Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neurochem Res ; 49(6): 1556-1576, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38160216

RESUMEN

Multiple sclerosis (MS) is a pathological condition characterized by the demyelination of nerve fibers, primarily attributed to the destruction of oligodendrocytes and subsequent motor neuron impairment. Ethidium bromide (EB) is a neurotoxic compound that induces neuronal degeneration, resulting in demyelination and symptoms resembling those observed in experimental animal models of multiple sclerosis (MS). The neurotoxic effects induced by EB in multiple sclerosis (MS) are distinguished by the death of oligodendrocytes, degradation of myelin basic protein (MBP), and deterioration of axons. Neurological complications related to MS have been linked to alterations in the signaling pathway known as smo-shh. Purmorphine (PUR) is a semi-synthetic compound that exhibits potent Smo-shh agonistic activity. It possesses various pharmacological properties, including antioxidant, anti-inflammatory, anti-apoptotic, and neuromodulatory effects. Hence, the current investigation was conducted to assess the neuroprotective efficacy of PUR (at doses of 5 and 10 mg/kg, administered intraperitoneally) both individually and in conjunction with Fingolimod (FING) (at a dose of 0.5 mg/kg, administered intraperitoneally) in the experimental model of MS induced by EB. The administration of EB was conducted via the intracerebropeduncle route (ICP) over a period of seven days in the brain of rats. The Wistar rats were allocated into six groups using randomization, each consisting of eight rats (n = 8 per group). The experimental groups in this study were categorized as follows: (I) Sham Control, (II) Vehicle Control, (III) PUR per se, (IV) EB, (V) EB + PUR5, (VI) EB + PUR10, (VII) EB + FING 0.5, and (VIII) EB + PUR10 + FING 0.5. On the final day of the experimental timeline, all animal subjects were euthanized, and subsequent neurochemical estimations were conducted on cerebrospinal fluid, blood plasma, and brain tissue samples. In addition, we conducted neurofilament (NFL) analysis and histopathological examination. We utilized the luxol myelin stain to understand better the degeneration associated with MS and its associated neurological complications. The findings of our study indicate that the activation of SMO-Shh by PUR has a mitigating effect on neurobehavioral impairments induced by EB, as well as a restorative effect on cellular and neurotransmitter abnormalities in an experimental model of MS.


Asunto(s)
Proteínas Hedgehog , Morfolinas , Esclerosis Múltiple , Neurogénesis , Purinas , Animales , Masculino , Ratas , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Etidio , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/metabolismo , Neurogénesis/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas Wistar , Receptor Smoothened/efectos de los fármacos , Receptor Smoothened/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Morfolinas/farmacología , Purinas/farmacología
2.
Pancreatology ; 20(6): 1115-1122, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32778368

RESUMEN

BACKGROUND/OBJECTIVES: Preclinical data indicated a functional and molecular interaction between Hedgehog (HH)/GLI and PI3K-AKT-mTOR pathways promoting pancreatic ductal adenocarcinoma (PDAC). A phase I study was conducted of Vismodegib and Sirolimus combination to evaluate maximum tolerated dose (MTD) and preliminary anti-tumor efficacy. METHODS: Cohort I included advanced solid tumors patients following a traditional 3 + 3 design. Vismodegib was orally administered at 150 mg daily with Sirolimus starting at 3 mg daily, increasing to 6 mg daily at dose level 2. Cohort II included only metastatic PDAC patients. Anti-tumor efficacy was evaluated every two cycles and target assessment at pre-treatment and after a single cycle. RESULTS: Nine patient were enrolled in cohort I and 22 patients in cohort II. Twenty-eight patients were evaluated for dose-limiting toxicities (DLTs). One DLT was observed in each cohort, consisting of grade 2 mucositis and grade 3 thrombocytopenia. The MTD for Vismodegib and Sirolimus were 150 mg daily and 6 mg daily, respectively. The most common grade 3-4 toxicities were fatigue, thrombocytopenia, dehydration, and infections. A total of 6 patients had stable disease. No partial or complete responses were observed. Paired biopsy analysis before and after the first cycle in cohort II consistently demonstrated reduced GLI1 expression. Conversely, GLI and mTOR downstream targets were not significantly affected. CONCLUSIONS: The combination of Vismodegib and Sirolimus was well tolerated. Clinical benefit was limited to stable disease in a subgroup of patients. Targeting efficacy demonstrated consistent partial decreases in HH/GLI signaling with limited impact on mTOR signaling. These findings conflict with pre-clinical models and warrant further investigations.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Proteínas Hedgehog/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/efectos de los fármacos , Adulto , Anciano , Anilidas/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Biopsia , Quimioterapia Combinada , Femenino , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Inmunosupresores/efectos adversos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Resultados Negativos , Metástasis de la Neoplasia , Piridinas/administración & dosificación , ARN Neoplásico/química , ARN Neoplásico/genética , Transducción de Señal/efectos de los fármacos , Sirolimus/efectos adversos , Resultado del Tratamiento
3.
Alcohol Clin Exp Res ; 44(7): 1366-1377, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32472575

RESUMEN

BACKGROUND: Ethanol (EtOH) has diverse effects on nervous system development, which includes development and survival of GABAergic neurons in a sonic hedgehog (Shh) and fibroblast growth factor (Fgf)-dependent mechanism. Cannabinoids also function as inhibitors of Shh signaling, raising the possibility that EtOH and cannabinoids may interact to broadly disrupt neuronal function during brain development. METHODS: Zebrafish embryos were exposed to a range of EtOH and/or cannabinoid receptor 1 (CB1R) agonist concentrations at specific developmental stages, in the absence or presence of morpholino oligonucleotides that disrupt shh expression. In situ hybridization was employed to analyze glutamic acid decarboxylase (gad1) gene expression as a marker of GABAergic neuron differentiation, and zebrafish behavior was analyzed using the novel tank diving test as a measure of risk-taking behavior. RESULTS: Combined acute subthreshold EtOH and CB1R agonist exposure results in a marked reduction in gad1 mRNA expression in zebrafish forebrain. Consistent with the EtOH and cannabinoid effects on Shh signaling, fgf8 mRNA overexpression rescues the EtOH- and cannabinoid-induced decrease in gad1 gene expression and also prevents the changes in behavior induced by EtOH and cannabinoids. CONCLUSIONS: These studies provide evidence that forebrain GABAergic neuron development and zebrafish risk-taking behavior are sensitive to both EtOH and cannabinoid exposure in a Shh- and Fgf-dependent mechanism, and provide additional evidence that a signaling pathway involving Shh and Fgf crosstalk is a critical target of EtOH and cannabinoids in FASD.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Factores de Crecimiento de Fibroblastos/genética , Neuronas GABAérgicas/efectos de los fármacos , Proteínas Hedgehog/genética , Neurogénesis/efectos de los fármacos , Proteínas de Pez Cebra/genética , Animales , Conducta Animal/efectos de los fármacos , Embrión no Mamífero , Expresión Génica , Glutamato Descarboxilasa/efectos de los fármacos , Glutamato Descarboxilasa/genética , Proteínas Hedgehog/efectos de los fármacos , Hibridación in Situ , Morfolinos , Neurogénesis/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Cannabinoide CB1/agonistas , Asunción de Riesgos , Pez Cebra , Proteínas de Pez Cebra/efectos de los fármacos
4.
Neurochem Res ; 44(2): 441-449, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30552546

RESUMEN

The thrombolytic activity of tissue plasminogen activator (tPA) has undisputed benefits. However, the documented neurotoxicity of tPA raises important issues. Currently, common treatments for stroke might not be optimum if exogenous tPA can pass through the blood-brain barrier and enter the brain, thus adding to the deleterious effects of tPA within the cerebral parenchyma. Here, we determined whether tPA could damage brain microvascular endothelial cells (BMECs) during cerebral ischemia. We showed that treatment of BMECs with tPA decreased trans-endothelial electrical resistance and cell proliferation, and blocked the cell cycle at the G0-G1 phase. In addition, the Sonic hedgehog (Shh) signaling pathway was involved in tPA-induced BMECs dysfunction. However, tPA-enhanced oxygen glucose deprivation-induced BMECs dysfunction was eliminated by Shh administration and the effects could be reversed by Shh inhibitors. Taken together, these results demonstrate that tPA administration might result in damage to the endothelial barrier owing to blocked Shh signaling pathway.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Glucosa/metabolismo , Proteínas Hedgehog/efectos de los fármacos , Oxígeno/metabolismo , Activador de Tejido Plasminógeno/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
5.
Proc Natl Acad Sci U S A ; 113(47): E7545-E7553, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27815529

RESUMEN

Inflammation disrupts tissue architecture and function, thereby contributing to the pathogenesis of diverse diseases; the signals that promote or restrict tissue inflammation thus represent potential targets for therapeutic intervention. Here, we report that genetic or pharmacologic Hedgehog pathway inhibition intensifies colon inflammation (colitis) in mice. Conversely, genetic augmentation of Hedgehog response and systemic small-molecule Hedgehog pathway activation potently ameliorate colitis and restrain initiation and progression of colitis-induced adenocarcinoma. Within the colon, the Hedgehog protein signal does not act directly on the epithelium itself, but on underlying stromal cells to induce expression of IL-10, an immune-modulatory cytokine long known to suppress inflammatory intestinal damage. IL-10 function is required for the full protective effect of small-molecule Hedgehog pathway activation in colitis; this pharmacologic augmentation of Hedgehog pathway activity and stromal IL-10 expression are associated with increased presence of CD4+Foxp3+ regulatory T cells. We thus identify stromal cells as cellular coordinators of colon inflammation and suggest their pharmacologic manipulation as a potential means to treat colitis.


Asunto(s)
Colitis/metabolismo , Sulfato de Dextran/efectos adversos , Proteínas Hedgehog/metabolismo , Interleucina-10/metabolismo , Transducción de Señal , Animales , Antígenos CD4/metabolismo , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Factores de Transcripción Forkhead/metabolismo , Proteínas Hedgehog/efectos de los fármacos , Humanos , Ratones , Mutación , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/farmacología , Linfocitos T Reguladores/metabolismo , Proteína con Dedos de Zinc GLI1/genética
6.
Med Sci Monit ; 24: 6695-6706, 2018 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-30244259

RESUMEN

BACKGROUND The aim of this study was to determine the role of icariin, a Chinese traditional herbal medicine extracted from Epimedium, in osteoarthritis (OA), using the murine anterior cruciate ligament transection (ACLT)-induced model of OA and micromass culture of murine chondrocytes. MATERIAL AND METHODS Twenty-four three-month-old C57/6J mice were randomly divided into three groups: the sham group (no surgery and joint injection with normal saline) (N=8); the ACLT + ICA group (ACLT surgery and icariin treatment) (N=8); and the ACLT group (ACLT surgery and joint injection with normal saline) (N=8). At 12 weeks after ACLT surgery, murine articular cartilage was harvested from all mice for histological evaluation of any differences in cartilage degeneration. In vitro micromass culture of mouse chondrocytes was used to study the effects of icariin on chondrocyte differentiation and growth from the three mouse groups. RESULTS Icariin treatment (mice in the ACLT + ICA group) significantly reduced degeneration of cartilage in OA with increased cartilage thickness, associated with increased expression of collagen type II alpha 1 (COL2A1), decreased chondrocyte hypertrophy, and decreased expression of collagen type X (ColX) and matrix metalloproteinase 13 (MMP13). In vitro, icariin promoted chondrocyte differentiation by upregulating the expression of agrrecan, Sox9 and parathyroid hormone-related protein (PHrP) and down-regulation of Indian hedgehog (Ihh) and genes regulated by Ihh. CONCLUSIONS In a mouse model of OA icariin treatment reduced destruction of cartilage, promoted chondrocyte differentiation, upregulated expression of PHrP and down-regulated the expression of Ihh.


Asunto(s)
Cartílago/efectos de los fármacos , Flavonoides/farmacología , Osteoartritis/tratamiento farmacológico , Animales , Ligamento Cruzado Anterior/efectos de los fármacos , Cartílago Articular/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Colágeno Tipo II/metabolismo , Modelos Animales de Enfermedad , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteoartritis de la Rodilla/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/efectos de los fármacos
7.
Alcohol Clin Exp Res ; 41(12): 2051-2065, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28921619

RESUMEN

BACKGROUND: Cell and molecular mechanisms mediating the cardiovascular effects of alcohol are not fully understood. Our aim was to determine the effect of moderate ethanol (EtOH) on sonic hedgehog (SHh) signaling in regulating possible stem cell antigen-1 positive (Sca1+ ) progenitor stem cell involvement during pathologic arterial remodeling. METHODS: Partial ligation or sham operation of the left carotid artery was performed in transgenic Sca1-enhanced green fluorescent protein (eGFP) mice gavaged with or without "daily moderate" EtOH. RESULTS: The EtOH group had reduced adventitial thickening and less neointimal formation, compared to ligated controls. There was expansion of eGFP-expressing (i.e., Sca1+ ) cells in remodeled vessels postligation (day 14), especially in the neo intima. EtOH treatment reduced the number of Sca1+ cells in ligated vessel cross-sections concomitant with diminished remodeling, compared to control ligated vessels. Moreover, EtOH attenuated SHh signaling in injured carotids as determined by immunohistochemical analysis of the target genes patched 1 and Gli2, and RT-PCR of whole-vessel Gli2 mRNA levels. Intraperitoneal injection of ligated Sca1-eGFP mice with the SHh signaling inhibitor cyclopamine diminished SHh target gene expression, reduced the number of Sca1+ cells, and ameliorated carotid remodeling. EtOH treatment of purified Sca1+ adventitial progenitor stem cells in vitro inhibited SHh signaling, and their rSHh-induced differentiation to vascular smooth muscle cells. CONCLUSIONS: EtOH reduces SHh-responsive Sca1+ progenitor cell myogenic differentiation/expansion in vitro and during arterial remodeling in response to ligation injury in vivo. Regulation of vascular Sca1+ progenitor cells in this way may be an important novel mechanism contributing to alcohol's cardiovascular protective effects.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Hedgehog/fisiología , Células Madre/fisiología , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/fisiología , Animales , Antígenos Ly/inmunología , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/fisiopatología , Proliferación Celular/fisiología , Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/efectos de los fármacos , Proteínas de la Membrana/inmunología , Ratones , Ratones Transgénicos , Receptor Patched-1/biosíntesis , Células Madre/inmunología , Alcaloides de Veratrum/farmacología , Proteína Gli2 con Dedos de Zinc/biosíntesis
8.
Am J Physiol Endocrinol Metab ; 310(10): E846-54, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27026086

RESUMEN

Thyroid hormone (TH) action is mediated through two nuclear TH receptors, THRα and THRß. Although the role of THRα is well established in bone, less is known about the relevance of THRß-mediated signaling in bone development. On ther basis of our recent finding that TH signaling is essential for initiation and formation of secondary ossification center, we evaluated the role of THRs in mediating TH effects on epiphysial bone formation. Two-day treatment of TH-deficient Tshr(-/-) mice with TH increased THRß1 mRNA level 3.4-fold at day 7 but had no effect on THRα1 mRNA level at the proximal tibia epiphysis. Treatment of serum-free cultures of tibias from 3-day-old mice with T3 increased THRß1 expression 2.1- and 13-fold, respectively, at 24 and 72 h. Ten-day treatment of Tshr(-/-) newborns (days 5-14) with THRß1 agonist GC1 at 0.2 or 2.0 µg/day increased BV/TV at day 21 by 225 and 263%, respectively, compared with vehicle treatment. Two-day treatment with GC1 (0.2 µg/day) increased expression levels of Indian hedgehog (Ihh) 100-fold, osterix 15-fold, and osteocalcin 59-fold compared with vehicle at day 7 in the proximal tibia epiphysis. Gel mobility shift assay demonstrated that a putative TH response element in the distal promoter of mouse Ihh gene interacted with THRß1. GC1 treatment (1 nM) increased Ihh distal promoter activity 20-fold after 48 h in chondroctyes. Our data suggest a novel role for THRß1 in secondary ossification at the epiphysis that involves transcriptional upregulation of Ihh gene.


Asunto(s)
Epífisis/metabolismo , Proteínas Hedgehog/genética , Osteogénesis/genética , ARN Mensajero/metabolismo , Receptores beta de Hormona Tiroidea/genética , Tibia/metabolismo , Animales , Desarrollo Óseo/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Tirotropina/genética , Transducción de Señal , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/agonistas , Tiroxina/farmacología , Triyodotironina/farmacología , Regulación hacia Arriba
9.
J Clin Periodontol ; 42(1): 29-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25385493

RESUMEN

OBJECTIVE: Cyclosporine-A (CsA)-induced gingival overgrowth may arise from an alteration in stoma matrix homeostasis. Sonic hedgehog (Shh) plays a key role during embryogenic development and fibrotic progression, and may be involved in CsA-altered gingival matrix homeostasis. METHODS: Using the reverse transcription-polymerase chain reaction and Western blot analysis, we investigated the mRNA and protein expressions of Shh, type 1 collagen (COL1), alpha-smooth muscle actin (α-SMA) and transforming growth factor-beta (TGF-ß) in human gingival fibroblasts after CsA treatments. The effect of Shh on CsA-induced alterations was further evaluated by the extra-supplement or inhibition of Shh or TGF-ß. RESULTS: Cyclosporine-A enhanced COL1, α-SMA, Shh and TGF-ß expressions in human gingival fibroblasts. The exogenous Shh/TGF-ß augmented the expression of COL1 and α-SMA, and the Shh/TGF-ß inhibition suppressed the CsA-enhanced COL1 and α-SMA expressions. Moreover, Shh mRNA and protein expressions increased if extra-supplementing the exogenous TGF-ß, whereas the CsA-upregulated Shh was mitigated by the TGF-ß pathway inhibitor. However, neither exogenous Shh nor the Shh pathway inhibitor alters TGF-ß expression or CsA-up-regulated TGF-ß expression. CONCLUSIONS: Shh, regulated by TGF-ß, mediates CsA-altered gingival matrix homeostasis.


Asunto(s)
Actinas/efectos de los fármacos , Colágeno Tipo I/efectos de los fármacos , Ciclosporina/farmacología , Fibroblastos/efectos de los fármacos , Encía/efectos de los fármacos , Proteínas Hedgehog/efectos de los fármacos , Inmunosupresores/farmacología , Factor de Crecimiento Transformador beta/efectos de los fármacos , Técnicas de Cultivo de Célula , Línea Celular , Matriz Extracelular/efectos de los fármacos , Encía/citología , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/fisiología , Homeostasis/efectos de los fármacos , Humanos , Receptor Cross-Talk/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/fisiología , Alcaloides de Veratrum/farmacología
10.
J Periodontal Res ; 49(6): 810-6, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24823913

RESUMEN

BACKGROUND AND OBJECTIVE: Sonic hedgehog protein (SHH) is a mitogen that stimulates cell proliferation. Cyclosporine A enhances the proliferation of gingival cells; however, the relationships of SHH to cyclosporine A or to cyclosporine A-enhanced gingival cell proliferation have not been described. MATERIAL AND METHODS: Here, we investigated SHH expression in gingiva in vitro and in vivo after cyclosporine A treatment and tested the effect of SHH inhibition on cyclosporine A-enhanced gingival fibroblast proliferation in vitro. RESULTS: In human gingival fibroblasts, cyclosporine A treatment increased the expression of SHH transcripts and SHH protein, and stimulated cell proliferation; the addition of cyclopamine, an SHH signaling inhibitor, suppressed cyclosporine A-enhanced cell proliferation. Up-regulated expression of SHH and up-regulation of proliferating cell nuclear antigen transcripts and protein were observed in the edentulous gingiva of cyclosporine A-treated rats. CONCLUSION: Cyclosporine A up-regulates gingival SHH expression in vitro and in vivo, and the inhibition of the SHH pathway counteracts the stimulatory effect of cyclosporine A on gingival fibroblast proliferation. Therefore, we suggest that SHH mediates a novel molecular mechanism for cyclosporine A-induced gingival complications.


Asunto(s)
Ciclosporina/farmacología , Encía/efectos de los fármacos , Proteínas Hedgehog/efectos de los fármacos , Mitógenos/farmacología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Proliferación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Encía/citología , Proteínas Hedgehog/análisis , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Arcada Edéntula/patología , Masculino , Modelos Animales , Proteínas Oncogénicas/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/análisis , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transactivadores/efectos de los fármacos , Regulación hacia Arriba , Alcaloides de Veratrum/farmacología , Proteína con Dedos de Zinc GLI1 , Dedos de Zinc/efectos de los fármacos
12.
Actas Dermosifiliogr ; 105(8): 744-51, 2014 Oct.
Artículo en Inglés, Español | MEDLINE | ID: mdl-24359667

RESUMEN

In January 2012, vismodegib (Erivedge, manufactured by Genentech) became the first selective inhibitor of the Hedgehog signaling pathway to be approved by the US Food and Drug Administration for the treatment of locally advanced and metastatic basal cell carcinoma. The drug selectively binds to Smoothened, a 7-helix transmembrane receptor, thereby inhibiting activation of transcription factors of the glioma-associated oncogene family and suppressing tumor proliferation and growth. Studies published to date have assessed the efficacy of vismodegib according to clinical and radiologic outcomes but little information is available on the molecular mechanisms underpinning the proven clinical efficacy of the drug. This review will cover recent data on the Hedgehog signaling pathway and data from clinical trials with vismodegib in the treatment of basal cell carcinoma, and will consider its use in other types of tumor.


Asunto(s)
Anilidas/uso terapéutico , Carcinoma Basocelular/tratamiento farmacológico , Piridinas/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Anilidas/farmacología , Ensayos Clínicos como Asunto , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/fisiología , Humanos , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos
13.
Prostate ; 73(16): 1810-23, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24105601

RESUMEN

BACKGROUND: Contribution of stromal Hedgehog (Hh) signaling is evident in the prostate gland in mice, but needs translation to human tissues if Hh therapeutics are to be used effectively. Our goal was to determine if primary human prostate fibroblasts contain cilia, and respond to prostate Hh signaling. METHODS: Primary human prostate cancer-associated (CAFs), and adjacent non-malignant (NPFs) fibroblasts isolated from human tissue specimens were analyzed using immunofluorescence, real-time PCR, and available array data. Cell culture and tissue recombination were used to determine responsiveness of human fibroblasts to Hh pathway manipulation and the paracrine effects of stromal Hh signaling, respectively. RESULTS: Prostatic fibroblasts were capable of forming primary cilia, with the capacity for active Hh signaling as seen by Smo co-localization to the tip of the primary cilium. Expression of genes known to represent a signature of active Hh signaling in the prostate (especially Fgf5 and Igfbp6) were increased in CAFs compared to NPFs. The level of canonical Hh genes and prostate Hh signature genes were rarely synchronous; with lower doses of Purmorphamine/BMS-833923 regulating canonical transcription factors, and higher doses effecting prostate Hh signature genes. Grafts consisting of NPFs with constitutively active Hh signaling induced increased proliferation and dedifferentiation of adjacent non-malignant BPH-1 epithelial cells. CONCLUSIONS: These data show that human prostatic fibroblasts have the capacity for Hh signaling and manipulation. Increased expression of a signature of prostatic Hh genes in the prostate tumor microenvironment suggests a role in the epithelial transformations driving prostate cancer (PCa).


Asunto(s)
Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular , Proteínas Hedgehog/fisiología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , Transducción de Señal/fisiología , Animales , Benzamidas/farmacología , Transformación Celular Neoplásica/patología , Células Cultivadas , Epitelio/patología , Fibroblastos/patología , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/genética , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Morfolinas/farmacología , Purinas/farmacología , Quinazolinas/farmacología , Células del Estroma/patología
14.
Biol Reprod ; 86(3): 80, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22133695

RESUMEN

The signaling molecule DHH, secreted by Sertoli cells, has essential regulatory functions in testicular differentiation. DHH is required for the differentiation of peritubular myoid cells that line the seminiferous cords and steroidogenic Leydig cells. The testicular cords in Dhh-null male mice lack a basal lamina and develop abnormally. To date, the DHH-signaling pathway has never been examined outside of any eutherian mammals. This study examined the effects of inhibition of DHH signaling in a marsupial mammal, the tammar wallaby, by culturing gonads in vitro in the presence of the hedgehog-signaling inhibitors cyclopamine and forskolin. Disruption of hedgehog signaling in the tammar testes caused highly disorganized cord formation. SOX9 protein remained strongly expressed in Sertoli cells, laminin distribution was highly fragmented, and germ cells were distributed around the cortical regions of treated testes in an ovarianlike morphology. This suggests that hedgehog signaling regulates cord formation in the tammar wallaby testis as it does in eutherian mammals. These data demonstrate that the hedgehog pathway has been highly conserved in mammals for at least 160 million years.


Asunto(s)
Proteínas Hedgehog/fisiología , Marsupiales/embriología , Marsupiales/fisiología , Túbulos Seminíferos/embriología , Transducción de Señal/fisiología , Testículo/embriología , Animales , Colforsina/farmacología , Desarrollo Fetal/fisiología , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/efectos de los fármacos , Masculino , Técnicas de Cultivo de Órganos , Factor de Transcripción SOX9/metabolismo , Túbulos Seminíferos/citología , Túbulos Seminíferos/efectos de los fármacos , Células de Sertoli/metabolismo , Testículo/citología , Testículo/efectos de los fármacos , Alcaloides de Veratrum/farmacología
15.
Eur Cell Mater ; 23: 121-32; discussion 133-4, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22370795

RESUMEN

Hydrostatic pressure (HP) is a key component of the in vivo joint environment and has been shown to enhance chondrogenesis of stem cells. The objective of this study was to investigate the interaction between HP and TGF-ß3 on both the initiation and maintenance of a chondrogenic phenotype for joint tissue derived stem cells. Pellets generated from porcine chondrocytes (CCs), synovial membrane derived stem cells (SDSCs) and infrapatellar fat pad derived stem cells (FPSCs) were subjected to 10 MPa of cyclic HP (4 h/day) and different concentrations of TGF-ß3 (0, 1 and 10 ng/mL) for 14 days. CCs and stem cells were observed to respond differentially to both HP and TGF-ß3 stimulation. HP in the absence of TGF-ß3 did not induce robust chondrogenic differentiation of stem cells. At low concentrations of TGF-ß3 (1 ng/mL), HP acted to enhance chondrogenesis of both SDSCs and FPSCs, as evident by a 3-fold increase in Sox9 expression and a significant increase in glycosaminoglycan accumulation. In contrast, HP had no effect on cartilage-specific matrix synthesis at higher concentrations of TGF-ß3 (10 ng/mL). Critically, HP appears to play a key role in the maintenance of a chondrogenic phenotype, as evident by a down-regulation of the hypertrophic markers type X collagen and Indian hedgehog in SDSCs irrespective of the cytokine concentration. In the context of stem cell based therapies for cartilage repair, this study demonstrates the importance of considering how joint specific environmental factors interact to regulate not only the initiation of chondrogenesis, but also the development of a stable hyaline-like repair tissue.


Asunto(s)
Cartílago/citología , Condrocitos/metabolismo , Condrogénesis/efectos de los fármacos , Presión Hidrostática , Articulación Patelofemoral/citología , Células Madre/metabolismo , Factor de Crecimiento Transformador beta3/administración & dosificación , Animales , Cartílago/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Colágeno Tipo X/efectos de los fármacos , Colágeno Tipo X/metabolismo , Glicosaminoglicanos/metabolismo , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Articulación Patelofemoral/metabolismo , Fenotipo , Factor de Transcripción SOX9/efectos de los fármacos , Factor de Transcripción SOX9/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos , Porcinos , Membrana Sinovial/citología , Membrana Sinovial/metabolismo
17.
Pharmazie ; 67(6): 475-81, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22822532

RESUMEN

The hedgehog (Hh) protein is involved in angiogenesis and cardiovascular development via activation of the classical ligand-dependent signaling transduction. So its potential therapeutic meaning of Hh signaling proteins to the ischemic heart diseases has been greatly explored. Recent studies show that up-regulated expression of hypoxia-induced factor-1 (HIF-1) and inflamemation in ischemic tissues activate the Hh signaling cascade in a GLI-dependent or independent way, resulting in elevated expression levels of pro-angiogenic and agiogenic factors to facilitate angiogenesis. In addition, Hh signaling pathway activation can promote residual myocardial progenitors, endogenous EPCs and MSCs differentiating into cardiomyocytes, inhibit cardiomyocyte apoptosis; thirdly, high level of exogenous Hh signaling can reduce myocardial ischemic/reperfusion injuries(I/R). In conclusion, three kinds of mechanisms induced by Hh signaling pathway participate in the heart repair after myocardial ischemia. Therefore, Hh agonists including Hh protein, Hh gene transfer and small molecule agonist could be part of a potential therapeutic strategy for acute or chronic ischemic heart disease.


Asunto(s)
Proteínas Hedgehog/fisiología , Isquemia Miocárdica/tratamiento farmacológico , Transducción de Señal/fisiología , Animales , Cardiotónicos/farmacología , Sistema Cardiovascular/crecimiento & desarrollo , Proteínas Hedgehog/efectos de los fármacos , Humanos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Transducción de Señal/efectos de los fármacos , Transactivadores/genética
19.
Neurol Res ; 44(4): 318-330, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34592910

RESUMEN

OBJECTIVE: Ischemic stroke is a major cause of death in the global population, with a high disability and mortality rate. Lack of regenerative ability is considered to be the fundamental cause. This study aims to determine the effect of Shh pathway, which mediates regenerative signaling in response to CNS injury, on myelin repair and Olig1 expression in focal ischemic lesions in the rat. METHODS: A model of middle cerebral artery occlusion (MCAO) was established using the intraluminal suture method where the middle cerebral artery (MCA) was restricted for 120 min. Cyclopamine, a specific inhibitor of Shh, or saline was administered 12h after MCAO surgery and lasted for 7d. After MCA occlusion, male Sprague-Dawley rats were randomly allocated to cyclopamine- or saline-treated groups. A group of no-injection animals after MCAO were used as control. The Shh signaling pathway, myelinogenesis-related factor MBP and Olig1 were tested using immunohistochemistry and RT-PCR assay. RESULTS: The levels of Shh and its component Gli1 were elevated from 1d up to 14d following ischemia, indicating that the Shh-Gli1 axis was broadly reactivated. Treatment with cyclopamine can partially block the Shh signaling pathway, prevent myelin repair, and decrease the Olig1 expression following ischemic stroke. CONCLUSION: That blockade of Shh signaling concurrently with the creation of a lesion aggravated ischemic myelin damage, probably via its downstream effects on Olig1 transcription. Shh plays a contributory role during regeneration in the CNS, thereby providing promising new therapeutic strategies to assist in recovery from ischemic stroke.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Hedgehog/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/metabolismo , Alcaloides de Veratrum/farmacología , Proteína con Dedos de Zinc GLI1/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/efectos de los fármacos , Modelos Animales de Enfermedad , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/efectos de los fármacos , Masculino , Regeneración Nerviosa/efectos de los fármacos , Proteínas del Tejido Nervioso/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína con Dedos de Zinc GLI1/efectos de los fármacos
20.
Curr Probl Cancer ; 45(6): 100736, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33894989

RESUMEN

Basal cell carcinomas occur in up to 39% of Caucasian men and 28% of women. Rarely it can present a management dilemma in patients with neglected locally advanced disease of large dimension or involvement of critical structures. The Hedgehog pathway is constitutively active in almost all basal cell carcinomas and patients with Naevoid Basal Cell Carcinoma Syndrome have germline mutations in the Patched tumor suppressor gene, a Hedgehog pathway component, on chromosome 9q. This case describes an elderly patient with an untreated sporadic Basal cell carcinoma whose dimensions precluded local management approaches. The Hedgehog pathway inhibitor Vismodegib had a dramatic response allowing definitive treatment to be pursued.


Asunto(s)
Anilidas/farmacología , Carcinoma Basocelular/tratamiento farmacológico , Carcinoma Basocelular/genética , Proteínas Hedgehog/efectos de los fármacos , Piridinas/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Anciano de 80 o más Años , Carcinoma Basocelular/patología , Humanos , Masculino , Terapia Neoadyuvante/métodos , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA