Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 132024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38591541

RESUMEN

Collective cell migration is fundamental for the development of organisms and in the adult for tissue regeneration and in pathological conditions such as cancer. Migration as a coherent group requires the maintenance of cell-cell interactions, while contact inhibition of locomotion (CIL), a local repulsive force, can propel the group forward. Here we show that the cell-cell interaction molecule, N-cadherin, regulates both adhesion and repulsion processes during Schwann cell (SC) collective migration, which is required for peripheral nerve regeneration. However, distinct from its role in cell-cell adhesion, the repulsion process is independent of N-cadherin trans-homodimerisation and the associated adherens junction complex. Rather, the extracellular domain of N-cadherin is required to present the repulsive Slit2/Slit3 signal at the cell surface. Inhibiting Slit2/Slit3 signalling inhibits CIL and subsequently collective SC migration, resulting in adherent, nonmigratory cell clusters. Moreover, analysis of ex vivo explants from mice following sciatic nerve injury showed that inhibition of Slit2 decreased SC collective migration and increased clustering of SCs within the nerve bridge. These findings provide insight into how opposing signals can mediate collective cell migration and how CIL pathways are promising targets for inhibiting pathological cell migration.


Asunto(s)
Cadherinas , Movimiento Celular , Inhibición de Contacto , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana , Regeneración Nerviosa , Proteínas del Tejido Nervioso , Células de Schwann , Células de Schwann/metabolismo , Células de Schwann/fisiología , Animales , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Ratones , Cadherinas/metabolismo , Cadherinas/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Regeneración Nerviosa/fisiología , Locomoción/fisiología , Adhesión Celular , Transducción de Señal
2.
EMBO J ; 39(1): e102030, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31774199

RESUMEN

Glioblastoma is one of the most lethal forms of adult cancer with a median survival of around 15 months. A potential treatment strategy involves targeting glioblastoma stem-like cells (GSC), which constitute a cell autonomous reservoir of aberrant cells able to initiate, maintain, and repopulate the tumor mass. Here, we report that the expression of the paracaspase mucosa-associated lymphoid tissue l (MALT1), a protease previously linked to antigen receptor-mediated NF-κB activation and B-cell lymphoma survival, inversely correlates with patient probability of survival. The knockdown of MALT1 largely impaired the expansion of patient-derived stem-like cells in vitro, and this could be recapitulated with pharmacological inhibitors, in vitro and in vivo. Blocking MALT1 protease activity increases the endo-lysosome abundance, impairs autophagic flux, and culminates in lysosomal-mediated cell death, concomitantly with mTOR inactivation and dispersion from endo-lysosomes. These findings place MALT1 as a new druggable target involved in glioblastoma and unveil ways to modulate the homeostasis of endo-lysosomes.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Endosomas/patología , Glioma/patología , Homeostasis , Lisosomas/patología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Células Madre Neoplásicas/patología , Anciano , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Endosomas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/metabolismo , Humanos , Activación de Linfocitos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Células Madre Neoplásicas/metabolismo , Proteolisis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Rep ; 25(10): 2755-2765.e5, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30517863

RESUMEN

The formation of myelinating Schwann cells (mSCs) involves the remarkable biogenic process, which rapidly generates the myelin sheath. Once formed, the mSC transitions to a stable homeostatic state, with loss of this stability associated with neuropathies. The histone deacetylases histone deacetylase 1 (HDAC1) and HDAC2 are required for the myelination transcriptional program. Here, we show a distinct role for HDAC3, in that, while dispensable for the formation of mSCs, it is essential for the stability of the myelin sheath once formed-with loss resulting in progressive severe neuropathy in adulthood. This is associated with the prior failure to downregulate the biogenic program upon entering the homeostatic state leading to hypertrophy and hypermyelination of the mSCs, progressing to the development of severe myelination defects. Our results highlight distinct roles of HDAC1/2 and HDAC3 in controlling the differentiation and homeostatic states of a cell with broad implications for the understanding of this important cell-state transition.


Asunto(s)
Histona Desacetilasas/metabolismo , Homeostasis , Vaina de Mielina/metabolismo , Células de Schwann/citología , Células de Schwann/enzimología , Envejecimiento/metabolismo , Animales , Ratones Endogámicos C57BL , Vaina de Mielina/ultraestructura , Ratas , Nervio Ciático/metabolismo , Nervio Ciático/ultraestructura , Transcripción Genética
4.
J Exp Neurosci ; 12: 1179069518759680, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29535551

RESUMEN

Glioblastoma multiforme are mortifying brain tumors that contain a subpopulation of tumor cells with stem-like properties, termed as glioblastoma stem-like cells (GSCs). These GSCs constitute an autonomous reservoir of aberrant cells able to initiate, maintain, and repopulate the tumor mass. A new therapeutic strategy would consist of targeting the GSC population. The GSCs are situated in perivascular niches, closely associated with brain microvascular endothelial cells thereby involved in bidirectional molecular and cellular interactions. In this scenario, the endothelium not only supplies oxygen and necessary nutrients but also seeds a protective microenvironment for tumor growth. Although GSC fate, plasticity, and survival are regulated by external cues emanating from endothelial cells, the nature of such angiocrine signals remains unknown. Our laboratory conclusively demonstrated that brain endothelial cells positively control the expansion of GSCs.1 Notably, we found that GSCs are addicted to the hormonal peptide apelin (APLN) secreted by surrounding endothelial cells, and identified the APLN/APLNR nexus as a promising druggable network in glioblastoma.

5.
Brain ; 140(11): 2939-2954, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29053791

RESUMEN

Glioblastoma are highly aggressive brain tumours that are associated with an extremely poor prognosis. Within these tumours exists a subpopulation of highly plastic self-renewing cancer cells that retain the ability to expand ex vivo as tumourspheres, induce tumour growth in mice, and have been implicated in radio- and chemo-resistance. Although their identity and fate are regulated by external cues emanating from endothelial cells, the nature of such signals remains unknown. Here, we used a mass spectrometry proteomic approach to characterize the factors released by brain endothelial cells. We report the identification of the vasoactive peptide apelin as a central regulator for endothelial-mediated maintenance of glioblastoma patient-derived cells with stem-like properties. Genetic and pharmacological targeting of apelin cognate receptor abrogates apelin- and endothelial-mediated expansion of glioblastoma patient-derived cells with stem-like properties in vitro and suppresses tumour growth in vivo. Functionally, selective competitive antagonists of apelin receptor were shown to be safe and effective in reducing tumour expansion and lengthening the survival of intracranially xenografted mice. Therefore, the apelin/apelin receptor signalling nexus may operate as a paracrine signal that sustains tumour cell expansion and progression, suggesting that apelin is a druggable factor in glioblastoma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Apelina , Receptores de Apelina , Neoplasias Encefálicas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales , Glioblastoma/tratamiento farmacológico , Células HEK293 , Humanos , Técnicas In Vitro , Espectrometría de Masas , Ratones , Terapia Molecular Dirigida , Proteómica , ARN Interferente Pequeño , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 7(41): 66865-66879, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27589691

RESUMEN

Glioblastoma multiforme (GBM) is a highly aggressive tumour of the central nervous system and is associated with an extremely poor prognosis. Within GBM exists a subpopulation of cells, glioblastoma-initiating cells (GIC), which possess the characteristics of progenitor cells, have the ability to initiate tumour growth and resist to current treatment strategies. We aimed at identifying novel specific inhibitors of GIC expansion through use of a large-scale chemical screen of approved small molecules. Here, we report the identification of the natural compound ß-escin as a selective inhibitor of GIC viability. Indeed, ß-escin was significantly cytotoxic in nine patient-derived GIC, whilst exhibiting no substantial effect on the other human cancer or control cell lines tested. In addition, ß-escin was more effective at reducing GIC growth than current clinically used cytotoxic agents. We further show that ß-escin triggers caspase-dependent cell death combined with a loss of stemness properties. However, blocking apoptosis could not rescue the ß-escin-induced reduction in sphere formation or stemness marker activity, indicating that ß-escin directly modifies the stem identity of GIC, independent of the induction of cell death. Thus, this study has repositioned ß-escin as a promising potential candidate to selectively target the aggressive population of initiating cells within GBM.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Escina/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Anciano , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Autorrenovación de las Células/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Escina/química , Femenino , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Estructura Molecular , Células Tumorales Cultivadas
8.
Front Pharmacol ; 6: 281, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26635611

RESUMEN

Glioblastoma multiforme (GBM) constitutes the most common and the most aggressive type of human tumors affecting the central nervous system. Prognosis remains dark due to the inefficiency of current treatments and the rapid relapse. Paralleling other human tumors, GBM contains a fraction of tumor initiating cells with the capacity to self-renew, initiate and maintain the tumor mass. These cells were found in close proximity to brain vasculature, suggesting functional interactions between brain tumor-initiating cells (BTICs) and endothelial cells within the so-called vascular niche. However, the mechanisms by which these cells impact on the endothelium plasticity and function remain unclear. Using culture of BTICs isolated from a cohort of 14 GBM patients, we show that BTICs secretome promotes brain endothelial cell remodeling in a VEGF-independent manner. Gene array analysis unmasked that BTICs-released factors drove the expression of Ptch2 in endothelial cells. Interestingly, BTICs produce desert hedgehog (DHH) ligand, enabling a paracrine DHH/Ptch2 signaling cascade that conveys elevated permeability and angiogenesis. Finally, DHH silencing in BTICs dramatically reduced tumor growth, as well as vascularization and intra-tumor permeability. Collectively, our data unveil a role for DHH in exacerbated tumor angiogenesis and permeability, which may ultimately favor glioblastoma growth, and thus place the DHH/Ptch2 nexus as a molecular target for novel therapies.

9.
Recent Pat CNS Drug Discov ; 9(2): 110-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25386916

RESUMEN

Evidence for the involvement of the Substance P (SP)/NK1 receptor system in the development and progression of cancer strongly supports its potential as a therapeutic target in malignancies. Novel strategies for approaching cancer treatment are urgently required particularly with regard to tumours of the central nervous system (CNS), which are notoriously difficult to effectively treat and associated with extremely poor prognosis for many patients. This is due, in part, to the presence of the highly specialised blood-brain barrier, which is known to restrict common treatments such as chemotherapy and hinder early tumour diagnosis. Additionally, tumours of the CNS are difficult to surgically resect completely, often contributing to the resurgence of the disease many years later. Interestingly, despite the presence of the blood-brain barrier, circulating tumour cells are able to gain entry to the brain and form secondary brain tumours; however, the underlying mechanisms of this process remain unclear. Tachykinins, in particular Substance P, have been implicated in early blood-brain barrier disruption via neurogenic inflammation in a number of other CNS pathologies. Recent evidence also suggests that Substance P may play a central role in the development of CNS tumours. It has been well established that a number of tumour cells express Substance P, NK1 receptors and mRNA for the tachykinin NK1 receptor. This increase in the Substance P/NK1 receptor system is known to induce proliferation and migration of tumour cells as well as stimulate angiogenesis, thus contributing to tumour progression. Accordingly, the NK1 receptor antagonist presents a novel target for anti-cancer therapy for which a number of patents have been filed. This review will examine the role of Substance P in the development of CNS tumours and its potential application as an anti-cancer agent.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Sustancia P/antagonistas & inhibidores , Sustancia P/uso terapéutico , Animales , Humanos , Receptores de Neuroquinina-1/metabolismo
10.
PLoS One ; 9(8): e104565, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25105800

RESUMEN

Red/near-infrared irradiation therapy (R/NIR-IT) delivered by laser or light-emitting diode (LED) has improved functional outcomes in a range of CNS injuries. However, translation of R/NIR-IT to the clinic for treatment of neurotrauma has been hampered by lack of comparative information regarding the degree of penetration of the delivered irradiation to the injury site and the optimal treatment parameters for different CNS injuries. We compared the treatment efficacy of R/NIR-IT at 670 nm and 830 nm, provided by narrow-band LED arrays adjusted to produce equal irradiance, in four in vivo rat models of CNS injury: partial optic nerve transection, light-induced retinal degeneration, traumatic brain injury (TBI) and spinal cord injury (SCI). The number of photons of 670 nm or 830 nm light reaching the SCI injury site was 6.6% and 11.3% of emitted light respectively. Treatment of rats with 670 nm R/NIR-IT following partial optic nerve transection significantly increased the number of visual responses at 7 days after injury (P ≤ 0.05); 830 nm R/NIR-IT was partially effective. 670 nm R/NIR-IT also significantly reduced reactive species and both 670 nm and 830 nm R/NIR-IT reduced hydroxynonenal immunoreactivity (P ≤ 0.05) in this model. Pre-treatment of light-induced retinal degeneration with 670 nm R/NIR-IT significantly reduced the number of Tunel+ cells and 8-hydroxyguanosine immunoreactivity (P ≤ 0.05); outcomes in 830 nm R/NIR-IT treated animals were not significantly different to controls. Treatment of fluid-percussion TBI with 670 nm or 830 nm R/NIR-IT did not result in improvements in motor or sensory function or lesion size at 7 days (P>0.05). Similarly, treatment of contusive SCI with 670 nm or 830 nm R/NIR-IT did not result in significant improvements in functional recovery or reduced cyst size at 28 days (P>0.05). Outcomes from this comparative study indicate that it will be necessary to optimise delivery devices, wavelength, intensity and duration of R/NIR-IT individually for different CNS injury types.


Asunto(s)
Lesiones Encefálicas/radioterapia , Traumatismos del Nervio Óptico/radioterapia , Degeneración Retiniana/radioterapia , Traumatismos de la Médula Espinal/radioterapia , Animales , Encéfalo/patología , Encéfalo/efectos de la radiación , Lesiones Encefálicas/patología , Femenino , Rayos Infrarrojos , Masculino , Nervio Óptico/patología , Nervio Óptico/efectos de la radiación , Traumatismos del Nervio Óptico/patología , Ratas Sprague-Dawley , Retina/patología , Retina/efectos de la radiación , Degeneración Retiniana/patología , Médula Espinal/patología , Médula Espinal/efectos de la radiación , Traumatismos de la Médula Espinal/patología
11.
PLoS One ; 9(5): e97002, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24818961

RESUMEN

The neuropeptide substance P (SP) has been implicated in the disruption of the blood-brain barrier (BBB) and development of cerebral edema in acute brain injury. Cerebral edema accumulates rapidly around brain tumors and has been linked to several tumor-associated deficits. Currently, the standard treatment for peritumoral edema is the corticosteroid dexamethasone, prolonged use of which is associated with a number of deleterious side effects. As SP is reported to increase in many cancer types, this study examined whether SP plays a role in the genesis of brain peritumoral edema. A-375 human melanoma cells were injected into the right striatum of male Balb/c nude mice to induce brain tumor growth, with culture medium injected in animals serving as controls. At 2, 3 or 4 weeks following tumor cell inoculation, non-treated animals were perfusion fixed for immunohistochemical detection of Albumin, SP and NK1 receptor. A further subgroup of animals was treated with a daily injection of the NK1 antagonist Emend (3 mg/kg), dexamethasone (8 mg/kg) or saline vehicle at 3 weeks post-inoculation. Animals were sacrificed a week later to determine BBB permeability using Evan's Blue and brain water content. Non-treated animals demonstrated a significant increase in albumin, SP and NK1 receptor immunoreactivity in the peritumoral area as well as increased perivascular staining in the surrounding brain tissue. Brain water content and BBB permeability was significantly increased in tumor-inoculated animals when compared to controls (p<0.05). Treatment with Emend and dexamethasone reduced BBB permeability and brain water content when compared to vehicle-treated tumor-inoculated mice. The increase in peritumoral staining for both SP and the NK1 receptor, coupled with the reduction in brain water content and BBB permeability seen following treatment with the NK1 antagonist Emend, suggests that SP plays a role in the genesis of peritumoral edema, and thus warrants further investigation as a potential anti-edematous treatment.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Neoplasias Encefálicas/complicaciones , Morfolinas/farmacología , Antagonistas del Receptor de Neuroquinina-1/farmacología , Receptores de Neuroquinina-1/metabolismo , Animales , Aprepitant , Barrera Hematoencefálica/metabolismo , Edema Encefálico/complicaciones , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Morfolinas/uso terapéutico , Antagonistas del Receptor de Neuroquinina-1/uso terapéutico , Permeabilidad/efectos de los fármacos , Sustancia P/metabolismo
12.
Recent Pat CNS Drug Discov ; 8(1): 13-23, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23477306

RESUMEN

Despite recent advances in cancer treatment and diagnosis, the prognosis for patients with CNS tumours remains extremely poor. This is, in part, due to the difficulty in completely removing tumours surgically, and also because of the presence of the blood brain barrier, which can prevent the entry of chemotherapeutic agents typically used in cancer treatment. Despite the presence of the blood brain barrier, tumour cells are capable of entering and colonising the brain to form secondary brain tumours. Additionally, tumour related disruption of the blood brain barrier is associated with the clinical presentation of many patients, with accompanying increases in intracranial pressure due, in part, to the development of vasogenic oedema. Vasogenic oedema results because the newly formed angiogenic vessels within brain tumours do not retain the highly selective properties of the blood brain barrier, and thus allow for the extravasation of plasma proteins and water into the brain parenchyma. Tachykinins, and in particular substance P, have been implicated in blood brain barrier disruption and the genesis of cerebral oedema in other CNS insults via a process known as neurogenic inflammation. Recent evidence suggests that substance P may play a similar role in CNS tumours. It has been well established that an upregulation of substance P and its receptors occurs in a number of different cancer types, including CNS neoplasms. In addition to disrupting blood brain barrier permeability, substance P and the NK1 receptors facilitate promotion of tumour growth and the development of cerebral oedema. Accordingly, recent patents describe the potential of NK1 receptor antagonists as anti-cancer agents suggesting that substance P may provide a novel cancer treatment target. This review will examine the role of substance P in the development of CNS tumours.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Sustancia P/antagonistas & inhibidores , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiopatología , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/fisiopatología , Humanos , Modelos Neurológicos , Terapia Molecular Dirigida , Antagonistas del Receptor de Neuroquinina-1/farmacología , Receptores de Neuroquinina-1/fisiología , Sustancia P/fisiología
13.
Anticancer Drugs ; 24(4): 344-54, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23407059

RESUMEN

Emend, an NK1 antagonist, and dexamethasone are used to treat complications associated with metastatic brain tumours and their treatment. It has been suggested that these agents exert anticancer effects apart from their current use. The effects of the NK1 antagonists, Emend and N-acetyl-L-tryptophan, and dexamethasone on tumour growth were investigated in vitro and in vivo at clinically relevant doses. For animal experiments, a stereotaxic injection model of Walker 256 rat breast carcinoma cells into the striatum of Wistar rats was used. Emend treatment led to a decrease in tumour cell viability in vitro, although this effect was not replicated by N-acetyl-L-tryptophan. Dexamethasone did not decrease tumour cell viability in vitro but decreased tumour volume in vivo, likely to be through a reduction in tumour oedema, as indicated by the increase in tumour cell density. None of the agents investigated altered tumour cell replication or apoptosis in vivo. Inoculated animals showed increased glial fibrillary acidic protein and ionized calcium-binding adapter molecule 1 immunoreactivity indicative of astrocytes and microglia in the peritumoral area, whereas treatment with Emend and dexamethasone reduced the labelling for both glial cells. These results do not support the hypothesis that NK1 antagonists or dexamethasone exert a cytotoxic action on tumour cells, although these conclusions may be specific to this model and cell line.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/secundario , Carcinoma/secundario , Dexametasona/farmacología , Neoplasias Mamarias Experimentales/secundario , Morfolinas/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Antagonistas del Receptor de Neuroquinina-1 , Triptófano/análogos & derivados , Animales , Aprepitant , Astrocitos/patología , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Proteínas de Unión al Calcio/análisis , Carcinoma/tratamiento farmacológico , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/trasplante , Cuerpo Estriado/patología , Dexametasona/administración & dosificación , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Proteína Ácida Fibrilar de la Glía/análisis , Humanos , Técnicas In Vitro , Masculino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Proteínas de Microfilamentos/análisis , Microglía/patología , Modelos Biológicos , Morfolinas/administración & dosificación , Morfolinas/uso terapéutico , Proteínas de Neoplasias/fisiología , Distribución Aleatoria , Ratas , Ratas Wistar , Receptores de Neuroquinina-1/fisiología , Triptófano/administración & dosificación , Triptófano/farmacología , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral
14.
Cancer Cell Int ; 13(1): 5, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23374226

RESUMEN

BACKGROUND: Metastatic brain tumours are a common end stage of breast cancer progression, with significant associated morbidity and high mortality. Walker 256 is a rat breast carcinoma cell line syngeneic to Wistar rats and commonly used to induce secondary brain tumours. Previously there has been the assumption that the same cancer cell line from different cell banks behave in a similar manner, although recent studies have suggested that cell lines may change their characteristics over time in vitro. METHODS: In this study internal carotid artery injection and direct cerebral inoculation models of secondary brain tumours were used to determine the tumorigenicity of Walker 256 cells obtained from two cell banks, the American Type Culture Collection (ATCC), and the Cell Resource Centre for Medical Research at Tohoku University (CRCTU). RESULTS: Tumour incidence and volume, plus immunoreactivity to albumin, IBA1 and GFAP, were used as indicators of tumorigenicity and tumour interaction with the host brain microenvironment. CRCTU Walker 256 cells showed greater incidence, larger tumour volume, pronounced blood-brain barrier disruption and prominent glial response when compared to ATCC cell line. CONCLUSIONS: These findings indicate that immortalised cancer cell lines obtained from different cell banks may have diverse characteristics and behaviour in vivo.

15.
Clin Exp Metastasis ; 30(1): 1-12, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22610781

RESUMEN

It is not yet known how tumour cells traverse the blood-brain barrier (BBB) to form brain metastases. Substance P (SP) release is a key component of neurogenic inflammation which has been recently shown to increase the permeability of the BBB following CNS insults, making it a possible candidate as a mediator of tumour cell extravasation into the brain. This study investigated the properties of the BBB in the early stages of tumour cell invasion into the brain, and the possible involvement of SP. Male Wistar rats were injected with Walker 256 breast carcinoma cells via the internal carotid artery and euthanised at 1, 3, 6 and 9 days post tumour inoculation. Culture medium-injected animals served as controls at 1 and 9 days. Evidence of tumour cell extravasation across the BBB was first observed at 3 days post-inoculation, which corresponded with significantly increased albumin (p < 0.05) and SP immunoreactivity (p < 0.01) and significantly reduced endothelial barrier antigen labelling of microvessels when compared to culture medium control animals (p < 0.001). By day 9 after tumour cell inoculation, 100 % of animals developed large intracranial neoplasms that had significantly increased albumin in the peri-tumoral area (p < 0.001). The increased SP immunoreactivity and altered BBB properties at 3 days post-inoculation that coincided with early tumour invasion may be indicative of a mechanism for tumour cell extravasation into the brain. Thus, extravasation of tumour cells into the brain to form cerebral metastases may be a SP-mediated process.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas/patología , Carcinoma 256 de Walker/patología , Neoplasias Mamarias Animales/patología , Sustancia P/metabolismo , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/secundario , Permeabilidad Capilar , Carcinoma 256 de Walker/metabolismo , Femenino , Inmunohistoquímica , Masculino , Neoplasias Mamarias Animales/metabolismo , Ratas , Ratas Wistar , Células Tumorales Cultivadas
16.
J Neuroimmunol ; 250(1-2): 59-65, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22722013

RESUMEN

Dexamethasone, the standard treatment for peritumoral brain oedema, inhibits classical inflammation. Neurogenic inflammation, which acts via substance P (SP), has been implicated in vasogenic oedema in animal models of CNS injury. SP is elevated within and outside CNS tumours. This study investigated the efficacy of NK1 receptor antagonists, which block SP, compared with dexamethasone treatment, in a rat model of tumorigenesis. Dexamethasone reverted normal brain water content and reduced Evans blue and albumin extravasation, while NK1 antagonists did not ameliorate oedema formation. We conclude that classical inflammation rather than neurogenic inflammation drives peritumoral oedema in this brain tumour model.


Asunto(s)
Antiinflamatorios/farmacología , Edema Encefálico/tratamiento farmacológico , Neoplasias Encefálicas/complicaciones , Dexametasona/farmacología , Inflamación/tratamiento farmacológico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/etiología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/metabolismo , Masculino , Antagonistas del Receptor de Neuroquinina-1 , Ratas , Ratas Wistar , Sustancia P/biosíntesis
17.
Recent Pat CNS Drug Discov ; 6(1): 31-40, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21073431

RESUMEN

Cancers of the brain are intrinsically more complicated to treat than systemic malignancies due to the unique anatomical features of the brain. The blood-brain barrier prevents chemotherapeutic agents from reaching brain neoplasms, and angiogenesis occurs as the metabolic needs of the tumour increase, thus further complicating treatment. The newly formed blood vessels form the blood-tumour barrier and are distinct from the blood-brain barrier in that they are more permeable. Being more permeable, these abnormal blood vessels lead to the formation of peri-tumoural edema, which is the cause of much morbidity and mortality associated with central nervous system neoplasms. While the cause of the increased permeability is unclear, kinins have been implicated in regulating the permeability of normal vasculature. Kinins are also known to exert many inflammatory actions affecting both normal and angiogenic blood vessels, as well as tumour cells. The vasodilatory and vascular permeabilizing effects of kinins, and particularly bradykinin and substance P, have been investigated with regard to delivery of chemotherapeutic agents to neoplastic brain tissue through both vascular barriers. In contrast, kinin receptor antagonists have been found to exert effects on tumour cells that result in decreased angiogenesis, tumour cell motility and growth. Thus, many recent patents describe kinin activity on brain vasculature, which may play an integral role in the development of treatments for malignancies in the central nervous system through amelioration of angiogenesis. In conjunction, patents that discuss the ability of kinins to decrease tumour cell migration and proliferation demonstrate that kinins may offer novel approaches to brain tumour therapy in the future.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Descubrimiento de Drogas , Cininas/farmacología , Moduladores de la Angiogénesis/metabolismo , Moduladores de la Angiogénesis/farmacología , Moduladores de la Angiogénesis/uso terapéutico , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/metabolismo , Progresión de la Enfermedad , Humanos , Cininas/metabolismo , Cininas/uso terapéutico
18.
Neurosci Lett ; 481(1): 26-9, 2010 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-20600611

RESUMEN

Angiotensin-converting enzyme (ACE) inhibitors are widely used as blood pressure medications in hypertensive individuals. However, ACE inhibitors also play an integral role in the breakdown of neuronal substance P, which has been recently implicated in the development of functional deficits following traumatic brain injury (TBI). The present study therefore examined the effects of ACE inhibitors on histological and motor outcome following TBI. Male Sprague-Dawley rats were treated with Captopril, Enalapril or equal volume saline for 7 days prior to the induction of diffuse TBI using the impact acceleration model. At 5h post-injury, animals administered Captopril demonstrated significantly increased substance P immunoreactivity compared to vehicle controls (p<0.01), and increased dark cell change that persisted to 7 days post-trauma. Captopril also resulted in exacerbated motor deficits compared to vehicle treated animals (p<0.05) as assessed by the rotarod test over a 7-day post-traumatic period. Administration of the alternative ACE inhibitor, Enalapril, likewise exacerbated motor deficits, confirming a class effect of ACE inhibitors rather than a compound effect specific to Captopril. We conclude that ACE inhibitors are deleterious to outcome following TBI, presumably by impairing the degradation of substance P and increasing substance P mediated neuronal injury.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Lesiones Encefálicas/complicaciones , Trastornos del Movimiento/etiología , Trastornos del Movimiento/patología , Análisis de Varianza , Animales , Presión Sanguínea/efectos de los fármacos , Captopril/efectos adversos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Enalapril/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante/métodos , Sustancia P/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...