid-derived suppressor cells isolated from bone marrow and the tumor microenvironment reveals unique gene expression patterns in the bone marrow and tumor microenvironment-derived myeloid-derived suppressor cells. It also suggests T-cell suppressive activity of myeloid-derived suppressor cells progressively increases toward the mid-to-late phase of the tumor and a significant differentiation bias of tumor site myeloid-derived suppressor cells toward macrophages, even in the presence of differentiating agents, indicating potential molecular characteristics of myeloid-derived suppressor cells in different stages of the tumor that can emerge as an intervention target.


Asunto(s)
Diferenciación Celular , Progresión de la Enfermedad , Células Supresoras de Origen Mieloide , Microambiente Tumoral , Animales , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Microambiente Tumoral/inmunología , Ratones , Regulación Neoplásica de la Expresión Génica , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Células de la Médula Ósea/metabolismo , Antígeno CD11b/metabolismo , Antígeno CD11b/genética , Médula Ósea/patología , Médula Ósea/metabolismo
3.
Phytomedicine ; 119: 155007, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37549537

RESUMEN

BACKGROUND: 1-2.88% of human populations are affected by psoriasis, one type of chronic inflammatory skin disease. Skin thickenings, erythema, scaling in skin are the most important symptoms of psoriasis. There are renewed interests amongst scientists in studying anti-inflammatory property of the plant extracts due to lower side effects and cost effectiveness. There are few reports suggesting anti-inflammatory activity of Premna herbacea roxb. but lacks systematic evaluation of these properties. METHODS: We, initially tested the anti-inflammatory activity of crude root methanolic extract in vitro, where it significantly reduced LPS generated ROS in splenic macrophages. We further tested the TLC and HPLC fraction in order to find active ingredient in Premna herbacea roxb. root extract that ameliorated the chronic inflammation of skin and performed GC-MS and LC-MS studies to identify active component. Upon finding significant anti-inflammatory effect of the crude root extract in vitro, We studied the efficacy of the Premna herbacea roxb. root extract in Imiquimod induced psoriasis like skin inflammation in male BALB/C mice that closely resembles human psoriasis. Immunophenotyping, Cytokine productions were observed by flow cytometry, status of gene expression was done by Real time PCR and nuclear co-localization was studied by confocal microscopy. RESULTS: We observed progressive increase in signs and symptoms of the disease in imiquimod treated diseased animals but the Premna herbacea roxb. Root Methanolic Extract (PHRME) reduced the thickening of the skin, redness and scaling in these animals. In our study, along with progression of the disease, the production of macrophages increases and with the application of PHRME, the percentage of macrophages have reduced. CONCLUSION: As per the previous Indigenous traditional knowledge regarding use of Premna herbacea roxb. against inflammatory disorder and lack of detail mechanistic study of the crude root extract prompted us to elucidate the efficacy of the root extract in vitro and in vivo psoriatic mice model. For the first time we have identified three putative bioactive active components (5­hydroxy-7-methoxyflavanone, 3-Hydroxy-7,8,2',3'-tetramethoxyflavone, 2,4',6'-trimethoxy chalcone) from Premna herbacea root methanolic extract (PHRME) and we suggest PHRME and purified active fractions influence NFκB and COX2 signaling pathway to suppress inflammatory conditions. All of the purified components show strong binding efficiency in our molecular docking analysis. Our study also suggests that Premna herbacea roxb. root extract may be explored as cost effective alternative for established treatment regimen as our study also indicates low side effect of the extract against pre-clinical psoriatic model.


Asunto(s)
Psoriasis , Ratones , Humanos , Animales , Imiquimod , Simulación del Acoplamiento Molecular , Ratones Endogámicos BALB C , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Piel , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Macrófagos/metabolismo , Modelos Animales de Enfermedad , Citocinas/metabolismo
4.
Adv Sci (Weinh) ; 10(29): e2302658, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37555802

RESUMEN

Topical chemotherapy approaches are relevant for certain skin cancer treatments. This study observes that cabazitaxel (CTX), a broad-spectrum second-generation taxane cytotoxic agent, can be dissolved in α-tocopherol at high concentrations exceeding 100 mg mL-1 . 2D nuclear magnetic resonance (NMR) analysis and molecular dynamics (MD) are used to study this phenomenon. The addition of 30% dimethyl sulfoxide (DMSO) to the α-tocopherol/CTX solution improves its working viscosity and enhances CTX permeation through human skin in vitro (over 5 µg cm-2 within 24 h), while no detectable drug permeates when CTX is dissolved in α-tocopherol alone. In a transepidermal water loss assay, the barrier impairment induced by CTX in 30% DMSO in α-tocopherol, but not in pure DMSO, is reversible 8 h after the formulation removal from the skin surface. Antitumor efficacy of the topical CTX formulation is evaluated in nude mice bearing A431 human squamous carcinoma skin cancer xenografts. With topical application of concentrated CTX solutions (75 mg mL-1 ), tumor growth is significantly suppressed compared to lower concentration groups (0, 25, or 50 mg mL-1 CTX). Taken together, these findings show that topical delivery of CTX using a DMSO and α-tocopherol solvent warrants further study as a treatment for skin malignancies.


Asunto(s)
Neoplasias Cutáneas , alfa-Tocoferol , Ratones , Animales , Humanos , alfa-Tocoferol/química , Dimetilsulfóxido/uso terapéutico , Ratones Desnudos , Taxoides , Neoplasias Cutáneas/tratamiento farmacológico
5.
Int Immunopharmacol ; 114: 109521, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36470118

RESUMEN

In clinical practice, major efforts are underway to identify appropriate drug combinations to boost anticancer activity while suppressing unwanted adverse effects. In this regard, we evaluated the efficacy of combination treatment with the widely used chemotherapeutic drug doxorubicin along with the TGFßRI inhibitor galunisertib (LY2157299) in aggressive B-cell non-Hodgkin lymphoma (B-NHL). The antiproliferative effects of these drugs as single agents or in combination against several B-NHL cell lines and the synergism of the drug combination were evaluated by calculating the combination index. To understand the putative molecular mechanism of drug synergism, the TGF-ß and stress signaling pathways were analyzed after combination treatment. An aggressive lymphoma model was used to evaluate the anticancer activity and post-therapeutic immune response of the drug combination in vivo. Galunisertib sensitized various B-NHL cells to doxorubicin and in combination synergistically increased apoptosis. The antitumor activity of the drug combinations involved upregulation of p-P38 MAPK and inhibition of the TGF-ß/Smad2/3 and PI3K/AKT signaling pathways. Combined drug treatment significantly reduced tumor growth and enhanced survival, indicating that the synergism between galunisertib and Dox observed in vitro was most likely retained in vivo. Based on the tumor-draining lymph node analysis, combination therapy results in better prognosis, including disappearance of disease-exacerbating regulatory T cells and prevention of CD8+ T-cell exhaustion by downregulating MDSCs. Galunisertib synergistically potentiates the doxorubicin-mediated antitumor effect without aggravating the toxic effects and the ability to kickstart the immune system, supporting the clinical relevance of targeting TGF-ßRI in combination with doxorubicin against lymphoma.


Asunto(s)
Linfoma , Neoplasias , Humanos , Fosfatidilinositol 3-Quinasas/farmacología , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Neoplasias/tratamiento farmacológico , Factor de Crecimiento Transformador beta , Sistema Inmunológico , Sinergismo Farmacológico , Línea Celular Tumoral , Apoptosis
6.
Anal Methods ; 14(33): 3196-3202, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-35938936

RESUMEN

A new benzorhodol-based non-fluorescent organic frame (DEB-CO) detects carbon monoxide (CO) selectively through a spirolactam ring-opening mechanism. Herein, the selective off-on fluorogenic behavior of this probe towards CO has been achieved without any assistance of precious and hazardous metals (e.g. Pd2+) as additional substrates. Moreover, the red-emissive probe motivated us to apply in situ tracing in mice and living cells. The selective off-on fluorogenic behavior of this probe towards CO originating from CORM-3 in vitro and in vivo with a limit of detection as low as 64.29 nM (for CORM-3) has been observed. Additionally, this probe is capable of sensing toxic carbon monoxide gas. This probe has also been utilized to detect intracellular CO in MCF7 cells (in vitro) and to spot the distribution of CO in mice (in vivo) by acquiring bioimages with the help of confocal microscopy, which indicates that DEB-CO is a smart competent probe for this purpose.


Asunto(s)
Monóxido de Carbono , Colorantes Fluorescentes , Animales , Humanos , Células MCF-7 , Metales , Ratones , Microscopía Confocal
7.
Carbohydr Polym ; 291: 119614, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35698411

RESUMEN

We report the solvent-free green synthesis of two Schiff bases, (E)-2-((2-hydroxy-3-methoxybenzylidene)amino)-4-methylphenol (SL1) and (E)-2-((2-hydroxybenzylidene) amino)-4-methylphenol (SL2), and their inclusion complexes with ß-cyclodextrin (ß-CD). The encapsulation phenomenon, structural characteristics and hydrolytic stabilities of the SL1, SL2 and their inclusion complexes are determined with a suite of spectroscopic, analytical and crystallographic analyses. Dose and time-dependent cytotoxicity study of SL1-ß-CD and SL2-ß-CD against two breast cancer cell lines, Michigan Cancer Foundation-7 (MCF-7) and metastatic mammary adenocarcinoma1 (MDA-MB-231), exhibit excellent inhibitory activity with significant non-cytotoxic concentrations and ensure a multifold elevation of bio-potency than the parent Schiff base compounds. The Annexin-V assay determines the efficacy of these inclusion complexes to trigger apoptosis, suggesting that SL2-ß-CD possesses better efficacy as an anti-cancer drug. To the best of our knowledge, we, for the first time, report the inclusion of nanocrystalline Schiff bases into ß-CD for multifold enrichment of bio-potency.


Asunto(s)
Antineoplásicos , beta-Ciclodextrinas , Antineoplásicos/química , Apoptosis , Humanos , Células MCF-7 , Bases de Schiff/química , beta-Ciclodextrinas/farmacología
8.
Front Pharmacol ; 13: 852065, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35444547

RESUMEN

Integrative medicine practices, such as Ayurveda, are popular in India and many South Asian countries, yet basic research to investigate the concepts, procedures, and medical benefits of ayurvedic products has received little attention and is not fully understood. Here, we report a functional nanodiamond-based traditional Ayurvedic herbomineral formulation, Heerak Bhasma (Ayu_ND), for the treatment of solid tumors called Dalton's lymphoma generated in CD1 mice. Ayu_ND-mediated immunostimulation significantly reduces tumor cell proliferation and induces apoptosis aided by the active participation of dendritic cells. Immunomodulatory Ayu_ND treatment is highly immunostimulatory and drives dendritic cells to produce TNF-α. Treatment with Ayu_ND significantly reduces the tumor volume, inhibits metastasis in distant vascularized organs, and increases the life span of tumor-bearing animals compared with untreated littermates. These events were associated with elevated serum levels of the protective cytokines IFN-γ and TNF-α and downregulated the disease, exacerbating TGF-ß. Ayu_ND-mediated therapeutic success was also accompanied by the depletion of regulatory T cells and enhanced vaccine-induced T-cell immunity, guided by the restoration of the memory CD8+ T-cell pool and prevention of PD-1-mediated T cell exhaustion. The results provide a basis for further evaluation of ayurvedic formulations and drug efficacy in treating cancers.

9.
Life Sci ; 253: 117731, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32353431

RESUMEN

BACKGROUND: Very little is known about the role inflammation and mechanism(s) that enables the tumor to evade host's anti-tumor immune function during very initial days of tumor establishment. Our study focuses on the immune response and local inflammation specially the pro-inflammatory and immune modifier components that are responsible for tumor-induced immune-suppression, tumor-associated macrophages (TAM) at tumor microenvironment in mouse model from very early to late phase of tumor progression. METHODS: 1 × 105 Ascites tumor, EAC in Swiss albino or Sarcoma-180 (S-180) in Balb c mice strain were inoculated intra-peritonially and grouped into Control (0 day or no tumor), initial phase (3 day tumor), early (7 Day), Late (14 day) and terminal (21 day tumor) sets. T cell activity, tumor niche macrophage, inflammatory signatures were studied using Confocal microscopy, flowcytometry, ELISA, q-RT PCR and Western blot. RESULTS: We observed increased T cell infiltration at a very early stage of tumorigenesis in the tumor site with elevated percentage of activated/memory T cells. But increased cellular death and functional suppression of tumor site T cells during final stages. We observed increased infiltration of TAMs with skewed M2 phenotype. Increased chemokine receptor expression could be noted on these TAMs. Using HIF-1α inhibitor and prostaglandin receptor antagonists we demonstrated crucial role of these factor in functional alteration in TAMs. HIF-1α inhibition and also by prostaglandin receptor inhibition reduced signature pro-inflammatory gene expression, migration of macrophages and T cell suppression capacity of TAMs. We also demonstrated that PGE2 can induce HIF-1α activation in relatively less hypoxic microenvironment during early stages of tumor. CONCLUSION: Altogether, these findings strongly suggest link between prostaglandin mediated early HIF-1α activation and subsequent hypoxia induced HIF-1α activation that further enhances prostaglandin synthesis driving the recruitment and functional alteration of tumor site macrophages.


Asunto(s)
Carcinoma de Ehrlich/patología , Dinoprostona/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/patología , Sarcoma 180/patología , Animales , Carcinoma de Ehrlich/inmunología , Movimiento Celular , Progresión de la Enfermedad , Inflamación/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Fenotipo , Sarcoma 180/inmunología , Linfocitos T/inmunología , Microambiente Tumoral/inmunología
10.
Langmuir ; 33(24): 6167-6181, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28564546

RESUMEN

Colloidal gels are three-dimensional networks of microgel particles and can be utilized to design microtissues where the differential adhesive interactions between the particles and cells, guided by their surface energetics, are engineered to spatially assemble the cellular and colloidal components into three-dimensional microtissues. In this work we utilized a colloidal interaction approach to design cell-polyurethane (PU) microgel bimodal microtissues using endothelial cells (ECs) as a normal cell model and a nonmalignant breast cancer cell line (MCF-7) as a cancer cell model. PU microgels were developed from a library of segmental polyurethanes with poly(ethylene glycol) soft segment and aliphatic diisocyanate/l-tyrosine based chain extender as hard segment to modulate the interactions between PU colloidal particles and cells. The surface energies of the microgel particles and cells were estimated using Zisman's critical surface tension and van Oss-Good-Chaudhury theory (vOGCT) from liquid contact angle analysis. Binary interaction potentials between colloidal PU particles and cells and the ternary interaction between colloidal PU particle, cell, and collagen I/Matrigel were calculated to explain the formation of microtissues and their spreading in extraneous biomatrix respectively by using classical and extended DLVO theory (XDLVO). Furthermore, rheological analysis and in silico simulations were used to analyze the assembly and spreading of the PU microgel based microtissues. In vitro experiments showed that ECs and MCF-7 displayed more differentiated (EC spreading/MCF-7 lumen formation) character when mixed with microgel particles that were stable in aqueous medium and more undifferentiated character (EC nonspreading/MCF-7 spreading) when mixed with microgel particles unstable in aqueous medium.


Asunto(s)
Poliuretanos/química , Diferenciación Celular , Colágeno Tipo I , Geles , Tensión Superficial
11.
Mater Sci Eng C Mater Biol Appl ; 54: 182-95, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26046282

RESUMEN

Segmental polyurethanes exhibit biphasic morphology and can control cell fate by providing distinct matrix guided signals to increase the chondrogenic potential of mesenchymal stem cells (MSCs). Polyethylene glycol (PEG) based hydrophilic polyurethanes can deliver differential signals to MSCs through their matrix phases where hard segments are cell-interactive domains and PEG based soft segments are minimally interactive with cells. These coordinated communications can modulate cell-matrix interactions to control cell shape and size for chondrogenesis. Biphasic character and hydrophilicity of polyurethanes with gel like architecture provide a synthetic matrix conducive for chondrogenesis of MSCs, as evidenced by deposition of cartilage-associated extracellular matrix. Compared to monophasic hydrogels, presence of cell interactive domains in hydrophilic polyurethanes gels can balance cell-cell and cell-matrix interactions. These results demonstrate the correlation between lineage commitment and the changes in cell shape, cell-matrix interaction, and cell-cell adhesion during chondrogenic differentiation which is regulated by polyurethane phase morphology, and thus, represent hydrophilic polyurethanes as promising synthetic matrices for cartilage regeneration.


Asunto(s)
Condrogénesis , Células Madre Mesenquimatosas/citología , Polietilenglicoles/química , Poliuretanos/química , Cartílago/citología , Comunicación Celular , Diferenciación Celular , Células Inmovilizadas/citología , Matriz Extracelular/química , Humanos , Hidrogeles/química , Interacciones Hidrofóbicas e Hidrofílicas
12.
Macromol Biosci ; 15(6): 747-55, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25771753

RESUMEN

Controlling hydrogel structures by combination of physical and chemical cross-links provides a novel system to regulate (stem) cell fate. In this study, we designed a polyethylene glycol (PEG)-based hydrogel where the polymer chains contain both physical and chemical cross-linking units in the same chain with self-assembling L-tyrosine-based dipeptides and photopolymerizable polyacrylate groups, respectively. It is shown that hydrogel architectures derived from this polymer are correlated to the cross-linking mechanisms. Combination of these cross-links controls three-dimensional gel architecture to regulate stem cell behavior in these hydrogels. Particularly, interaction of mesenchymal stem cells with the hydrogel enabled cellular aggregation to enhance chondrogenic differentiation as observed from the deposition of chondrogenic matrix. Increased chondrogenesis was due to enhanced cell-cell adhesion, which was mediated by gel morphology. This study shows the interplay of physical and chemical cross-links in hydrogels to regulate stem cell function and provides a novel molecular engineering tool for controlling hydrogel properties.


Asunto(s)
Condrogénesis , Reactivos de Enlaces Cruzados/química , Dipéptidos/química , Hidrogeles/química , Células Madre Mesenquimatosas/metabolismo , Polietilenglicoles/química , Adhesión Celular , Humanos , Células Madre Mesenquimatosas/citología
13.
Nat Protoc ; 9(2): 233-45, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24407352

RESUMEN

Cell therapies enable unprecedented treatment options to replace tissues, destroy tumors and facilitate regeneration. The greatest challenge facing cell therapy is the inability to control the fate and function of cells after transplantation. We have developed an approach to control cell phenotype in vitro and after transplantation by engineering cells with intracellular depots that continuously release phenotype-altering agents for days to weeks. The platform enables control of cells' secretome, viability, proliferation and differentiation, and the platform can be used to deliver drugs or other factors (e.g., dexamethasone, rhodamine and iron oxide) to the cell's microenvironment. The preparation, efficient internalization and intracellular stabilization of ∼1-µm drug-loaded microparticles are critical for establishing sustained control of cell phenotype. Herein we provide a protocol to generate and characterize micrometer-sized agent-doped poly(lactic-co-glycolic) acid (PLGA) particles by using a single-emulsion evaporation technique (7 h), to uniformly engineer cultured cells (15 h), to confirm particle internalization and to troubleshoot commonly experienced obstacles.


Asunto(s)
Ingeniería Celular/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Sistemas de Liberación de Medicamentos/métodos , Ácido Láctico/metabolismo , Fenotipo , Ácido Poliglicólico/metabolismo , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
14.
PLoS One ; 8(4): e60473, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23565253

RESUMEN

Current clinical therapies for critical-sized bone defects (CSBDs) remain far from ideal. Previous studies have demonstrated that engineering bone tissue using mesenchymal stem cells (MSCs) is feasible. However, this approach is not effective for CSBDs due to inadequate vascularization. In our previous study, we have developed an injectable and porous nano calcium sulfate/alginate (nCS/A) scaffold and demonstrated that nCS/A composition is biocompatible and has proper biodegradability for bone regeneration. Here, we hypothesized that the combination of an injectable and porous nCS/A with bone morphogenetic protein 2 (BMP2) gene-modified MSCs and endothelial progenitor cells (EPCs) could significantly enhance vascularized bone regeneration. Our results demonstrated that delivery of MSCs and EPCs with the injectable nCS/A scaffold did not affect cell viability. Moreover, co-culture of BMP2 gene-modified MSCs and EPCs dramatically increased osteoblast differentiation of MSCs and endothelial differentiation of EPCs in vitro. We further tested the multifunctional bone reconstruction system consisting of an injectable and porous nCS/A scaffold (mimicking the nano-calcium matrix of bone) and BMP2 genetically-engineered MSCs and EPCs in a rat critical-sized (8 mm) caviarial bone defect model. Our in vivo results showed that, compared to the groups of nCS/A, nCS/A+MSCs, nCS/A+MSCs+EPCs and nCS/A+BMP2 gene-modified MSCs, the combination of BMP2 gene -modified MSCs and EPCs in nCS/A dramatically increased the new bone and vascular formation. These results demonstrated that EPCs increase new vascular growth, and that BMP2 gene modification for MSCs and EPCs dramatically promotes bone regeneration. This system could ultimately enable clinicians to better reconstruct the craniofacial bone and avoid donor site morbidity for CSBDs.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Regeneración Ósea/fisiología , Células Endoteliales/citología , Células Madre Mesenquimatosas/citología , Cráneo/citología , Células Madre/citología , Ingeniería de Tejidos/métodos , Animales , Proteína Morfogenética Ósea 2/genética , Supervivencia Celular/fisiología , Inmunohistoquímica , Masculino , Osteoblastos/citología , Osteogénesis/fisiología , Ratas , Andamios del Tejido
16.
J Biomed Mater Res A ; 101(8): 2151-63, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23255285

RESUMEN

Cell-matrix interaction is a key regulator for controlling stem cell fate in regenerative tissue engineering. These interactions are induced and controlled by the nanoscale features of extracellular matrix and are mimicked on synthetic matrices to control cell structure and functions. Recent studies have shown that nanostructured matrices can modulate stem cell behavior and exert specific role in tissue regeneration. In this study, we have demonstrated that nanostructured phase morphology of synthetic matrix can control adhesion, proliferation, organization and migration of human mesenchymal stem cells (MSCs). Nanostructured biodegradable polyurethanes (PU) with segmental composition exhibit biphasic morphology at nanoscale dimensions and can control cellular features of MSCs. Biodegradable PU with polyester soft segment and hard segment composed of aliphatic diisocyanates and dipeptide chain extender were designed to examine the effect polyurethane phase morphology. By altering the polyurethane composition, morphological architecture of PU was modulated and its effect was examined on MSC. Results show that MSCs can sense the nanoscale morphology of biphasic polyurethane matrix to exhibit distinct cellular features and, thus, signifies the relevance of matrix phase morphology. The role of nanostructured phases of a synthetic matrix in controlling cell-matrix interaction provides important insights for regulation of cell behavior on synthetic matrix and, therefore, is an important tool for engineering tissue regeneration.


Asunto(s)
Materiales Biocompatibles/metabolismo , Células Madre Mesenquimatosas/citología , Poliuretanos/metabolismo , Andamios del Tejido/química , Materiales Biocompatibles/química , Adhesión Celular , Línea Celular , Movimiento Celular , Proliferación Celular , Humanos , Células Madre Mesenquimatosas/metabolismo , Poliuretanos/química
17.
Blood ; 118(25): e184-91, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22034631

RESUMEN

One of the greatest challenges in cell therapy is to minimally invasively deliver a large quantity of viable cells to a tissue of interest with high engraftment efficiency. Low and inefficient homing of systemically delivered mesenchymal stem cells (MSCs), for example, is thought to be a major limitation of existing MSC-based therapeutic approaches, caused predominantly by inadequate expression of cell surface adhesion receptors. Using a platform approach that preserves the MSC phenotype and does not require genetic manipulation, we modified the surface of MSCs with a nanometer-scale polymer construct containing sialyl Lewis(x) (sLe(x)) that is found on the surface of leukocytes and mediates cell rolling within inflamed tissue. The sLe(x) engineered MSCs exhibited a robust rolling response on inflamed endothelium in vivo and homed to inflamed tissue with higher efficiency compared with native MSCs. The modular approach described herein offers a simple method to potentially target any cell type to specific tissues via the circulation.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Oligosacáridos/química , Animales , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Células HL-60 , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Integrina beta1/metabolismo , Células Madre Mesenquimatosas/química , Ratones , Ratones Endogámicos BALB C , Selectinas/metabolismo , Antígeno Sialil Lewis X , Antígenos Thy-1/metabolismo , Trasplante Heterólogo
18.
Nat Nanotechnol ; 6(8): 524-31, 2011 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-21765401

RESUMEN

The ability to explore cell signalling and cell-to-cell communication is essential for understanding cell biology and developing effective therapeutics. However, it is not yet possible to monitor the interaction of cells with their environments in real time. Here, we show that a fluorescent sensor attached to a cell membrane can detect signalling molecules in the cellular environment. The sensor is an aptamer (a short length of single-stranded DNA) that binds to platelet-derived growth factor (PDGF) and contains a pair of fluorescent dyes. When bound to PDGF, the aptamer changes conformation and the dyes come closer to each other, producing a signal. The sensor, which is covalently attached to the membranes of mesenchymal stem cells, can quantitatively detect with high spatial and temporal resolution PDGF that is added in cell culture medium or secreted by neighbouring cells. The engineered stem cells retain their ability to find their way to the bone marrow and can be monitored in vivo at the single-cell level using intravital microscopy.


Asunto(s)
Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/métodos , Comunicación Celular/fisiología , Membrana Celular/metabolismo , Análisis de la Célula Individual/instrumentación , Análisis de la Célula Individual/métodos , Animales , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/metabolismo , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Colorantes Fluorescentes , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Imagen Molecular , Factor de Crecimiento Derivado de Plaquetas/análisis , Factor de Crecimiento Derivado de Plaquetas/metabolismo
19.
FASEB J ; 25(9): 3045-56, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21653192

RESUMEN

Nature has evolved effective cell adhesion mechanisms to deliver inflammatory cells to inflamed tissue; however, many culture-expanded therapeutic cells are incapable of targeting diseased tissues following systemic infusion, which represents a great challenge in cell therapy. Our aim was to develop simple approaches to program cell-cell interactions that would otherwise not exist toward cell targeting and understanding the complex biology of cell-cell interactions. We employed a chemistry approach to engineer P- or L-selectin binding nucleic acid aptamers onto mesenchymal stem cells (MSCs) to enable them to engage inflamed endothelial cells and leukocytes, respectively. We show for the first time that engineered cells with a single artificial adhesion ligand can recapitulate 3 critical cell interactions in the inflammatory cell adhesion cascade under dynamic flow conditions. Aptamer-engineered MSCs adhered on respective selectin surfaces under static conditions >10 times more efficiently than controls including scrambled-DNA modified MSCs. Significantly, engineered MSCs can be directly captured from the flow stream by selectin surfaces or selectin-expressing cells under flow conditions (≤2dyn/cm²). The simple chemistry approach and the versatility of aptamers permit the concept of engineered cell-cell interactions to be generically applicable for targeting cells to diseased tissues and elucidating the biology of cell-cell interactions.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Células Endoteliales/fisiología , Inflamación/metabolismo , Células Madre Mesenquimatosas/fisiología , Animales , Células Cultivadas , Selectina L/metabolismo , Selectina-P/metabolismo , Propiedades de Superficie
20.
Methods Mol Biol ; 698: 505-23, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21431540

RESUMEN

By leveraging the capacity to promote regeneration, stem cell therapies offer enormous hope for solving some of the most tragic illnesses, diseases, and tissue defects world-wide. However, a significant barrier to the effective implementation of cell therapies is the inability to target a large quantity of viable cells with high efficiency to tissues of interest. Systemic infusion is desired as it minimizes the invasiveness of cell therapy, and maximizes practical aspects of repeated doses. However, cell types such as mesenchymal stem cells exhibit a poor homing capability or lose their capacity to home following culture expansion (i.e. FASEB J 21:3197-3207, 2007; Circulation 108:863-868, 2003; Stroke: A Journal of Cerebral Circulation 32:1005-1011; Blood 104:3581-3587, 2004). To address this challenge, we have developed a simple platform technology to chemically attach cell adhesion molecules to the cell surface to improve the homing efficiency to specific tissues. This chemical approach involves a stepwise process including (1) treatment of cells with sulfonated biotinyl-N-hydroxy-succinimide to introduce biotin groups on the cell surface, (2) addition of streptavidin that binds to the biotin on the cell surface and presents unoccupied binding sites, and (3) attachment of biotinylated targeting ligands that promote adhesive interactions with vascular endothelium. Specifically, in our model system, a biotinylated cell rolling ligand, sialyl Lewisx (SLeX), found on the surface of leukocytes (i.e., the active site of the P-selectin glycoprotein ligand (PSGL-1)), is conjugated on MSC surface. The SLeX engineered MSCs exhibit a rolling response on a P-selectin coated substrate under shear stress conditions. This indicates that this approach can be used to potentially target P-selectin expressing endothelium in the more marrow or at sites of inflammation. Importantly, the surface modification has no adverse impact on MSCs' native phenotype including their multilineage differentiation capacity, viability, proliferation, and adhesion kinetics. We anticipate that the present approach to covalently modify the cell surface and immobilize required ligands is not limited to MSCs or the SLeX ligand. Therefore, this technology should have broad implications on cell therapies that utilize systemic administration and require targeting of cells to specific tissues. The approach may also be useful to promote specific cell-cell interactions. In this protocol, we describe the conjugation of SLeX on MSC surface and methods to study cell rolling behaviors of SLeX-modified MSCs on a P-selectin coated substrate using an in vitro flow chamber assay. We also provide a brief description of cell characterization assays that can be used to examine the impact of the chemical modification regimen.


Asunto(s)
Ingeniería/métodos , Células Madre Mesenquimatosas/química , Adipogénesis , Animales , Biotinilación , Adhesión Celular , Técnicas de Cultivo de Célula , Proliferación Celular , Supervivencia Celular , Citometría de Flujo , Humanos , Cinética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Imagen Molecular , Osteogénesis , Selectina-P/metabolismo , Coloración y Etiquetado
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA