Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 320(4): H1370-H1392, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33543687

RESUMEN

Age-related blood-brain barrier (BBB) disruption and cerebromicrovascular rarefaction contribute importantly to the pathogenesis of both vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). Recent advances in geroscience research enable development of novel interventions to reverse age-related alterations of the cerebral microcirculation for prevention of VCID and AD. To facilitate this research, there is an urgent need for sensitive and easy-to-adapt imaging methods that enable longitudinal assessment of changes in BBB permeability and brain capillarization in aged mice and that could be used in vivo to evaluate treatment efficiency. To enable longitudinal assessment of changes in BBB permeability in aged mice equipped with a chronic cranial window, we adapted and optimized two different intravital two-photon imaging approaches. By assessing relative fluorescence changes over the baseline within a volume of brain tissue, after qualitative image subtraction of the brain microvasculature, we confirmed that, in 24-mo-old C57BL/6J mice, cumulative permeability of the microvessels to fluorescent tracers of different molecular masses (0.3 to 40 kDa) is significantly increased compared with that of 5-mo-old mice. Real-time recording of vessel cross-sections showed that apparent solute permeability of single microvessels is significantly increased in aged mice vs. young mice. Cortical capillary density, assessed both by intravital two-photon microscopy and optical coherence tomography was also decreased in aged mice vs. young mice. The presented methods have been optimized for longitudinal (over the period of 36 wk) in vivo assessment of cerebromicrovascular health in preclinical geroscience research.NEW & NOTEWORTHY Methods are presented for longitudinal detection of age-related increase in blood-brain barrier permeability and microvascular rarefaction in the mouse cerebral cortex by intravital two-photon microscopy and optical coherence tomography.


Asunto(s)
Envejecimiento/patología , Barrera Hematoencefálica/diagnóstico por imagen , Permeabilidad Capilar , Corteza Cerebral/irrigación sanguínea , Microscopía Intravital , Microscopía de Fluorescencia por Excitación Multifotónica , Rarefacción Microvascular , Microvasos/diagnóstico por imagen , Tomografía de Coherencia Óptica , Factores de Edad , Envejecimiento/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Masculino , Ratones Endogámicos C57BL , Densidad Microvascular , Microvasos/metabolismo , Microvasos/patología , Factores de Tiempo
2.
Int J Mol Sci ; 22(7)2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33810538

RESUMEN

Spreading depolarization (SD) is a wave of mass depolarization that causes profound perfusion changes in acute cerebrovascular diseases. Although the astrocyte response is secondary to the neuronal depolarization with SD, it remains to be explored how glial activity is altered after the passage of SD. Here, we describe post-SD high frequency astrocyte Ca2+ oscillations in the mouse somatosensory cortex. The intracellular Ca2+ changes of SR101 labeled astrocytes and the SD-related arteriole diameter variations were simultaneously visualized by multiphoton microscopy in anesthetized mice. Post-SD astrocyte Ca2+ oscillations were identified as Ca2+ events non-synchronized among astrocytes in the field of view. Ca2+ oscillations occurred minutes after the Ca2+ wave of SD. Furthermore, fewer astrocytes were involved in Ca2+ oscillations at a given time, compared to Ca2+ waves, engaging all astrocytes in the field of view simultaneously. Finally, our data confirm that astrocyte Ca2+ waves coincide with arteriolar constriction, while post-SD Ca2+ oscillations occur with the peak of the SD-related vasodilation. This is the first in vivo study to present the post-SD astrocyte Ca2+ oscillations. Our results provide novel insight into the spatio-temporal correlation between glial reactivity and cerebral arteriole diameter changes behind the SD wavefront.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio , Calcio/metabolismo , Depresión de Propagación Cortical , Oscilometría , Animales , Arteriolas/metabolismo , Astrocitos/citología , Circulación Cerebrovascular , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía , Neuronas , Corteza Somatosensorial/metabolismo , Vasodilatación
3.
Int J Mol Sci ; 22(11)2021 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-34204159

RESUMEN

By upregulation of cell adhesion molecules and secretion of proinflammatory cytokines, cells of the neurovascular unit, including pericytes and endothelial cells, actively participate in neuroinflammatory reactions. As previously shown, both cell types can activate inflammasomes, cerebral endothelial cells (CECs) through the canonical pathway, while pericytes only through the noncanonical pathway. Using complex in vitro models, we demonstrate here that the noncanonical inflammasome pathway can be induced in CECs as well, leading to a further increase in the secretion of active interleukin-1ß over that observed in response to activation of the canonical pathway. In parallel, a more pronounced disruption of tight junctions takes place. We also show that CECs respond to inflammatory stimuli coming from both the apical/blood and the basolateral/brain directions. As a result, CECs can detect factors secreted by pericytes in which the noncanonical inflammasome pathway is activated and respond with inflammatory activation and impairment of the barrier properties. In addition, upon sensing inflammatory signals, CECs release inflammatory factors toward both the blood and the brain sides. Consequently, CECs activate pericytes by upregulating their expression of NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3), an inflammasome-forming pattern recognition receptor. In conclusion, cerebral pericytes and endothelial cells mutually activate each other in inflammation.


Asunto(s)
Encéfalo/patología , Comunicación Celular , Células Endoteliales/patología , Inflamasomas/metabolismo , Pericitos/patología , Transducción de Señal , Animales , Inflamación/metabolismo , Inflamación/patología , Porcinos , Uniones Estrechas/metabolismo
4.
Int J Mol Sci ; 20(21)2019 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-31684130

RESUMEN

With age, our cognitive skills and abilities decline. Maybe starting as an annoyance, this decline can become a major impediment to normal daily life. Recent research shows that the neurodegenerative disorders responsible for age associated cognitive dysfunction are mechanistically linked to the state of the microvasculature in the brain. When the microvasculature does not function properly, ischemia, hypoxia, oxidative stress and related pathologic processes ensue, further damaging vascular and neural function. One of the most important and specialized functions of the brain microvasculature is the blood-brain barrier (BBB), which controls the movement of molecules between blood circulation and the brain parenchyma. In this review, we are focusing on tight junctions (TJs), the multiprotein complexes that play an important role in establishing and maintaining barrier function. After a short introduction of the cell types that modulate barrier function via intercellular communication, we examine how age, age related pathologies and the aging of the immune system affects TJs. Then, we review how the TJs are affected in age associated neurodegenerative disorders: Alzheimer's disease and Parkinson's disease. Lastly, we summarize the TJ aspects of Huntington's disease and schizophrenia. Barrier dysfunction appears to be a common denominator in neurological disorders, warranting detailed research into the molecular mechanisms behind it. Learning the commonalities and differences in the pathomechanism of the BBB injury in different neurological disorders will predictably lead to development of new therapeutics that improve our life as we age.


Asunto(s)
Envejecimiento , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Microvasos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Uniones Estrechas/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/irrigación sanguínea , Humanos , Enfermedad de Parkinson/metabolismo
5.
Neurobiol Dis ; 119: 41-52, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30053571

RESUMEN

Recurrent spreading depolarizations occur in the cerebral cortex from minutes up to weeks following acute brain injury. Clinical evidence suggests that the immediate reduction of cerebral blood flow in response to spreading depolarization importantly contributes to lesion progression as the wave propagates over vulnerable tissue zones, characterized by potassium concentration already elevated prior to the passage of spreading depolarization. Here we demonstrate with two-photon microscopy in anesthetized mice that initial vasoconstriction in response to SD triggered experimentally with 1 M KCl is coincident in space and time with the large extracellular accumulation of potassium, as shown with a potassium indicator fluorescent dye. Moreover, pharmacological manipulations in combination with the use of potassium-sensitive microelectrodes suggest that large-conductance Ca2+-activated potassium (BK) channels and L-type voltage-gated calcium channels play significant roles in the marked initial vasoconstriction under elevated baseline potassium. We propose that potassium efflux through BK channels is a central component in the devastating neurovascular effects of spreading depolarizations in tissue at risk.


Asunto(s)
Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/fisiología , Circulación Cerebrovascular/fisiología , Depresión de Propagación Cortical/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Animales , Corteza Cerebral/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Depresión de Propagación Cortical/efectos de los fármacos , Indoles/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley
6.
Am J Physiol Heart Circ Physiol ; 313(5): H1000-H1012, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28801521

RESUMEN

Aging is associated with chronic inflammation partly mediated by increased levels of damage-associated molecular patterns, which activate pattern recognition receptors (PRRs) of the innate immune system. Furthermore, many aging-related disorders are associated with inflammation. PRRs, such as Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain-like receptors (NLRs), are expressed not only in cells of the innate immune system but also in other cells, including cells of the neurovascular unit and cerebral vasculature forming the blood-brain barrier. In this review, we summarize our present knowledge about the relationship between activation of PRRs expressed by cells of the neurovascular unit-blood-brain barrier, chronic inflammation, and aging-related pathologies of the brain. The most important damage-associated molecular pattern-sensing PRRs in the brain are TLR2, TLR4, and NLR family pyrin domain-containing protein-1 and pyrin domain-containing protein-3, which are activated during physiological and pathological aging in microglia, neurons, astrocytes, and possibly endothelial cells and pericytes.


Asunto(s)
Envejecimiento/metabolismo , Barrera Hematoencefálica/metabolismo , Inflamasomas/metabolismo , Inflamación/metabolismo , Microvasos/metabolismo , Acoplamiento Neurovascular , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Factores de Edad , Envejecimiento/inmunología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/fisiopatología , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Inflamación/inmunología , Inflamación/fisiopatología , Microvasos/inmunología , Microvasos/fisiopatología , Proteínas NLR/inmunología , Proteínas NLR/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
7.
Brain Behav Immun ; 64: 220-231, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28432035

RESUMEN

Cerebral pericytes are mural cells embedded in the basement membrane of capillaries. Increasing evidence suggests that they play important role in controlling neurovascular functions, i.e. cerebral blood flow, angiogenesis and permeability of the blood-brain barrier. These cells can also influence neuroinflammation which is highly regulated by the innate immune system. Therefore, we systematically tested the pattern recognition receptor expression of brain pericytes. We detected expression of NOD1, NOD2, NLRC5, NLRP1-3, NLRP5, NLRP9, NLRP10 and NLRX mRNA in non-treated cells. Among the ten known human TLRs, TLR2, TLR4, TLR5, TLR6 and TLR10 were found to be expressed. Inflammatory mediators induced the expression of NLRA, NLRC4 and TLR9 and increased the levels of NOD2, TLR2, inflammasome-forming caspases and inflammasome-cleaved interleukins. Oxidative stress, on the other hand, upregulated expression of TLR10 and NLRP9. Activation of selected pattern recognition receptors can lead to inflammasome assembly and caspase-dependent secretion of IL-1ß. TNF-α and IFN-γ increased the levels of pro-IL-1ß and pro-caspase-1 proteins; however, no canonical activation of NLRP1, NLRP2, NLRP3 or NLRC4 inflammasomes could be observed in human brain vascular pericytes. On the other hand, we could demonstrate secretion of active IL-1ß in response to non-canonical inflammasome activation, i.e. intracellular LPS or infection with E. coli bacteria. Our in vitro results indicate that pericytes might have an important regulatory role in neuroinflammation.


Asunto(s)
Encéfalo/metabolismo , Inflamasomas/metabolismo , Pericitos/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Interleucina-1beta/metabolismo , Transducción de Señal
8.
Am J Physiol Heart Circ Physiol ; 310(11): H1702-14, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27059078

RESUMEN

The blood-brain barrier (BBB) is the main interface controlling molecular and cellular traffic between the central nervous system (CNS) and the periphery. It consists of cerebral endothelial cells (CECs) interconnected by continuous tight junctions, and closely associated pericytes and astrocytes. Different parts of the CNS have diverse functions and structures and may be subject of different pathologies, in which the BBB is actively involved. It is largely unknown, however, what are the cellular and molecular differences of the BBB in different regions of the brain. Using in silico, in vitro, and ex vivo techniques we compared the expression of BBB-associated genes and proteins (i.e., markers of CECs, brain pericytes, and astrocytes) in the cortical grey matter and white matter. In silico human database analysis (obtained from recalculated data of the Allen Brain Atlas), qPCR, Western blot, and immunofluorescence studies on porcine and mouse brain tissue indicated an increased expression of glial fibrillary acidic protein in astrocytes in the white matter compared with the grey matter. We have also found increased expression of genes of the junctional complex of CECs (occludin, claudin-5, and α-catenin) in the white matter compared with the cerebral cortex. Accordingly, occludin, claudin-5, and α-catenin proteins showed increased expression in CECs of the white matter compared with endothelial cells of the cortical grey matter. In parallel, barrier properties of white matter CECs were superior as well. These differences might be important in the pathogenesis of diseases differently affecting distinct regions of the brain.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Moléculas de Adhesión Celular/metabolismo , Corteza Cerebral/metabolismo , Sustancia Gris/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Sustancia Blanca/metabolismo , Animales , Astrocitos/metabolismo , Simulación por Computador , Femenino , Humanos , Masculino , Ratones , Estructura Molecular , Pericitos/metabolismo , Porcinos , Uniones Estrechas/metabolismo
9.
Am J Pathol ; 185(8): 2206-18, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26216285

RESUMEN

The intestinal epithelium is a dynamic barrier that maintains the distinct environments of intestinal tissue and lumen. Epithelial barrier function is defined principally by tight junctions, which, in turn, depend on the regulated expression of claudin family proteins. Claudins are expressed differentially during intestinal epithelial cell (IEC) differentiation. However, regulatory mechanisms governing claudin expression during epithelial differentiation are incompletely understood. We investigated the molecular mechanisms regulating claudin-7 during IEC differentiation. Claudin-7 expression is increased as epithelial cells differentiate along the intestinal crypt-luminal axis. By using model IECs we observed increased claudin-7 mRNA and nascent heteronuclear RNA levels during differentiation. A screen for potential regulators of the CLDN7 gene during IEC differentiation was performed using a transcription factor/DNA binding array, CLDN7 luciferase reporters, and in silico promoter analysis. We identified hepatocyte nuclear factor 4α as a regulatory factor that bound endogenous CLDN7 promoter in differentiating IECs and stimulated CLDN7 promoter activity. These findings support a role of hepatocyte nuclear factor 4α in controlling claudin-7 expression during IEC differentiation.


Asunto(s)
Diferenciación Celular/genética , Claudinas/metabolismo , Células Epiteliales/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Mucosa Intestinal/metabolismo , Células CACO-2 , Claudinas/genética , Células Epiteliales/citología , Regulación de la Expresión Génica , Células HT29 , Factor Nuclear 4 del Hepatocito/genética , Humanos , Mucosa Intestinal/citología , Regiones Promotoras Genéticas
10.
EMBO Rep ; 12(4): 314-20, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21372850

RESUMEN

Expression of the tight junction protein junctional adhesion molecule-A (JAM-A) has been linked to proliferation and tumour progression. However, a direct role for JAM-A in regulating proliferative processes has not been shown. By using complementary in vivo and in vitro approaches, we demonstrate that JAM-A restricts intestinal epithelial cell (IEC) proliferation in a dimerization-dependent manner, by inhibiting Akt-dependent ß-catenin activation. Furthermore, IECs from transgenic JAM-A(-/-)/ß-catenin/T-cell factor reporter mice showed enhanced ß-catenin-dependent transcription. Finally, inhibition of Akt reversed colonic crypt hyperproliferation in JAM-A-deficient mice. These data establish a new link between JAM-A and IEC homeostasis.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , beta Catenina/metabolismo , Animales , Moléculas de Adhesión Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Humanos , Immunoblotting , Ratones , Ratones Mutantes , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Ribonucleósidos/farmacología , Transducción de Señal/genética , Uniones Estrechas/genética , Uniones Estrechas/metabolismo , beta Catenina/genética
11.
Pharmaceutics ; 14(7)2022 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-35890313

RESUMEN

The failures of anti-ß-amyloid immunotherapies suggested that the very low fraction of injected antibodies reaching the brain parenchyma due to the filtering effect of the BBB may be a reason for the lack of therapeutic effect. However, there is no treatment, as yet, for the amyotrophic lateral sclerosis (ALS) despite substantial evidence existing of the involvement of TDP-43 protein in the evolution of ALS. To circumvent this filtering effect, we have developed a novel approach to facilitate the penetration of antibody fragments (Fabs) into the brain parenchyma. Leveraging the homing properties of endothelial progenitor cells (EPCs), we transfected, ex vivo, such cells with vectors encoding anti-ß-amyloid and anti-TDP43 Fabs turning them into an "antibody fragment factory". When injected these cells integrate into the BBB, where they secrete anti-TDP43 Fabs. The results showed the formation of tight junctions between the injected engineered EPCs and the unlabeled resident endothelial cells. When the EPCs were further modified to express the anti-TDP43 Fab, we could observe integration of these cells into the vasculature and the secretion of Fabs. Results confirm that production and secretion of Fabs at the BBB level leads to their migration to the brain parenchyma where they might exert a therapeutic effect.

12.
J Cereb Blood Flow Metab ; 42(4): 584-599, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34427145

RESUMEN

Spreading depolarizations (SDs) indicate injury progression and predict worse clinical outcome in acute brain injury. We demonstrate in rodents that acute brain swelling upon cerebral ischemia impairs astroglial glutamate clearance and increases the tissue area invaded by SD. The cytotoxic extracellular glutamate accumulation (>15 µM) predisposes an extensive bulk of tissue (4-5 mm2) for a yet undescribed simultaneous depolarization (SiD). We confirm in rat brain slices exposed to osmotic stress that SiD is the pathological expansion of prior punctual SD foci (0.5-1 mm2), is associated with astrocyte swelling, and triggers oncotic neuron death. The blockade of astrocytic aquaporin-4 channels and Na+/K+/Cl- co-transporters, or volume-regulated anion channels mitigated slice edema, extracellular glutamate accumulation (<10 µM) and SiD occurrence. Reversal of slice swelling by hyperosmotic mannitol counteracted glutamate accumulation and prevented SiD. In contrast, inhibition of glial metabolism or inhibition of astrocyte glutamate transporters reproduced the SiD phenotype. Finally, we show in the rodent water intoxication model of cytotoxic edema that astrocyte swelling and altered astrocyte calcium waves are central in the evolution of SiD. We discuss our results in the light of evidence for SiD in the human cortex. Our results emphasize the need of preventive osmotherapy in acute brain injury.


Asunto(s)
Edema Encefálico , Lesiones Encefálicas , Animales , Ratas , Astrocitos/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/metabolismo , Edema/metabolismo , Ácido Glutámico/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo
13.
Neuropharmacology ; 192: 108612, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34023338

RESUMEN

Dimethyltryptamine (DMT), an endogenous ligand of sigma-1 receptors (Sig-1Rs), acts against systemic hypoxia, but whether DMT may prevent cerebral ischemic injury is unexplored. Here global forebrain ischemia was created in anesthetized rats and aggravated with the induction of spreading depolarizations (SDs) and subsequent short hypoxia before reperfusion. Drugs (DMT, the selective Sig-1R agonist PRE-084, the Sig-1R antagonist NE-100, or the serotonin receptor antagonist asenapine) were administered intravenously alone or in combination while physiological variables and local field potential from the cerebral cortex was recorded. Neuroprotection and the cellular localization of Sig-1R were evaluated with immunocytochemistry. Plasma and brain DMT content was measured by 2D-LC-HRMS/MS. The affinity of drugs for cerebral Sig-1R was evaluated with a radioligand binding assay. Both DMT and PRE-084 mitigated SDs, counteracted with NE-100. Further, DMT attenuated SD when co-administered with asenapine, compared to asenapine alone. DMT reduced the number of apoptotic and ferroptotic cells and supported astrocyte survival. The binding affinity of DMT to Sig-1R matched previously reported values. Sig-1Rs were associated with the perinuclear cytoplasm of neurons, astrocytes and microglia, and with glial processes. According to these data, DMT may be considered as adjuvant pharmacological therapy in the management of acute cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Depresión de Propagación Cortical/efectos de los fármacos , N,N-Dimetiltriptamina/farmacología , Enfermedades Neurodegenerativas/metabolismo , Receptores sigma/metabolismo , Animales , Encéfalo/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Depresión de Propagación Cortical/fisiología , Relación Dosis-Respuesta a Droga , Masculino , N,N-Dimetiltriptamina/uso terapéutico , Enfermedades Neurodegenerativas/prevención & control , Ratas , Ratas Sprague-Dawley , Receptores sigma/agonistas , Receptor Sigma-1
14.
Mol Oncol ; 14(9): 2040-2057, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32534480

RESUMEN

Brain metastases are life-threatening complications of triple-negative breast cancer, melanoma, and a few other tumor types. Poor outcome of cerebral secondary tumors largely depends on the microenvironment formed by cells of the neurovascular unit, among which pericytes are the least characterized. By using in vivo and in vitro techniques and human samples, here we show that pericytes play crucial role in the development of metastatic brain tumors by directly influencing key steps of the development of the disease. Brain pericytes had a prompt chemoattractant effect on breast cancer cells and established direct contacts with them. By secreting high amounts of extracellular matrix proteins, pericytes enhanced adhesion of both melanoma and triple-negative cancer cells, which might be particularly important in the exclusive perivascular growth of these tumor cells. In addition, pericytes secreted insulin-like growth factor 2 (IGF2), which had a very significant pro-proliferative effect on mammary carcinoma, but not on melanoma cells. By inhibiting IGF2 signaling using silencing or picropodophyllin (PPP), we could block the proliferation-increasing effect of pericytes on breast cancer cells. Administration of PPP (a blood-brain barrier-permeable substance) significantly decreased the size of brain tumors in mice inoculated with triple-negative breast cancer cells. Taken together, our results indicate that brain pericytes have significant pro-metastatic features, especially in breast cancer. Our study underlines the importance of targeting pericytes and the IGF axis as potential strategies in brain metastatic diseases.


Asunto(s)
Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Pericitos/metabolismo , Animales , Encéfalo/patología , Adhesión Celular , Comunicación Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Ratones , Pericitos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
Acta Neuropathol Commun ; 7(1): 133, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31426859

RESUMEN

Therapeutic resistance of cerebral secondary tumours largely depends on unique aspects linked to the neurovascular unit, especially cerebral endothelial cells and astrocytes. By using advanced microscopy techniques, here we explored novel mechanisms related to the neurovascular unit during extravasation and proliferation of triple negative breast cancer cells in the brain. Metastatic mammary carcinoma cells arrested and elongated within one hour in cerebral microvessels, but their number decreased by almost 80% in the first two days. Interestingly, malignant cells induced vasoconstriction and development of intraluminal endothelial plugs, which isolated invading cells from the circulation. During diapedesis - which usually took place on day four and five after inoculation of the tumour cells - continuity of cerebral endothelial tight junctions remained intact, indicating migration of cancer cells through the transcellular pathway. In addition, metastatic cells induced formation of multiluminal vessels and claudin-5-positive endothelial blebs. However, even severe endothelial blebbing could be reversed and the vessel morphology was restored shortly after the tumour cells completed transendothelial migration. Similar to neuro-inflammatory leukocytes, tumour cells migrated not only through the endothelial layer, but through the glia limitans perivascularis as well. Nevertheless, along with the growth of metastatic lesions by co-option of pre-existing capillaries, astrocytes and astrocyte end-feet were gradually expelled from the vessels to the border of the tumour. Taken together, we identified previously unknown mechanisms involved in the reaction of brain resident cells to invading breast cancer cells. Our results contribute to a better understanding of the complex cross-talk between tumour cells and host cells in the brain, which is essential for the identification of new therapeutic targets in this devastating disease.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/patología , Neoplasias de la Mama/patología , Movimiento Celular/fisiología , Células Endoteliales/patología , Animales , Encéfalo/diagnóstico por imagen , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Neoplasias de la Mama/diagnóstico por imagen , Células Cultivadas , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/patología , Técnicas de Cocultivo , Femenino , Ratones , Ratones Endogámicos BALB C
16.
J Neurochem ; 107(1): 116-26, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18673450

RESUMEN

Because of the relative impermeability of the blood-brain barrier (BBB), many drugs are unable to reach the CNS in therapeutically relevant concentration. One method to deliver drugs to the CNS is the osmotic opening of the BBB using mannitol. Hyperosmotic mannitol induces a strong phosphorylation on tyrosine residues in a broad spectrum of proteins in cerebral endothelial cells, the principal components of the BBB. Previously, we have shown that among targets of tyrosine phosphorylation are beta-catenin, extracellular signal-regulated kinase 1/2 and the non-receptor tyrosine kinase Src. The aim of this study was to identify new signalling pathways activated by hypertonicity in cerebral endothelial cells. Using an antibody array and immunoprecipitation we identified the receptor tyrosine kinase Axl to become tyrosine phosphorylated in response to hyperosmotic mannitol. Besides activation, Axl was also cleaved in response to osmotic stress. Degradation of Axl proved to be metalloproteinase- and proteasome-dependent and resulted in 50-55 kDa C-terminal products which remained phosphorylated even after degradation. Specific knockdown of Axl increased the rate of apoptosis in hyperosmotic mannitol-treated cells; therefore, we assume that activation of Axl may be a protective mechanism against hypertonicity-induced apoptosis. Our results identify Axl as an important element of osmotic stress-induced signalling.


Asunto(s)
Barrera Hematoencefálica/enzimología , Arterias Cerebrales/enzimología , Células Endoteliales/enzimología , Proteínas Oncogénicas/metabolismo , Presión Osmótica/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular , Arterias Cerebrales/citología , Arterias Cerebrales/efectos de los fármacos , Regulación hacia Abajo/fisiología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Soluciones Hipertónicas/farmacología , Manitol/farmacología , Metaloproteasas/metabolismo , Proteínas Oncogénicas/genética , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tirosina Quinasa del Receptor Axl
17.
Neurochem Int ; 50(1): 219-28, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16997427

RESUMEN

Peripheral inflammation can aggravate local brain inflammation and neuronal death. The blood-brain barrier (BBB) is a key player in the event. On a relevant in vitro model of primary rat brain endothelial cells co-cultured with primary rat astroglia cells lipopolysaccharide (LPS)-induced changes in several BBB functions have been investigated. LPS-treatment resulted in a dose- and time-dependent decrease in the integrity of endothelial monolayers: transendothelial electrical resistance dropped, while flux of permeability markers fluorescein and albumin significantly increased. Immunostaining for junctional proteins ZO-1, claudin-5 and beta-catenin was significantly weaker in LPS-treated endothelial cells than in control monolayers. LPS also reduced the intensity and changed the pattern of ZO-1 immunostaining in freshly isolated rat brain microvessels. The activity of P-glycoprotein, an important efflux pump at the BBB, was also inhibited by LPS. At the same time production of reactive oxygen species and nitric oxide was increased in brain endothelial cells treated with LPS. Pentosan polysulfate, a polyanionic polysaccharide could reduce the deleterious effects of LPS on BBB permeability, and P-glycoprotein activity. LPS-stimulated increase in the production of reactive oxygen species and nitric oxide was also decreased by pentosan treatment. The protective effect of pentosan for brain endothelium can be of therapeutical significance in bacterial infections affecting the BBB.


Asunto(s)
Encéfalo/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Lipopolisacáridos/farmacología , Poliéster Pentosan Sulfúrico/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Barrera Hematoencefálica , Encéfalo/irrigación sanguínea , Endotelio Vascular/citología , Potenciales de la Membrana/efectos de los fármacos , Poliéster Pentosan Sulfúrico/farmacocinética , Ratas
18.
Phys Med Biol ; 52(20): 6261-74, 2007 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-17921584

RESUMEN

Cerebral endothelial cells interconnected by tight and adherens junctions constitute the structural basis of the blood-brain barrier. Extracellular calcium ions have been reported to play an important role in the formation and maintenance of the junctional complex. However, little is known about the action of calcium depletion on the structural characteristics of cerebral endothelial cells. Using atomic force microscopy we analyzed the effect of calcium depletion and readdition on the shape and size of living brain endothelial cells. It was found that the removal of extracellular calcium from confluent cell cultures induced the dissociation of the cells from each other accompanied by an increase in their height. After readdition of calcium a gradual recovery was observed until total confluency was regained. We have also demonstrated that Rho-kinase plays an important role in the calcium-depletion-induced disassembly of endothelial tight and adherens junctions. The Rho-kinase inhibitor Y27632 could prevent the morphological changes induced by a lack of calcium as well. Our results suggest that calcium depletion induces Rho-kinase-dependent cytoskeletal changes that may be partly responsible for the disassembly of the junctional complex.


Asunto(s)
Barrera Hematoencefálica/citología , Barrera Hematoencefálica/fisiología , Encéfalo/citología , Encéfalo/fisiología , Calcio/metabolismo , Células Endoteliales/citología , Células Endoteliales/fisiología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Calcio/administración & dosificación , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Líquido Extracelular/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Ratas , Quinasas Asociadas a rho/metabolismo
19.
Nat Cell Biol ; 19(11): 1326-1335, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29058719

RESUMEN

In vitro differentiation of human intestinal organoids (HIOs) from pluripotent stem cells is an unparalleled system for creating complex, multicellular three-dimensional structures capable of giving rise to tissue analogous to native human tissue. Current methods for generating HIOs rely on growth in an undefined tumour-derived extracellular matrix (ECM), which severely limits the use of organoid technologies for regenerative and translational medicine. Here, we developed a fully defined, synthetic hydrogel based on a four-armed, maleimide-terminated poly(ethylene glycol) macromer that supports robust and highly reproducible in vitro growth and expansion of HIOs, such that three-dimensional structures are never embedded in tumour-derived ECM. We also demonstrate that the hydrogel serves as an injection vehicle that can be delivered into injured intestinal mucosa resulting in HIO engraftment and improved colonic wound repair. Together, these studies show proof-of-concept that HIOs may be used therapeutically to treat intestinal injury.


Asunto(s)
Colon/efectos de los fármacos , Hidrogeles/farmacología , Intestinos/efectos de los fármacos , Organoides/efectos de los fármacos , Regeneración/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Ratones , Células Madre Pluripotentes/efectos de los fármacos
20.
Curr Pharm Des ; 22(35): 5400-5414, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27464718

RESUMEN

The intestinal epithelium serves as an important barrier between luminal contents and underlying tissue compartments. Barrier properties are established and maintained by the multiprotein complexes in the tight junction (TJ), adherens junction (AJ) and desmosomes (DM). Compromised intestinal epithelial barrier function resulting from changes in junctional protein expression and organization is associated with exposure to luminal antigens that result in mucosal inflammation, which in turn further exacerbates the epithelial barrier compromise. In this review we discuss regulatory mechanisms by which intercellular junction proteins are influenced by inflammatory mediators to influence intestinal epithelial barrier function and address potential therapeutic strategies to strengthen the barrier and restore mucosal homeostasis.


Asunto(s)
Sistemas de Liberación de Medicamentos , Mediadores de Inflamación/farmacología , Mucosa Intestinal/efectos de los fármacos , Uniones Estrechas/efectos de los fármacos , Humanos , Mediadores de Inflamación/química , Mucosa Intestinal/metabolismo , Uniones Estrechas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA