Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mil Psychol ; 36(2): 158-167, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38377251

RESUMEN

The military environment involves stressful situations that may trigger or aggravate suicidal behaviors, such as suicide attempts (SAs), which significantly increase the likelihood of future suicide. This cross-sectional study aims to assess risk factors for severe SAs and non-suicidal self-injury (NSSI) among Israel Defense Forces (IDF) soldiers. Data were retrieved from an IDF computerized self-harm surveillance database and were based on the criteria of the Columbia Suicide Severity Rating Scale (C-SSRS) and the Suicide Attempt Self-Injury Interview (SASII). The cohort included all 1,238 occurrences of self-harm behavior, during 2017-2021. Other investigated variables included adjustment difficulty (AD, as per IDF definition) and psychiatric diagnosis (PD) as reported by mental health officers (MHOs) during recruitment. Higher rates of adjustment difficulties were found among soldiers who had conducted NSSIs. Higher rates of previous psychiatric diagnoses were found among individuals with SAs, and their risk of dying by suicide during military service was twice as high (OR = 2.356; p < .001). If the latter also served in a combat unit, the risk was almost fourfold (OR = .3.860; p < .001). The current study demonstrates a clear difference between IDF soldiers who conduct NSSI vs. those conducting SA with regard to adjustment difficulty (as per IDF definition) and PD.


Asunto(s)
Personal Militar , Conducta Autodestructiva , Humanos , Personal Militar/psicología , Israel/epidemiología , Estudios Transversales , Conducta Autodestructiva/epidemiología , Factores de Riesgo
2.
Int J Mol Sci ; 23(22)2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36430552

RESUMEN

Tumor Treating Fields (TTFields) are electric fields that exert physical forces to disrupt cellular processes critical for cancer cell viability and tumor progression. TTFields induce anti-mitotic effects through the disruption of the mitotic spindle and abnormal chromosome segregation, which trigger several forms of cell death, including immunogenic cell death (ICD). The efficacy of TTFields concomitant with anti-programmed death-1 (anti-PD-1) treatment was previously shown in vivo and is currently under clinical investigation. Here, the potential of TTFields concomitant with anti- PD-1/anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) or anti-programmed death-ligand 1 (anti-PD-L1) immune checkpoint inhibitors (ICI) to improve therapeutic efficacy was examined in lung tumor-bearing mice. Increased circulating levels of high mobility group box 1 protein (HMGB1) and elevated intratumoral levels of phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α) were found in the TTFields-treated mice, indicative of ICD induction. The concomitant application of TTFields and ICI led to a significant decrease in tumor volume as compared to all other groups. In addition, significant increases in the number of tumor-infiltrating immune cells, specifically cytotoxic T-cells, were observed in the TTFields plus anti-PD-1/anti-CTLA-4 or anti-PD-L1 groups. Correspondingly, cytotoxic T-cells isolated from these tumors showed higher levels of IFN-γ production. Collectively, these results suggest that TTFields have an immunoactivating role that may be leveraged for concomitant treatment with ICI to achieve better tumor control by enhancing antitumor immunity.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Ratones , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Supervivencia Celular/fisiología , Huso Acromático
3.
Cancers (Basel) ; 13(7)2021 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-33801627

RESUMEN

Focused ultrasound (FUS) has shown promise as a non-invasive treatment modality for solid malignancies. FUS targeting to tumors has been shown to initiate pro-inflammatory immune responses within the tumor microenvironment. Pulsed FUS (pFUS) can alter the expression of cytokines, chemokines, trophic factors, cell adhesion molecules, and immune cell phenotypes within tissues. Here, we investigated the molecular and immune cell effects of pFUS on murine B16 melanoma and 4T1 breast cancer flank tumors. Temporal changes following sonication were evaluated by proteomics, RNA-seq, flow-cytometry, and histological analyses. Proteomic profiling revealed molecular changes occurring over 24 h post-pFUS that were consistent with a shift toward inflamed tumor microenvironment. Over 5 days post-pFUS, tumor growth rates were significantly decreased while flow cytometric analysis revealed differences in the temporal migration of immune cells. Transcriptomic analyses following sonication identified differences in gene expression patterns between the two tumor types. Histological analyses further demonstrated reduction of proliferation marker, Ki-67 in 4T1, but not in B16 tumors, and activated cleaved-caspase 3 for apoptosis remained elevated up to 3 days post-pFUS in both tumor types. This study revealed diverse biological mechanisms following pFUS treatment and supports its use as a possible adjuvant to ablative tumor treatment to elicit enhanced anti-tumor responses and slow tumor growth.

4.
JCI Insight ; 5(22)2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33208556

RESUMEN

Successful implantation is associated with a unique spatial pattern of vascular remodeling, characterized by profound peripheral neovascularization surrounding a periembryo avascular niche. We hypothesized that hyaluronan controls the formation of this distinctive vascular pattern encompassing the embryo. This hypothesis was evaluated by genetic modification of hyaluronan metabolism, specifically targeted to embryonic trophoblast cells. The outcome of altered hyaluronan deposition on uterine vascular remodeling and postimplantation development were analyzed by MRI, detailed histological examinations, and RNA sequencing of uterine NK cells. Our experiments revealed that disruption of hyaluronan synthesis, as well as its increased cleavage at the embryonic niche, impaired implantation by induction of decidual vascular permeability, defective vascular sinus folds formation, breach of the maternal-embryo barrier, elevated MMP-9 expression, and interrupted uterine NK cell recruitment and function. Conversely, enhanced deposition of hyaluronan resulted in the expansion of the maternal-embryo barrier and increased diffusion distance, leading to compromised implantation. The deposition of hyaluronan at the embryonic niche is regulated by progesterone-progesterone receptor signaling. These results demonstrate a pivotal role for hyaluronan in successful pregnancy by fine-tuning the periembryo avascular niche and maternal vascular morphogenesis.


Asunto(s)
Decidua/irrigación sanguínea , Implantación del Embrión , Embrión de Mamíferos/fisiología , Ácido Hialurónico/farmacología , Células Asesinas Naturales/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Útero/fisiología , Animales , Diferenciación Celular , Decidua/efectos de los fármacos , Decidua/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Femenino , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Masculino , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos C57BL , Embarazo , Transducción de Señal , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/fisiología , Útero/citología , Útero/efectos de los fármacos , Remodelación Vascular , Viscosuplementos/farmacología
5.
Artículo en Inglés | MEDLINE | ID: mdl-32850728

RESUMEN

Non-ablative ultrasound (US)-based techniques to improve targeted tropism of systemically infused cell therapies, particularly mesenchymal stromal cell (MSC), have gained attention in recent years. Mechanotransduction following targeted US sonications have been shown to modulate tissue microenvironments by upregulating cytokines, chemokines, and trophic factors in addition to vascular cell adhesion molecules (CAM) that are necessary to promote tropism of MSC. While numerous US treatment parameters have demonstrated increased MSC homing, it remains unclear how the different mechanical US forces [i.e., acoustic radiation forces (ARF) or cavitation forces] influence tissue microenvironments. This study sonicated murine muscle tissue with pulsed focused ultrasound (pFUS) at 0.5 or 1.15 MHz each over a range of US intensities. Following sonication, tissue was assayed for the prostaglandins (PG) PGH2 and PGE2 as indicators of microenvironmental changes that would support MSC tropism. PGH2 and PGE2 levels were correlated to physical pFUS parameters and acoustic emissions measured by hydrophone. While ARF (pFUS with absence of cavitation signatures) was sufficient to increase PGH2 and PGE2, non-linear curve fitting revealed a frequency-independent relationship between prostaglandin production and mechanical index (MI), which accounts for increased cavitation probabilities of lower frequencies. The prostaglandin data suggested molecular changes in muscle would be particularly sensitive to cavitation. Therefore, low-intensity pulsed ultrasound (LIPUS) at 1 MHz was administered with low ARF (MI = 0.2) in combination with intravenous (IV) infusions of microbubble (MB) contrast agents. This combination upregulated prostaglandins and CAM without ultrasound-mediated microbubble destruction and ultimately promoted tropism of IV-infused MSC. This study revealed that accentuating non-destructive MB cavitation by US using parameters similar to diagnostic US contrast imaging increased MSC homing. Such approaches are particularly attractive to overcome clinical translation barriers of many still-experimental US parameters used in previous stem cell tropism studies.

6.
Cancers (Basel) ; 12(2)2020 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-32033171

RESUMEN

Image-guided focused ultrasound (FUS) has been successfully employed as an ablative treatment for solid malignancies by exposing immune cells to tumor debris/antigens, consequently inducing an immune response within the tumor microenvironment (TME). To date, immunomodulation effects of non-ablative pulsed-FUS (pFUS) on the TME are poorly understood. In this study, the temporal differences of cytokines, chemokines, and trophic factors (CCTFs) and immune cell populations induced by pFUS were interrogated in murine B16 melanoma or 4T1 breast cancer cells subcutaneously inoculated into C57BL/6 or BALB/c mice. Natural history growth characteristics during the course of 11 days showed a progressive increase in size for both tumors, and proteomic analysis revealed a shift toward an immunosuppressive TME. With respect to tumor natural growth, pFUS applied to tumors on days 1, 5, or 9 demonstrated a decrease in the growth rate 24 h post-sonication. Flow cytometry analysis of tumors, LNs, and Sp, as well as CCTF profiles, relative DNA damage, and adaptive T-cell localization within tumors, demonstrated dynamic innate and adaptive immune-modulation following pFUS in early time points of B16 tumors and in advanced 4T1 tumors. These results provide insight into the temporal dynamics in the treatment-associated TME, which could be used to evaluate an immunomodulatory approach in different tumor types.

7.
Ultrasound Med Biol ; 45(12): 3232-3245, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31530419

RESUMEN

Non-ablative pulsed focused ultrasound (pFUS) targets non-thermal forces that activate local molecular and cellular immune responses. Optimal parameters to stimulate immunotherapeutic tumor microenvironments (TME) and responses in different tumor types remain uninvestigated. Flank B16 murine melanoma and 4T1 breast tumors received 1 MHz pFUS at 1-8 MPa peak negative pressures (PNP) and were analyzed 24 hr post-sonication. Necrosis or hemorrhage were unaltered in both tumors, but pFUS induced DNA strand breaks in tumor cells at PNP ≥6 MPa. pFUS at >4 MPa suppressed anti-inflammatory cytokines in B16 tumors. pFUS to 4T1 tumors decreased anti-inflammatory cytokines and increased pro-inflammatory cytokines and cell adhesion molecules. pFUS at 6 MPa increased calreticulin and alterations in check-point proteins along with tumoral and splenic immune cell changes that could be consistent with a shift towards an anti-TME. pFUS-induced TME alterations shows promise in generating anti-tumor immune responses, but non-uniform responses between tumor types require additional investigation to assess pFUS as a suitable anti-tumor therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Melanoma/metabolismo , Proteómica/métodos , Microambiente Tumoral , Ondas Ultrasónicas , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
8.
Arterioscler Thromb Vasc Biol ; 39(8): 1602-1613, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31189431

RESUMEN

OBJECTIVE: The early embryo implantation is characterized by enhanced uterine vascular permeability at the site of blastocyst attachment, followed by extracellular-matrix remodeling and angiogenesis. Two TG (transglutaminase) isoenzymes, TG2 (tissue TG) and FXIII (factor XIII), catalyze covalent cross-linking of the extracellular-matrix. However, their specific role during embryo implantation is not fully understood. Approach and Results: For mapping the distribution as well as the enzymatic activities of TG2 and FXIII towards blood-borne and resident extracellular-matrix substrates, we synthetized selective and specific low molecular weight substrate analogs for each of the isoenzymes. The implantation sites were challenged by genetically modifying the trophoblast cells in the outer layer of blastocysts, to either overexpress or deplete TG2 or FXIII, and the angiogenic response was studied by dynamic contrast-enhanced-magnetic resonance imaging. Dynamic contrast-enhanced-magnetic resonance imaging revealed a decrease in the permeability of decidual vasculature surrounding embryos in which FXIII were overexpressed in trophoblast cell. Reduction in decidual blood volume fraction was demonstrated when either FXIII or TG2 were overexpressed in embryonic trophoblast cell and was elevated when trophoblast cell was depleted of FXIII. These results were corroborated by histological analysis. CONCLUSIONS: In this study, we report on the isoenzyme-specific roles of TG2 and FXIII during the early days of mouse pregnancy and further reveal their involvement in decidual angiogenesis. Our results reveal an important magnetic resonance imaging-detectable function of embryo-derived TG2 and FXIII on regulating maternal angiogenesis during embryo implantation in mice.Visual Overview: An online visual overview is available for this article.


Asunto(s)
Implantación del Embrión/fisiología , Factor XIII/fisiología , Proteínas de Unión al GTP/fisiología , Imagen por Resonancia Magnética/métodos , Neovascularización Fisiológica/fisiología , Transglutaminasas/fisiología , Animales , Femenino , Fibrinógeno/fisiología , Ratones , Embarazo , Proteína Glutamina Gamma Glutamiltransferasa 2
9.
J Mol Neurosci ; 68(1): 135-143, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30903486

RESUMEN

Nerve growth factor (NGF) promotes pleiotropic gene transcription-dependent biological effects, in neuronal and non-neuronal cells, including survival, proliferation, differentiation, neuroprotection, pain, and angiogenesis. It is hypothesized that during odontogenesis, NGF may be implicated in morphogenetic and mineralization events by affecting proliferation and/or differentiation of dental cells. Tuftelin belongs to the enamel associated teeth proteins and is thought to play a role in enamel mineralization. We previously reported that tuftelin transcript and protein, which are ubiquitously expressed in various tissues of embryos, adults, and tumors, were significantly upregulated during NGF-induced PC12 differentiation. To further confirm the involvement of tuftelin in the differentiation process, we established a tuftelin-knockdown neuronal PC12 cell model, using a non-cytotoxic siRNA directed towards sequences at the 3' UTR of the tuftelin gene. Using real-time PCR, we quantified tuftelin mRNA expression and found that tuftelin siRNA, but not scrambled siRNA or transfection reagents, efficiently depleted about 60% of NGF-induced tuftelin mRNA transcripts. The effect of tuftelin siRNA was quantified up to 6 days of NGF-induced differentiation. Using immunofluorescence and western blot analyses, we also found a direct correlation between reduction of 60-80% in tuftelin protein expression and inhibition of about 50-70% in NGF-induced differentiation of the cells, as was detected after 3-6 days of treatment. These results demonstrate an important role for tuftelin in NGF-induced differentiation of PC12 cells. Tuftelin could be a useful target for drug development in disease where neurotrophin therapy is required.


Asunto(s)
Proteínas del Esmalte Dental/metabolismo , Neurogénesis/genética , Animales , Proteínas del Esmalte Dental/genética , Factor de Crecimiento Nervioso/farmacología , Neurogénesis/efectos de los fármacos , Células PC12 , Ratas
10.
Toxins (Basel) ; 10(12)2018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30486274

RESUMEN

Chlorotoxin (CTX) is a 36-amino-acid disulfide-containing peptide derived from the venom of the scorpion Leiurus quinquestriatus. CTX alters physiology in numerous ways. It interacts with voltage gated chloride channels, Annexin-2, and matrix metalloproteinase-2 (MMP-2). CTX-based bioconjugates have been widely subjected to phase I/II clinical trials and have shown substantial promise. Many studies have demonstrated that CTX preferentially binds to neuroectodermal tumors, such as glioblastoma, without cross-reactivity to normal brain cells. With its ability to penetrate the blood-brain-barrier (BBB) and its tyrosine residue allows covalent conjugation with functional moieties, CTX is an attractive platform to explore development of diagnostic and therapeutic agents for gliomas. In this review, we outline CTX structure and its molecular targets, summarize molecular variations of CTX developed for glioma imaging, and discuss future trends and perspectives for CTX conjugates as a theranostic agent.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Glioma/diagnóstico por imagen , Venenos de Escorpión/uso terapéutico , Animales , Humanos , Imagen Molecular , Imagen Multimodal , Venenos de Escorpión/química
11.
FASEB J ; : fj201701568R, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29856660

RESUMEN

Glioblastoma is an aggressive and invasive brain malignancy with high mortality rates despite current treatment modalities. In this study, we show that a 7-gene signature, previously found to govern the switch of glioblastomas from dormancy to aggressive tumor growth, correlates with improved overall survival of patients with glioblastoma. Using glioblastoma dormancy models, we validated the role of 2 genes from the signature, thrombospondin-1 ( TSP-1) and epidermal growth factor receptor ( EGFR), as regulators of glioblastoma dormancy and explored their therapeutic potential. EGFR up-regulation was reversed using EGFR small interfering RNA polyplex, antibody, or small-molecule inhibitor. The diminished function of TSP-1 was augmented via a peptidomimetic. The combination of EGFR inhibition and TSP-1 restoration led to enhanced therapeutic efficacy in vitro, in 3-dimensional patient-derived spheroids, and in a subcutaneous human glioblastoma model in vivo. Systemic administration of the combination therapy to mice bearing intracranial murine glioblastoma resulted in marginal therapeutic outcomes, probably due to brain delivery challenges, p53 mutation status, and the aggressive nature of the selected cell line. Nevertheless, this study provides a proof of concept for exploiting regulators of tumor dormancy for glioblastoma therapy. This therapeutic strategy can be exploited for future investigations using a variety of therapeutic entities that manipulate the expression of dormancy-associated genes in glioblastoma as well as in other cancer types.-Tiram, G., Ferber, S., Ofek, P., Eldar-Boock, A., Ben-Shushan, D., Yeini, E., Krivitsky, A., Blatt, R., Almog, N., Henkin, J., Amsalem, O., Yavin, E., Cohen, G., Lazarovici, P., Lee, J. S., Ruppin, E., Milyavsky, M., Grossman, R., Ram, Z., Calderón, M., Haag, R., Satchi-Fainaro, R. Reverting the molecular fingerprint of tumor dormancy as a therapeutic strategy for glioblastoma.

12.
Biochim Biophys Acta Gen Subj ; 1861(3): 615-623, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28063984

RESUMEN

BACKGROUND: Peptide and protein toxins are essential tools to dissect and probe the biology of their target receptors. Venoms target vital physiological processes to evoke pain. Snake venoms contain various factors with the ability to evoke, enhance and sustain pain sensation. While a number of venom-derived toxins were shown to directly target TRPV1 channels expressed on somatosensory nerve terminals to evoke pain response, such toxins were yet to be identified in snake venoms. METHODS: We screened Echis coloratus saw-scaled viper venom's protein fractions isolated by reversed phase HPLC for their ability to activate TRPV1 channels. To this end, we employed heterologous systems to analyze TRPV1 and NGF pathways by imaging and electrophysiology, combined with molecular biology, biochemical, and pharmacological tools. RESULTS: We identified TRPV1 activating proteins in the venom of Echis coloratus that produce a channel-dependent increase in intracellular calcium and outwardly rectifying currents in neurons and heterologous systems. Interestingly, channel activation was not mediated by any of its known toxin binding sites. Moreover, although NGF neurotropic activity was detected in this venom, TRPV1 activation was independent of NGF receptors. CONCLUSIONS: Echis coloratus venom contains proteins with the ability to directly activate TRPV1. This activity is independent of the NGF pathway and is not mediated by known TRPV1 toxins' binding sites. GENERAL SIGNIFICANCE: Our results could facilitate the discovery of new toxins targeting TRPV1 to enhance current understanding of this receptor activation mechanism. Furthermore, the findings of this study provide insight into the mechanism through which snakes' venom elicit pain.


Asunto(s)
Proteínas/metabolismo , Canales Catiónicos TRPV/metabolismo , Venenos de Víboras/metabolismo , Viperidae/metabolismo , Animales , Sitios de Unión/fisiología , Calcio/metabolismo , Línea Celular , Células HEK293 , Humanos , Factor de Crecimiento Nervioso/metabolismo , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Toxinas Biológicas/metabolismo
13.
Ultrasound Med Biol ; 41(9): 2449-57, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26003010

RESUMEN

The purpose of this work was to investigate whether low-frequency, low-intensity (20 kHz, <100 mW/cm(2), spatial-peak, temporal-peak intensity) ultrasound, delivered with a lightweight (<100 g), tether-free, fully wearable, battery-powered applicator, is capable of reducing inflammation in a mouse model of rheumatoid arthritis. The therapeutic, acute, anti-inflammatory effect was estimated from the relative swelling induced in mice hindlimb paws. In an independent, indirect approach, the inflammation was bio-imaged by measuring glycolytic activity with near-infrared labeled 2-deoxyglucose. The outcome of the experiments indicated that the combination of ultrasound exposure and topical application of 0.1% (w/w) betamethasone gel resulted in statistically significantly (p < 0.05) enhanced anti-inflammatory activity in comparison with drug or ultrasound treatment alone. The present study underscores the potential benefits of low-frequency, low-intensity ultrasound-assisted drug delivery. However, the proof of concept presented indicates the need for additional experiments to systematically evaluate and optimize the potential of, and the conditions for, tolerable low-frequency, low-intensity ultrasound-promoted non-invasive drug delivery.


Asunto(s)
Artritis/diagnóstico , Artritis/terapia , Betametasona/administración & dosificación , Electroporación/métodos , Sonicación/métodos , Administración Tópica , Animales , Antiinflamatorios/administración & dosificación , Carragenina , Terapia Combinada/métodos , Sinergismo Farmacológico , Masculino , Ratones , Ratones Endogámicos ICR , Sonicación/instrumentación , Resultado del Tratamiento
14.
J Mol Neurosci ; 54(3): 574-85, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25078264

RESUMEN

Nerve growth factor (NGF) treatment causes a profound down-regulation of epidermal growth factor (EGF) receptors (EGFR) during the neuronal differentiation of PC12 cells. This process was characterized by a progressive decrease in EGFR level, as measured by (125)I-EGF binding and Scatchard analysis, tyrosine phosphorylation, Western blotting, and bio-imaging using EGF-labeled with a near-infrared probe. Differentiation of the cells with NGF for 5-7 days produces a 95 % reduction in the amount of (35)S-methionine-labeled EGFR. This down-regulation does not occur in PC12-nnr5 cells, which lack the TrkA NGF receptor but is reconstituted in these cells upon their stable transfection with TrkA. The process of NGF-induced EGFR down-regulation was inhibited by K252a, a TrkA antagonist and by anti-TrkA antibodies but not by Thx-B, a blocker of the interaction of NGF with p75(NTR) receptors. NGF-induced (heterologous) down-regulation, but not EGF-induced (homologous) down-regulation of EGFR, was blocked in Ras-deficient PC12 cells. NGF treatment for 5-7 days of PC12 cells, grown in suspension or in 3D collagen gels, induces down-regulation of EGFR independent of neurite outgrowth. The messenger RNA (mRNA) for EGFR decreased in a comparable fashion. This process was correlated temporally with a decrease in the transcription of the EGFR gene. Treatment with NGF also increased the cellular content of GCF2, a putative inhibitory transcription factor of the EGFR gene. The temporal increase in GCF2, like the decrease in the EGFR mRNA, was not seen in TrkA deficient PC12 cells nor in cells expressing dominant-negative Ras. The results suggest that NGF-induced down-regulation of the EGFR is under transcriptional control, is TrkA and Ras-dependent, may involve transcriptional repression by GCF2, and independent of mechanisms that lead to NGF-induced neurite outgrowth in PC12cells. This heterologous down-regulation of EGFR would appear to be an efficient mean of desensitizing the neuron to proliferative stimuli, thereby representing a safety latch for initiating and sustaining NGF-induced neuronal differentiation.


Asunto(s)
Regulación hacia Abajo , Receptores ErbB/metabolismo , Factor de Crecimiento Nervioso/farmacología , Transcripción Genética , Animales , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Células PC12 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptor trkA/metabolismo , Proteínas ras/metabolismo
15.
J Pharmacol Exp Ther ; 350(3): 506-19, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24939421

RESUMEN

Obtustatin and viperistatin, members of the disintegrin protein family, served as lead compounds for the synthesis of linear and cyclic peptides containing the KTS binding motif. The most active linear peptide, a viperistatin analog, indicated the importance of Cys(19) and Cys(29), as well as the presence of Arg at position 24 for their biologic activity, and was used as the basic sequence for the synthesis of cyclic peptides. Vimocin (compound 6) and vidapin (compound 10) showed a high potency (IC50 = 0.17 nM) and intermediate efficacy (20 and 40%) in inhibition of adhesion of α1/α2 integrin overexpressor cells to respective collagens. Vimocin was more active in inhibition of the wound healing (53%) and corneal micropocket (17%) vascularization, whereas vidapin was more potent in inhibition of migration in the Matrigel tube formation assay (90%). Both compounds similarly inhibited proliferation (50-90%) of endothelial cells, and angiogenesis induced by vascular endothelial growth factor (80%) and glioma (55%) in the chorioallantoic membrane assay. These peptides were not toxic to endothelial cell cultures and caused no acute toxicity upon intravenous injection in mice, and were stable for 10-30 hours in human serum. The in vitro and in vivo potency of the peptides are consistent with conformational ensembles and "bioactive" space shared by obtustatin and viperistatin. These findings suggest that vimocin and vidapin can serve as dual α1ß1/α2ß1 integrin antagonists in antiangiogenesis and cancer therapy.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Integrina alfa1beta1/antagonistas & inhibidores , Integrina alfa2beta1/antagonistas & inhibidores , Péptidos Cíclicos/farmacología , Venenos de Víboras/farmacología , Inhibidores de la Angiogénesis/química , Animales , Bovinos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa1beta1/metabolismo , Integrina alfa2beta1/metabolismo , Masculino , Ratones , Péptidos Cíclicos/química , Codorniz , Ratas , Venenos de Víboras/química
16.
J Neurotrauma ; 31(16): 1405-16, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24640955

RESUMEN

Treatment of traumatic brain injury (TBI) is still an unmet need. Cell therapy by human umbilical cord blood (HUCB) has shown promising results in animal models of TBI and is under evaluation in clinical trials. HUCB contains different cell populations but to date, only mesenchymal stem cells have been evaluated for therapy of TBI. Here we present the neurotherapeutic effect, as evaluated by neurological score, using a single dose of HUCB-derived mononuclear cells (MNCs) upon intravenous (IV) administration one day post-trauma in a mouse model of closed head injury (CHI). Delayed (eight days post-trauma) intracerebroventricular administration of MNCs showed improved neurobehavioral deficits thereby extending the therapeutic window for treating TBI. Further, we demonstrated for the first time that HUCB-derived pan-hematopoietic CD45 positive (CD45(+)) cells, isolated by magnetic sorting and characterized by expression of CD45 and CD11b markers (96-99%), improved the neurobehavioral deficits upon IV administration, which persisted for 35 days. The therapeutic effect was in a direct correlation to a reduction in the lesion volume and decreased by pre-treatment of the cells with anti-human-CD45 antibody. At the site of brain injury, 1.5-2 h after transplantation, HUCB-derived cells were identified by near infrared scanning and immunohistochemistry using anti-human-CD45 and anti-human-nuclei antibodies. Nerve growth factor and vascular endothelial growth factor levels were differentially expressed in both ipsilateral and contralateral brain hemispheres, thirty-five days after CHI, measured by enzyme-linked immunosorbent assay. These findings indicate the neurotherapeutic potential of HUCB-derived CD45(+) cell population in a mouse model of TBI and propose their use in the clinical setting of human TBI.


Asunto(s)
Lesiones Encefálicas/terapia , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Recuperación de la Función , Animales , Separación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Antígenos Comunes de Leucocito/inmunología , Masculino , Ratones
17.
J Biomater Sci Polym Ed ; 25(6): 608-24, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24568316

RESUMEN

One of the challenges in regenerative medicine is the development of novel biodegradable materials to build scaffolds that will support multiple cell types for tissue engineering. Here we describe the preparation, characterization, and cytocompatibility of homo- and hetero-polyesters of α-hydroxy amino acid derivatives with or without lactic acid conjugation. The polymers were prepared by a direct condensation method and characterized using gel permeation chromatography, (1)H-nuclear magnetic resonance spectroscopy, Fourier transform infrared spectroscopy, differential scanning calorimetry, optical activity, and solubility. The surface charge of the polymers was evaluated using zeta potential measurements. The polymers were coated onto glass cover slips followed by characterization using nano-surface profiler, thin film reflectometry, and atomic force microscopy (AFM). Their interaction with endothelial and neuronal cells was assessed using adhesion, proliferation, and differentiation assays. Of the characterized polymers, Poly-HOVal-LA, but not Poly-(D)HOPhe, significantly augmented nerve growth factor (NGF)-induced neuronal differentiation of the PC12 pheochromcytoma cells. In contrast, Poly-HOLeu increased by 20% the adhesion of endothelial cells, but did not affect PC12 cell differentiation. NGF-induced Erk1/2 phosphorylation in PC12 cells grown on the different polymers was similar to the effect observed for cells cultured on collagen type I. While no significant association could be established between charge and the differentiative/proliferative properties of the polymers, AFM analysis indicated augmentation of NGF-induced neuronal differentiation on smooth polymer surfaces. We conclude that overall selective cytocompatibility and bioactivity might render α-hydroxy amino acid polymers useful as extracellular matrix-mimicking materials for tissue engineering.


Asunto(s)
Aminoácidos/química , Materiales Biocompatibles/química , Poliésteres/química , Polímeros/química , Animales , Materiales Biocompatibles/efectos adversos , Materiales Biocompatibles/farmacología , Adhesión Celular/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Poliésteres/efectos adversos , Poliésteres/farmacología , Polímeros/efectos adversos , Polímeros/farmacología , Ratas , Ingeniería de Tejidos/métodos
18.
Toxins (Basel) ; 5(8): 1332-42, 2013 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-23899970

RESUMEN

The effect of the venom of the Chactoid family of scorpions on blood pressure was scantly investigated and was addressed in the present study using the venom of the Israeli scorpion, Scorpio maurus palmatus. Blood pressure in rats was monitored via cannulated femoral artery, while venom and toxins were introduced into femoral vein. Venom injection elicited a biphasic effect, expressed first by a fast and transient hypotensive response, which lasted up to 10 min, followed by a hypertensive response, which lasted up to one hour. It was found that these effects resulted from different venom components. Phospholipase A2 produced the hypotensive effect, while a non-enzymatic neurotoxic polypeptide fraction produced the hypertensive effect. Surprisingly, the main neurotoxic polypeptide to mice had no effect on blood pressure. In vitro experiments indicated that the hypertensive factors caused histamine release from the peritoneal mast cells, but this effect is assumed to be not relevant to their in vivo effect. In spite of the cytotoxic activity of phospholipase A2, it did not release histamine. These findings suggest that the effects of venom and isolated fractions on blood pressure parameters are mediated by different mechanisms, which deserve further pharmacological investigation.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Liberación de Histamina/efectos de los fármacos , Mastocitos/efectos de los fármacos , Venenos de Escorpión/toxicidad , Toxinas Biológicas/toxicidad , Animales , Insectos/efectos de los fármacos , Masculino , Ratones , Neurotoxinas/toxicidad , Fosfolipasas A2/metabolismo , Ratas , Ratas Wistar , Escorpiones/química
19.
Int J Mol Sci ; 14(7): 14669-88, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23857061

RESUMEN

In this study, we present the applicability of imaging epidermal growth factor (EGF) receptor levels in preclinical models of COLO205 carcinoma cells in vitro, mice with orthotopic tumors and ex vivo colorectal tumor biopsies, using EGF-labeled with IRDye800CW (EGF-NIR). The near infrared (NIR) bio-imaging of COLO205 cultures indicated specific and selective binding, reflecting EGF receptors levels. In vivo imaging of tumors in mice showed that the highest signal/background ratio between tumor and adjacent tissue was achieved 48 hours post-injection. Dissected colorectal cancer tissues from different patients demonstrated ex vivo specific imaging using the NIR bio-imaging platform of the heterogeneous distributed EGF receptors. Moreover, in the adjacent gastrointestinal tissue of the same patients, which by Western blotting was demonstrated as EGF receptor negative, no labeling with EGF-NIR probe was detected. Present results support the concept of tumor imaging by measuring EGF receptor levels using EGF-NIR probe. This platform is advantageous for EGF receptor bio-imaging of the NCI-60 recommended panel of tumor cell lines including 6-9 colorectal cell lines, since it avoids radioactive probes and is appropriate for use in the clinical setting using NIR technologies in a real-time manner.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Receptores ErbB/metabolismo , Espectroscopía Infrarroja Corta , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Ratones , Ratones Desnudos , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Trasplante Heterólogo , Imagen de Cuerpo Entero
20.
J Mol Neurosci ; 49(2): 334-46, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22878912

RESUMEN

The aim of our work is to utilize the crosstalk between the vascular and the neuronal system to enhance directed neuritogenesis in uniaxial guidance scaffolds for the repair of spinal cord injury. In this study, we describe a method for angioneural regenerative engineering, i.e., for generating biodegradable scaffolds, produced by a combination of controlled freezing (freeze-casting) and lyophilization, which contain longitudinally oriented channels, and provide uniaxial directionality to support and guide neuritogenesis from neuronal cells in the presence of endothelial cells. The optimized scaffolds, composed of 2.5 % gelatin and 1 % genipin crosslinked, were characterized by an elastic modulus of ~51 kPa and longitudinal channels of ~50 µm diameter. The scaffolds support the growth of endothelial cells, undifferentiated or NGF-differentiated PC12 cells, and primary cultures of fetal chick forebrain neurons. The angioneural crosstalk, as generated by first forming endothelial cell monolayers in the scaffolds followed by injection of neuronal cells, leads to the outgrowth of long aligned neurites in the PC12/endothelial cell co-cultures also in the absence of exogenously added nerve growth factor. Neuritogenesis was not observed in the scaffolds in the absence of the endothelial cells. This methodology is a promising approach for neural tissue engineering and may be applicable for regenerative spinal cord injury repair.


Asunto(s)
Células Endoteliales/citología , Neurogénesis , Neuronas/citología , Andamios del Tejido/química , Animales , Embrión de Pollo , Módulo de Elasticidad , Liofilización/instrumentación , Liofilización/métodos , Gelatina , Iridoides , Factor de Crecimiento Nervioso/farmacología , Células-Madre Neurales/citología , Células PC12 , Ratas , Traumatismos de la Médula Espinal/terapia , Ingeniería de Tejidos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...