Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Immunother Cancer ; 11(10)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37793853

RESUMEN

BACKGROUND: SGN-B7H4V is a novel investigational vedotin antibody-drug conjugate (ADC) comprising a B7-H4-directed human monoclonal antibody conjugated to the cytotoxic payload monomethyl auristatin E (MMAE) via a protease-cleavable maleimidocaproyl valine citrulline (mc-vc) linker. This vedotin linker-payload system has been clinically validated in multiple Food and Drug Administration approved agents including brentuximab vedotin, enfortumab vedotin, and tisotumab vedotin. B7-H4 is an immune checkpoint ligand with elevated expression on a variety of solid tumors, including breast, ovarian, and endometrial tumors, and limited normal tissue expression. SGN-B7H4V is designed to induce direct cytotoxicity against target cells by binding to B7-H4 on the surface of target cells and releasing the cytotoxic payload MMAE upon internalization of the B7-H4/ADC complex. METHODS: B7-H4 expression was characterized by immunohistochemistry across multiple solid tumor types. The ability of SGN-B7H4V to kill B7-H4-expressing tumor cells in vitro and in vivo in a variety of xenograft tumor models was also evaluated. Finally, the antitumor activity of SGN-B7H4V as monotherapy and in combination with an anti-programmed cell death-1 (PD-1) agent was evaluated using an immunocompetent murine B7-H4-expressing Renca tumor model. RESULTS: Immunohistochemistry confirmed B7-H4 expression across multiple solid tumors, with the highest prevalence in breast, endometrial, and ovarian tumors. In vitro, SGN-B7H4V killed B7-H4-expressing tumor cells by MMAE-mediated direct cytotoxicity and antibody-mediated effector functions including antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis. In vivo, SGN-B7H4V demonstrated strong antitumor activity in multiple xenograft models of breast and ovarian cancer, including xenograft tumors with heterogeneous B7-H4 expression, consistent with the ability of vedotin ADCs to elicit a bystander effect. In an immunocompetent murine B7-H4-expressing tumor model, SGN-B7H4V drove robust antitumor activity as a monotherapy that was enhanced when combined with an anti-PD-1 agent. CONCLUSION: The immune checkpoint ligand B7-H4 is a promising molecular target expressed by multiple solid tumors. SGN-B7H4V demonstrates robust antitumor activity in preclinical models through multiple potential mechanisms. Altogether, these preclinical data support the evaluation of SGN-B7H4V as a monotherapy in the ongoing phase 1 study of SGN-B7H4V in advanced solid tumors (NCT05194072) and potential future clinical combinations with immunotherapies.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Animales , Humanos , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Línea Celular Tumoral , Modelos Animales de Enfermedad , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Inmunoconjugados/química , Inmunohistoquímica , Ligandos
2.
PLoS One ; 16(9): e0257094, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34506571

RESUMEN

Although the Trans-Himalayan region (THR) is an important endemic and rendezvous area of peste des petits ruminants (PPR), monitoring and prevention measurements are difficult to execute because of the rough geographical conditions. Besides, a heterogeneous breeding system and the poor veterinary service of susceptible animals compound the existing problems. Here, we propose a forecasting system to define the key points of PPR prevention and aid the countries in saving time, labor, and products to achieve the goal of the global eradication project of PPR. The spatial distribution of PPR was predicted in the THR for the first time using a niche model that was constructed with a combination of eco-geographical, anthropoid, meteorological, and host variables. The transboundary least-cost paths (LCPs) of small ruminants in the THR were also calculated. Our results reveal that the low-elevation area of the THR had a higher PPR risk and was mainly dominated by human variables. The high-elevation area had lower risk and was mainly dominated by natural variables. Eight LCPs representing corridors among India, Nepal, Bhutan, Bangladesh, and China were obtained. This confirmed the potential risk of transboundary communication by relying on PPR contamination on the grasslands for the first time. The predicted potential risk communication between the two livestock systems and landscapes (high and low elevation) might play a role in driving PPR transboundary transmission.


Asunto(s)
Ecosistema , Ganado/virología , Peste de los Pequeños Rumiantes/epidemiología , Peste de los Pequeños Rumiantes/transmisión , Altitud , Animales , Geografía , Modelos Biológicos , Reproducibilidad de los Resultados , Factores de Riesgo
3.
J Immunol Methods ; 492: 112955, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33383062

RESUMEN

Identifying engineered T cells in situ is important to understand the location, persistence, and phenotype of these cells in patients after adoptive T cell therapy. While engineered cells are routinely characterized in fresh tissue or blood from patients by flow cytometry, it is difficult to distinguish them from endogenous cells in formalin-fixed, paraffin-embedded (FFPE) tissue biopsies. To overcome this limitation, we have developed a method for characterizing engineered T cells in fixed tissue using in situ hybridization (ISH) to the woodchuck hepatitis post-transcriptional regulatory element (WPRE) common in many lentiviral vectors used to transduce chimeric antigen receptor T (CAR-T) and T cell receptor T (TCR-T) cells, coupled with alternative permeabilization conditions that allows subsequent multiplex immunohistochemical (mIHC) staining within the same image. This new method provides the ability to mark the cells by ISH, and simultaneously stain for cell-associated proteins to immunophenotype CAR/TCR modified T cells within tumors, as well as assess potential roles of these cells in on-target/off-tumor toxicity in other tissue.


Asunto(s)
Inmunohistoquímica/métodos , Inmunofenotipificación/métodos , Receptores Quiméricos de Antígenos/análisis , Linfocitos T/inmunología , Animales , Biopsia , Ingeniería Celular , Técnicas de Cocultivo , Vectores Genéticos/genética , Virus de la Hepatitis B de la Marmota/genética , Humanos , Hibridación Fluorescente in Situ , Lentivirus/genética , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Adhesión en Parafina , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Piel/citología , Piel/inmunología , Piel/patología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Fijación del Tejido , Transducción Genética , Quimera por Trasplante
4.
Nat Med ; 22(8): 933-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27322743

RESUMEN

Individuals who have mutations in the breast-cancer-susceptibility gene BRCA1 (hereafter referred to as BRCA1-mutation carriers) frequently undergo prophylactic mastectomy to minimize their risk of breast cancer. The identification of an effective prevention therapy therefore remains a 'holy grail' for the field. Precancerous BRCA1(mut/+) tissue harbors an aberrant population of luminal progenitor cells, and deregulated progesterone signaling has been implicated in BRCA1-associated oncogenesis. Coupled with the findings that tumor necrosis factor superfamily member 11 (TNFSF11; also known as RANKL) is a key paracrine effector of progesterone signaling and that RANKL and its receptor TNFRSF11A (also known as RANK) contribute to mammary tumorigenesis, we investigated a role for this pathway in the pre-neoplastic phase of BRCA1-mutation carriers. We identified two subsets of luminal progenitors (RANK(+) and RANK(-)) in histologically normal tissue of BRCA1-mutation carriers and showed that RANK(+) cells are highly proliferative, have grossly aberrant DNA repair and bear a molecular signature similar to that of basal-like breast cancer. These data suggest that RANK(+) and not RANK(-) progenitors are a key target population in these women. Inhibition of RANKL signaling by treatment with denosumab in three-dimensional breast organoids derived from pre-neoplastic BRCA1(mut/+) tissue attenuated progesterone-induced proliferation. Notably, proliferation was markedly reduced in breast biopsies from BRCA1-mutation carriers who were treated with denosumab. Furthermore, inhibition of RANKL in a Brca1-deficient mouse model substantially curtailed mammary tumorigenesis. Taken together, these findings identify a targetable pathway in a putative cell-of-origin population in BRCA1-mutation carriers and implicate RANKL blockade as a promising strategy in the prevention of breast cancer.


Asunto(s)
Proteína BRCA1/genética , Conservadores de la Densidad Ósea/farmacología , Neoplasias de la Mama/prevención & control , Mama/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Denosumab/farmacología , Organoides/efectos de los fármacos , Ligando RANK/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Células Madre/efectos de los fármacos , Animales , Conservadores de la Densidad Ósea/uso terapéutico , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinogénesis/genética , Reparación del ADN , Denosumab/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Heterocigoto , Humanos , Inmunohistoquímica , Ratones , Terapia Molecular Dirigida , Mutación , Trasplante de Neoplasias , Organoides/metabolismo , Pilocarpina/análogos & derivados , Mastectomía Profiláctica , Ligando RANK/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Proteínas Supresoras de Tumor , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Cell Physiol ; 230(9): 2098-107, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25640014

RESUMEN

Cell loss due to apoptosis induced by oxidative stress is a major hurdle for endothelial progenitor cells (EPCs)-based therapy. Sirtuin 1 (SIRT1) plays important roles in many pathophysiological processes by deacetylating various substrates, including forkhead transcription factor (FOXO). However, after deacetylation, the fate of FOXO protein remains to be explored. In the present study, we investigated whether SIRT1 exerted a protective effect on hydrogen peroxide (H(2)O(2))-induced EPCs apoptosis and, if so, what the underlying mechanism might be. EPCs were isolated and obtained from human umbilical cord blood by density gradient centrifugation and identified by morphology, tube formation ability, cell surface markers, and the ability to take up acetylated low-density lipoprotein (Dil-Ac-LDL) and bind ulex europaeus agglutinin 1 (FITC-UEA-1). Immunofluorescence showed that SIRT1 is localized in the nucleus of EPCs in the presence or absence of H(2)O(2). SIRT1 protein level in EPCs was increased by the treatment with H(2)O(2) for 24 h. Incubation of EPCs with H(2)O(2) dose dependently induced EPCs apoptosis. SIRT1 overexpression reduced the rate of EPCs apoptosis induced by H(2)O(2), whereas SIRT1 downregulation and EX527, a specific SIRT1 inhibitor, exerted the opposite effect. SIRT1 overexpression decreased the total FOXO3a protein expression, whereas SIRT1 downregulation and EX527 increased the amount of FOXO3a protein. SIRT1 reduced FOXO3a transcriptional activity according to Bim expression. Co-immunoprecipitation assay showed that SIRT1 could bind to FOXO3a, reduce its acetylation level and increase its ubiquitination level. To sum up, our work demonstrated that SIRT1 had a pivotally protective role in the regulation of EPCs apoptosis induced by H(2)O(2) and that SIRT1 protected against apoptosis by inhibiting FOXO3a via FOXO3a ubiquitination and subsequent degradation.


Asunto(s)
Apoptosis/efectos de los fármacos , Factores de Transcripción Forkhead/genética , Estrés Oxidativo/genética , Sirtuina 1/genética , Células Progenitoras Endoteliales/efectos de los fármacos , Sangre Fetal , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Humanos , Peróxido de Hidrógeno/administración & dosificación , Proteolisis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Ubiquitinación/genética
6.
J Bone Oncol ; 4(3): 59-68, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27556008

RESUMEN

Receptor activator of nuclear factor kappa-B ligand (RANKL) is an essential mediator of osteoclast formation, function and survival. In patients with solid tumor metastasis to the bone, targeting the bone microenvironment by inhibition of RANKL using denosumab, a fully human monoclonal antibody (mAb) specific to RANKL, has been demonstrated to prevent tumor-induced osteolysis and subsequent skeletal complications. Recently, a prominent functional role for the RANKL pathway has emerged in the primary bone tumor giant cell tumor of bone (GCTB). Expression of both RANKL and RANK is extremely high in GCTB tumors and denosumab treatment was associated with tumor regression and reduced tumor-associated bone lysis in GCTB patients. In order to address the potential role of the RANKL pathway in another primary bone tumor, this study assessed human RANKL and RANK expression in human primary osteosarcoma (OS) using specific mAbs, validated and optimized for immunohistochemistry (IHC) or flow cytometry. Our results demonstrate RANKL expression was observed in the tumor element in 68% of human OS using IHC. However, the staining intensity was relatively low and only 37% (29/79) of samples exhibited≥10% RANKL positive tumor cells. RANK expression was not observed in OS tumor cells. In contrast, RANK expression was clearly observed in other cells within OS samples, including the myeloid osteoclast precursor compartment, osteoclasts and in giant osteoclast cells. The intensity and frequency of RANKL and RANK staining in OS samples were substantially less than that observed in GCTB samples. The observation that RANKL is expressed in OS cells themselves suggests that these tumors may mediate an osteoclastic response, and anti-RANKL therapy may potentially be protective against bone pathologies in OS. However, the absence of RANK expression in primary human OS cells suggests that any autocrine RANKL/RANK signaling in human OS tumor cells is not operative, and anti-RANKL therapy would not directly affect the tumor.

7.
Oncol Lett ; 8(5): 2096-2102, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25295097

RESUMEN

Pancreatic cancer (PC) is the fourth leading cause of cancer-related mortality in the United States. There is no effective serum biomarker for the early diagnosis of PC at present. Although serum UL16-binding protein 2 (ULBP2) and macrophage inhibitory cytokine-1 (MIC-1) levels are reported to be elevated in PC patients, the diagnostic and prognostic value of ULBP2 and MIC-1 alone or in combination remains unknown. The aim of the present case-control study was to compare the diagnostic value of ULBP2, MIC-1 and carbohydrate antigen 19-9 (CA19-9) in 359 serum samples, consisting of 152 cases of PC, 20 cases of pre-pancreatic cancer, 91 cases of chronic pancreatitis (CP) and 96 normal controls (NC). All patients were followed up for a median of 2 years. It was found that the serum levels of ULBP2, MIC-1 and CA19-9 were significantly higher in the PC patients compared with those in the NC group. In distinguishing PC from the CP, the highest sensitivity and specificity were ULBP2 (0.878) and CA19-9 (0.816), respectively. The area under the receiver operating characteristic curve of ULBP2 was 0.923, which was the highest of the three biomarkers. MIC-1 was the optimal choice for the diagnosis of early-stage PC (area under the curve, 0.831). Overall, MIC-1 in combination with ULBP2 improved the diagnostic accuracy in differentiating PC from CP and NC. In addition, a higher level of MIC-1 was correlated with a poorer prognosis, as calculated by the Kaplan-Meier test (P=0.039). Patients with serum MIC-1 levels of ≥1,932 ng/ml had a median survival time of 15.62±2.44 months (mean ± standard deviation) vs. 18.66±2.43 months in patients with a lower level of MIC-1. Overall, combined detection of serum MIC-1 and ULBP2 improved the diagnostic accuracy in differentiating PC from CP and NC, and serum MIC-1 level alone was a predictor of survival in the patients with PC.

8.
Am J Physiol Gastrointest Liver Physiol ; 306(9): G788-95, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24627565

RESUMEN

Communication between neurons and glia in the dorsal root ganglia (DRG) and the central nervous system is critical for nociception. Both glial activation and proinflammatory cytokine induction underlie this communication. We investigated whether satellite glial cell (SGC) and tumor necrosis factor-α (TNF-α) activation in DRG participates in a 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced rat model of visceral hyperalgesia. In TNBS-treated rats, TNF-α expression increased in DRG and was colocalized to SGCs enveloping a given neuron. These SGCs were activated as visualized under electron microscopy: they had more elongated processes projecting into the connective tissue space and more gap junctions. When nerves attached to DRG (L6-S1) were stimulated with a series of electrical stimulations, TNF-α were released from DRG in TNBS-treated animals compared with controls. Using a current clamp, we noted that exogenous TNF-α (2.5 ng/ml) increased DRG neuron activity, and visceral pain behavioral responses were reversed by intrathecal administration of anti-TNF-α (10 µg·kg(-1)·day(-1)). Based on our findings, TNF-α and SGC activation in neuron-glial communication are critical in inflammatory visceral hyperalgesia.


Asunto(s)
Comunicación Celular , Colitis/metabolismo , Colon/inervación , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Mediadores de Inflamación/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Umbral del Dolor , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales/administración & dosificación , Conducta Animal , Comunicación Celular/efectos de los fármacos , Extensiones de la Superficie Celular/inmunología , Extensiones de la Superficie Celular/metabolismo , Células Cultivadas , Colitis/inducido químicamente , Colitis/inmunología , Colitis/fisiopatología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/inmunología , Ganglios Espinales/fisiopatología , Uniones Comunicantes/inmunología , Uniones Comunicantes/metabolismo , Hiperalgesia/inducido químicamente , Hiperalgesia/inmunología , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Masculino , Neuroglía/inmunología , Neuronas/efectos de los fármacos , Neuronas/inmunología , Umbral del Dolor/efectos de los fármacos , Presión , Ratas , Ratas Sprague-Dawley , Ácido Trinitrobencenosulfónico , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba
9.
Breast Cancer Res ; 15(4): R62, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23938070

RESUMEN

INTRODUCTION: Menopausal hormone therapies vary widely in their effects on breast cancer risk, and the mechanisms underlying these differences are unclear. The primary goals of this study were to characterize the mammary gland transcriptional profile of estrogen + progestin therapy in comparison with estrogen-alone or tibolone and investigate pathways of cell proliferation in a postmenopausal primate model. METHODS: Ovariectomized female cynomolgus macaque monkeys were randomized into the following groups: placebo (Con), oral conjugated equine estrogens (CEE), CEE with medroxyprogesterone acetate (MPA) (CEE + MPA), and tibolone given at a low or high dose (Lo or Hi Tib). All study treatment doses represented human clinical dose equivalents and were administered in the diet over a period of 2 years. RESULTS: Treatment with CEE + MPA had the greatest effect on global mRNA profiles and markers of mammary gland proliferation compared to CEE or tibolone treatment. Changes in the transcriptional patterns resulting from the addition of MPA to CEE were related to increased growth factors and decreased estrogen receptor (ER) signaling. Specific genes induced by CEE + MPA treatment included key members of prolactin receptor (PRLR)/signal transducer and activator of transcription 5 (STAT5), epidermal growth factor receptor (EGFR), and receptor activator of nuclear factor kappa B (RANK)/receptor activator of nuclear factor kappa B ligand (RANKL) pathways that were highly associated with breast tissue proliferation. In contrast, tibolone did not affect breast tissue proliferation but did elicit a mixed pattern of ER agonist activity. CONCLUSION: Our findings indicate that estrogen + progestin therapy results in a distinct molecular profile compared to estrogen-alone or tibolone therapy, including upregulation of key growth factor targets associated with mammary carcinogenesis in mouse models. These changes may contribute to the promotional effects of estrogen + progestin therapy on breast cancer risk.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Posmenopausia , Progestinas/metabolismo , Transducción de Señal , Animales , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Análisis por Conglomerados , Células Epiteliales/metabolismo , Moduladores de los Receptores de Estrógeno/farmacología , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Perfilación de la Expresión Génica , Terapia de Reemplazo de Hormonas , Humanos , Inmunohistoquímica , Macaca fascicularis , Glándulas Mamarias Animales/efectos de los fármacos , Norpregnenos/farmacología , Progestinas/farmacología , Ligando RANK/genética , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Microvasc Res ; 90: 48-54, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23920411

RESUMEN

OBJECTIVES: Increased oxidative stress has been suggested to contribute to the functional impairment of endothelial progenitor cells (EPCs). The Forkhead box O transcription factors (FoxOs) are critical regulators involved in various cellular processes including cell apoptosis. Here, we investigated whether FoxOs are required in oxidative stress induced EPC apoptosis. METHODS AND RESULTS: EPCs were cultured from cord blood derived mononuclear cells and treated with hydrogen peroxide (H2O2) for induction of oxidative stress. Incubation with H2O2 dose dependently reduced viability and increased apoptosis in EPCs. Western blotting showed that EPCs predominantly expressed FoxO3a and the expression was markedly increased upon H2O2 treatment. Transduction with adenoviral vectors expressing either a wide-type or a non-phosphorylatable, constitutively active mutant of FoxO3a led to further increased apoptosis of EPCs after H2O2 treatment. Conversely, FoxO3a silencing rescued EPCs from these H2O2 induced deleterious effects. Overexpression of FoxO3a also increased the level of the pro-apoptotic protein Bim, whereas FoxO3a silencing downregulated H2O2 induced Bim expression. Furthermore, Matrigel assay demonstrated that FoxO3a overexpression significantly impaired the tube forming ability of EPCs, whereas its silencing completely protected EPCs from H2O2 induced decrease of capillary formation. CONCLUSIONS: These data suggest that oxidative stress induced impairment of EPC survival is mediated through a FoxO3a dependant mechanism, possibly by transcriptional regulation of Bim. Our data indicate FoxO3a as a potential therapeutic target for improvement of EPC number and function in patients with ischemic heart disease.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Peróxido de Hidrógeno/toxicidad , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Células Madre/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Células Endoteliales/patología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Humanos , Proteínas de la Membrana/metabolismo , Mutación , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Células Madre/patología , Transducción Genética , Transfección
11.
Int J Pharm ; 436(1-2): 88-96, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22789913

RESUMEN

A drug-loaded nanofiber mesh which could achieve time-engineeringed biphasic release was fabricated through sequential electrospinning. The drug to polymer ratio of each single mesh was allocated and designed before the tri-layered meshes were created. The resultant meshes had the following construction: (i) the first drug-loaded mesh (top side), (ii) the second drug-loaded mesh (second side), and (iii) the third drug-loaded mesh (bottom side). The drug release speed and duration were controlled by designing morphological features of the electrospun meshes such as the fiber diameter and mesh thickness. An in vitro release experiment revealed that the tri-layered construction with distinct morphological features of each component mesh can provide biphasic drug release. The time-engineeringed dual release system using the multilayered electrospun nanofiber meshes was proved to be a useful formulation when achieving controlled drug release at different times.


Asunto(s)
Celulosa/análogos & derivados , Sistemas de Liberación de Medicamentos , Cetoprofeno/química , Nanofibras/química , Polivinilos/química , Pirrolidinas/química , Celulosa/química , Química Farmacéutica/métodos , Preparaciones de Acción Retardada/química , Microscopía Electrónica de Rastreo , Nanofibras/ultraestructura
12.
J Dig Dis ; 13(4): 225-31, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22435508

RESUMEN

OBJECTIVE: The aim of this study was to study the role of fractalkine (FKN) in the development of severe acute pancreatitis (SAP) in animal model. METHODS: Serum FKN levels in rat model (control, SAP6 h, 16 h, 24 h and 48 h) were determined by enzyme-linked immunosorbent assay. FKN mRNA and protein levels in the pancreas tissue were measured by reverse transcription polymerase chain reaction (RT-PCR), Western blot and immunohistochemistry. RESULTS: Serum FKN level in the SAP rat increased significantly (P < 0.05 compared with the control group). FKN mRNA and protein levels in pancreas and lung increased significantly and reached the peak at 16 h after the induction of SAP, while those in kidney reached the peak at 48 h. Immunohistochemistry showed the overexpression of FKN in pancreas, lung and kidney tissue. CONCLUSION: FKN involves in the progression of SAP and might be a valuable marker for the assessment of SAP.


Asunto(s)
Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Páncreas/metabolismo , Pancreatitis/metabolismo , Índice de Severidad de la Enfermedad , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Expresión Génica/fisiología , Riñón/metabolismo , Pulmón/metabolismo , Pancreatitis/diagnóstico , Pancreatitis/mortalidad , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
13.
Inflammation ; 35(3): 1023-30, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22213034

RESUMEN

Based on the function of chemokine fractalkine (FKN), acting as both adhesion and chemoattractant, FKN plays a role in acute inflammatory response. In this study, we investigated the mechanism of FKN mediated upregulation inflammation in severe acute pancreatitis (SAP) rat models. Western blot, reverse transcriptase-polymerase chain reaction, and immunofluorescence demonstrated that FKN and its receptor CX3CR1 were overexpressed in cerulein-stimulated AR42J cells. AG490 and FKN-siRNA inhibited activation of Janus kinase/signal transducers and activators of transcription (Jak/Stat) in cerulein-stimulated AR42J cells. Following exposure AG490 and FKN-siRNA inhibited tumor necrosis factor-alpha expression by enzyme-linked immunosorbent assay and immunohistochemistry in vivo the SAP rat models. These results showed FKN and CX3CR1 were involved inflammatory response in cerulein-stimulated AR42J cells. FKN upregulates inflammation through CX3CR1 and the Jak/Stat pathway in SAP rat models.


Asunto(s)
Quimiocina CX3CL1/metabolismo , Quinasas Janus/metabolismo , Pancreatitis/metabolismo , Receptores de Quimiocina/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C , Línea Celular , Ceruletida/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Mediadores de Inflamación , Quinasas Janus/antagonistas & inhibidores , Pancreatitis/inducido químicamente , Interferencia de ARN , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Receptores de Quimiocina/genética , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
14.
Anal Biochem ; 423(1): 26-35, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22285698

RESUMEN

The development of protein chips has suffered from problems regarding long-term protein stability and activity. We present a protein sensor surface for immunodetection that is prepared by a DNA-directed protein immobilization method on a mixed self-assembled monolayer (SAM). By this approach, an immobilized single-stranded DNA (ssDNA) surface can be transferred/modified into a protein chip by flowing in ssDNA-conjugated protein when the protein chip measurement is needed. Therefore, the long-term stability of the protein chip will not be a problem for various applications. We tried various compositions for the SAM layer, the length of the ssDNA spacer, the end-point nucleotide composition, and the processes of ssDNA immobilization of the SAM for an optimized condition for shifting the DNA chip to a protein chip. The evaluations were made by using surface plasmon resonance. Our results indicated that a 50:1 ratio of oligo(ethylene glycol) (OEG)/COOH-terminated OEG and DNA sequences with 20mer are the best conditions found here for making a protein chip via a DNA-directed immobilization (DDI) method. The designed end-point nucleotide composition contains a few guanines or cytosines, and ssDNA immobilization of the SAM by dehybridizing immobilized double-stranded DNA (dsDNA) can improve the hybridization efficiency.


Asunto(s)
ADN de Cadena Simple/química , Glicol de Etileno/química , Inmunoensayo/métodos , Proteínas/análisis , Resonancia por Plasmón de Superficie , Anticuerpos/química , Anticuerpos/inmunología , Secuencia de Bases , ADN de Cadena Simple/metabolismo , Muramidasa/análisis , Muramidasa/inmunología , Análisis por Matrices de Proteínas , Proteínas/inmunología , Albúmina Sérica/análisis , Albúmina Sérica/inmunología
15.
West Indian med. j ; 60(2): 107-113, Mar. 2011. ilus
Artículo en Inglés | LILACS | ID: lil-672734

RESUMEN

BACKGROUND AND AIMS: Interferon-α (IFN-α) treatment is associated with up-regulation of epidermal growth factor receptor (EGFR) expression and marked growth inhibition of colon cancer cell lines. We aimed to determine the effect of combining IFN-α and gefitinib in the growth of human colon cancer cell lines. METHODS: Two human colon cancer cell lines SW480 and LOVO were treated with IFN-α alone or gefitinib alone or IFN-α plus gefitinib. Proliferation of colon cancer cells was measured by methyl thiazolyl tetrazolium (MTT) assay; the apoptosis rate was analysed by flow cytometry (FCM). The expression of XIAP, XAF1 mRNA was detected by RT-PCR and the expression of XIAP, XAF1 protein was detected by western blotting. RESULTS: Methyl thiazolyl tetrazolium showed that IFN-α, gefitinib and IFN-α plus gefitinib significantly inhibited SW480 and LOVO cells in a dose-dependent manner (p < 0.05). The FCM revealed that IFN α, gefitinib and IFN-α plus gefitinib could markedly upgrade the apoptosis rate (p < 0.05). The expression of XIAP mRNA down-regulated markedly (p < 0.05) while the expression of XAF1 mRNA up-regulated significantly (p < 0.05). The expression of XIAP protein was down-regulated markedly (p < 0.05) while the expression of XAF1 protein was up-regulated significantly (p < 0.05). CONCLUSION: IFN-α promotes the antiproliferaative effect of gefitinib on human colon cancer cell lines and the mechanism may be related to up-regulation expression of EGFR by IFN-α.


ANTECEDENTES Y OBJETIVOS: El tratamiento con interferón α (IFN-α) se halla asociado con la regulación por incremento de la expresión del receptor del factor de crecimiento epidérmico y la acentuada inhibición del crecimiento de las líneas celulares del cáncer colorrectal. El presente trabajo tuvo por objetivo determinar el efecto que se produce al combinar el IFN-α y el gefitinib en el crecimiento de las líneas celulares del cáncer de colon. MÉTODOS: Dos líneas celulares de cáncer del colon en humanos - SW480 y LOVO - fueron tratadas con IFN-α solamente, gefitinib solamente, o IFN-α más gefitinib. La proliferación de las células cancerosas del colon se midió mediante ensayo de metil tiazolil tetrazolio (MTT); la tasa de apoptosis se analizó mediante citometría de flujo (CMF); la expresión de XIAP/XAF1 mRNA fue detectada mediante RT-PCR y la expresión de la proteína XIAP/XAF1 fue detectada mediante inmunoblot (western blot). RESULTADOS: El MTT mostró que el IFN-α, el gefitinib, y el IFN-α más gefitinib inhibían de forma significativa las células SW480 y LOVO en dependencia de la dosis (p < 0.05). La CMF reveló que el IFN-α, el gefitinib, y el IFN-α más gefitinib podían aumentar notablemente la tasa de apoptosis (p < 0.05). La expresión de XIAP mRNA tuvo una marcada regulación por decremento (p < 0.05) mientras que la expresión de XAF1 mRNA tuvo una significativa regulación por incremento (p < 0.05); la expresión de la proteína XIAP fue notablemente regulada por decremento (p < 0.05) mientras que la expresión de la proteína XAF1 fue regulada por incremento de manera significativa (p < 0.05). CONCLUSIÓN: El IFN-α promueve el efecto antiproliferativo del gefitinib sobre las líneas celulares del cáncer colorrectal, y el mecanismo puede hallarse relacionado con la expresión de la regulación por incremento del EGFR mediante el IFN-α.


Asunto(s)
Humanos , Antineoplásicos/farmacología , Neoplasias del Colon/patología , Interferón-alfa/farmacología , Quinazolinas/farmacología , Receptores ErbB/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Relación Dosis-Respuesta a Droga , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores ErbB/metabolismo
16.
J Biomater Sci Polym Ed ; 21(12): 1545-58, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20537240

RESUMEN

Time-lapse digital images were taken every 30 s of PC12 cells cultured on polystyrene dishes, collagen-coated dishes and poly(L-lysine)-coated dishes in high-serum medium, low-serum medium and neurite outgrowth factor (NGF)-containing medium to investigate their diffusion coefficients (i.e., self-diffusion coefficients), D and specific movement (i.e., specific frequent movement) using image analysis and Fast Fourier Transform (FFT) analysis. D for these cells was found to fluctuate as a function of time, D varying between 0 and 0.08 microm(2)/s. The trend observed upon examination of average D values was: D in high-serum medium > or = D in low-serum medium > or = D in NGF-containing medium. Image analysis and FFT analysis of single cells cultured on polymeric dishes in these three media did not have any specific frequency of cell movement between 0 and 0.0167 Hz. The high diffusion coefficient and high amplitude of power spectra of PC12 cells in high-serum medium might be attributed to the high energy necessary for their continual suppression of the mitogen-activated protein kinase (MAPK) cascade and for them to maintain their undifferentiated state.


Asunto(s)
Movimiento Celular , Procesamiento de Imagen Asistido por Computador/métodos , Imagen Molecular , Polímeros , Animales , Colágeno , Difusión , Análisis de Fourier , Luz , Microscopía , Células PC12 , Polilisina , Poliestirenos , Ratas , Análisis Espectral , Propiedades de Superficie , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...