Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Immunity ; 54(11): 2439-2441, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758333

RESUMEN

In this issue of Immunity, Vega-Pérez et al. (2021) reveal the formation of a dynamic multicellular aggregate within a fibrin scaffold consisting of large peritoneal macrophages, B1 cells, neutrophils, and monocytes during antibacterial immunity in the peritoneum. Anticoagulants targeting thrombin or peritoneal macrophage depletion by clodronate impaired efficient control of E. coli infection.


Asunto(s)
Escherichia coli , Fibrina , Animales , Coagulación Sanguínea , Ratones , Ratones Endogámicos C57BL , Monocitos
2.
J Neurosci ; 39(47): 9343-9359, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31591156

RESUMEN

Subcortical white matter stroke is a common stroke subtype. White matter stroke stimulates adjacent oligodendrocyte progenitor cells (OPCs) to divide and migrate to the lesion, but stroke OPCs have only a limited differentiation into mature oligodendrocytes. To understand the molecular systems that are active in OPC responses in white matter stroke, OPCs were virally labeled and laser-captured in the region of partial damage adjacent to the infarct in male mice. RNAseq indicates two distinct OPC transcriptomes associated with the proliferative and limited-regeneration phases of OPCs after stroke. Molecular pathways related to nuclear receptor activation, ECM turnover, and lipid biosynthesis are activated during proliferative OPC phases after stroke; inflammatory and growth factor signaling is activated in the later stage of limited OPC differentiation. Within ECM proteins, Matrilin-2 is induced early after stroke and then rapidly downregulated. Prediction of upstream regulators of the OPC stroke transcriptome identifies several candidate molecules, including Inhibin A-a negative regulator of Matrilin-2. Inhibin A is induced in reactive astrocytes after stroke, including in humans. In functional assays, Matrilin-2 induces OPC differentiation, and Inhibin A inhibits OPC Matrilin-2 expression and inhibits OPC differentiation. In vivo, Matrilin-2 promotes motor recovery after white matter stroke, and promotes OPC differentiation and ultrastructural evidence of remyelination. These studies show that white matter stroke induces an initial proliferative and reparative response in OPCs, but this is blocked by a local cellular niche where reactive astrocytes secrete Inhibin A, downregulating Matrilin-2 and blocking myelin repair and recovery.SIGNIFICANCE STATEMENT Stroke in the cerebral white matter of the brain is common. The biology of damage and recovery in this stroke subtype are not well defined. These studies use cell-specific RNA sequencing and gain-of-function studies to show that white matter stroke induces a glial signaling niche, present in both humans and mice, between reactive astrocytes and oligodendrocyte progenitor cells. Astrocyte secretion of Inhibin A and downregulation of oligodendrocyte precursor production of Matrilin-2 limit OPC differentiation, tissue repair, and recovery in this disease.


Asunto(s)
Astrocitos/patología , Oligodendroglía/patología , Recuperación de la Función , Accidente Cerebrovascular/patología , Sustancia Blanca/patología , Animales , Astrocitos/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Oligodendroglía/fisiología , Ratas , Recuperación de la Función/fisiología , Accidente Cerebrovascular/genética , Sustancia Blanca/fisiología
3.
Proc Natl Acad Sci U S A ; 113(52): E8453-E8462, 2016 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-27956620

RESUMEN

White matter stroke is a distinct stroke subtype, accounting for up to 25% of stroke and constituting the second leading cause of dementia. The biology of possible tissue repair after white matter stroke has not been determined. In a mouse stroke model, white matter ischemia causes focal damage and adjacent areas of axonal myelin disruption and gliosis. In these areas of only partial damage, local white matter progenitors respond to injury, as oligodendrocyte progenitors (OPCs) proliferate. However, OPCs fail to mature into oligodendrocytes (OLs) even in regions of demyelination with intact axons and instead divert into an astrocytic fate. Local axonal sprouting occurs, producing an increase in unmyelinated fibers in the corpus callosum. The OPC maturation block after white matter stroke is in part mediated via Nogo receptor 1 (NgR1) signaling. In both aged and young adult mice, stroke induces NgR1 ligands and down-regulates NgR1 inhibitors during the peak OPC maturation block. Nogo ligands are also induced adjacent to human white matter stroke in humans. A Nogo signaling blockade with an NgR1 antagonist administered after stroke reduces the OPC astrocytic transformation and improves poststroke oligodendrogenesis in mice. Notably, increased white matter repair in aged mice is translated into significant poststroke motor recovery, even when NgR1 blockade is provided during the chronic time points of injury. These data provide a perspective on the role of NgR1 ligand function in OPC fate in the context of a specific and common type of stroke and show that it is amenable to systemic intervention to promote recovery.


Asunto(s)
Envejecimiento , Vaina de Mielina/química , Receptor Nogo 1/metabolismo , Accidente Cerebrovascular/fisiopatología , Sustancia Blanca/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Axones/metabolismo , Encéfalo/patología , Diferenciación Celular , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Humanos , Ligandos , Ratones , Ratones Transgénicos , Oligodendroglía/citología , Remielinización , Células Madre/citología , Rehabilitación de Accidente Cerebrovascular , Sustancia Blanca/patología
4.
Methods Mol Biol ; 2561: 87-101, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36399266

RESUMEN

Cerebrovascular dysfunction is a hallmark of Alzheimer's disease (AD) that is linked to cognitive decline. However, blood-brain barrier (BBB) disruption in AD is focal and requires sensitive methods to detect extravasated blood proteins and vasculature in large brain volumes. Fibrinogen, a blood coagulation factor, is deposited in AD brains at sites of BBB disruption and cerebrovascular damage. This chapter presents the methodology of fibrinogen immunolabeling-enabled three-dimensional (3D) imaging of solvent-cleared organs (iDISCO) which, when combined with immunolabeling of amyloid ß (Aß) and vasculature, enables sensitive detection of focal BBB vascular abnormalities, and reveals the spatial distribution of Aß plaques and fibrin deposits, in large tissue volumes from cleared human brains. Overall, fibrinogen iDISCO enables the investigation of neurovascular and neuroimmune mechanisms driving neurodegeneration in disease.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Fibrinógeno/metabolismo , Imagenología Tridimensional , Placa Amiloide
5.
Trends Pharmacol Sci ; 42(9): 772-788, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34334250

RESUMEN

The p75 neurotrophin receptor (p75NTR) functions at the molecular nexus of cell death, survival, and differentiation. In addition to its contribution to neurodegenerative diseases and nervous system injuries, recent studies have revealed unanticipated roles of p75NTR in liver repair, fibrinolysis, lung fibrosis, muscle regeneration, and metabolism. Linking these various p75NTR functions more precisely to specific mechanisms marks p75NTR as an emerging candidate for therapeutic intervention in a wide range of disorders. Indeed, small molecule inhibitors of p75NTR binding to neurotrophins have shown efficacy in models of Alzheimer's disease (AD) and neurodegeneration. Here, we outline recent advances in understanding p75NTR pleiotropic functions in vivo, and propose an integrated view of p75NTR and its challenges and opportunities as a pharmacological target.


Asunto(s)
Enfermedad de Alzheimer , Receptor de Factor de Crecimiento Nervioso , Enfermedad de Alzheimer/tratamiento farmacológico , Muerte Celular , Humanos
6.
bioRxiv ; 2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34671772

RESUMEN

Blood clots are a central feature of coronavirus disease-2019 (COVID-19) and can culminate in pulmonary embolism, stroke, and sudden death. However, it is not known how abnormal blood clots form in COVID-19 or why they occur even in asymptomatic and convalescent patients. Here we report that the Spike protein from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds to the blood coagulation factor fibrinogen and induces structurally abnormal blood clots with heightened proinflammatory activity. SARS-CoV-2 Spike virions enhanced fibrin-mediated microglia activation and induced fibrinogen-dependent lung pathology. COVID-19 patients had fibrin autoantibodies that persisted long after acute infection. Monoclonal antibody 5B8, targeting the cryptic inflammatory fibrin epitope, inhibited thromboinflammation. Our results reveal a procoagulant role for the SARS-CoV-2 Spike and propose fibrin-targeting interventions as a treatment for thromboinflammation in COVID-19. ONE-SENTENCE SUMMARY: SARS-CoV-2 spike induces structurally abnormal blood clots and thromboinflammation neutralized by a fibrin-targeting antibody.

7.
Nat Neurosci ; 24(1): 19-23, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33318667

RESUMEN

Microglial surveillance is a key feature of brain physiology and disease. Here, we found that Gi-dependent microglial dynamics prevent neuronal network hyperexcitability. By generating MgPTX mice to genetically inhibit Gi in microglia, we show that sustained reduction of microglia brain surveillance and directed process motility induced spontaneous seizures and increased hypersynchrony after physiologically evoked neuronal activity in awake adult mice. Thus, Gi-dependent microglia dynamics may prevent hyperexcitability in neurological diseases.


Asunto(s)
Quinasa 1 del Receptor Acoplado a Proteína-G/fisiología , Microglía/fisiología , Red Nerviosa/fisiología , Animales , Señalización del Calcio , Movimiento Celular , Convulsivantes , Electroencefalografía , Vigilancia Inmunológica , Ratones , Microglía/enzimología , Microglía/ultraestructura , Enfermedades del Sistema Nervioso/fisiopatología , Fenómenos Fisiológicos del Sistema Nervioso , Pilocarpina , Convulsiones/fisiopatología , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
8.
Cancer Res ; 66(20): 9837-44, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17047044

RESUMEN

The transforming growth factor-beta (TGF-beta) signaling pathway is a tumor-suppressor pathway that is commonly inactivated in colon cancer. TGF-beta is a secreted ligand that mediates its effects through a transmembrane heteromeric receptor complex, which consists of type I (TGFBR1) and type II subunits (TGFBR2). Approximately 30% of colon cancers carry TGFBR2 mutations, demonstrating that it is a common target for mutational inactivation in this cancer. To assess the functional role of TGFBR2 inactivation in the multistep progression sequence of colon cancer, we generated a mouse model that recapitulates two common genetic events observed in human colon cancer by mating Apc(1638N/wt) mice with mice that are null for Tgfbr2 in the intestinal epithelium, Villin-Cre;Tgfbr2(E2flx/E2flx) mice. In this model, we observed a dramatic increase in the number of intestinal adenocarcinomas in the Apc(1638N/wt);Villin-Cre;Tgfbr2(E2flx/E2flx) mice (called Apc(1638N/wt);Tgfbr2(IEKO)) compared with those mice with intact Tgfbr2 (Apc(1638N/wt);Tgfbr2(E2flx/E2flx)). Additionally, in vitro analyses of epithelial tumor cells derived from the Apc(1638N/wt);Tgfbr2(IEKO) mice showed enhanced expression and activity of matrix metalloproteinase MMP-2 and MMP-9, as well as increased TGF-beta1 secretion in the conditioned medium. Similarly, primary tumor tissues from the Apc(1638N/wt);Tgfbr2(IEKO) mice also showed elevated amounts of TGF-beta1 as well as higher MMP-2 activity in comparison with Apc(1638N/wt);Tgfbr2(E2flx/E2flx)-derived tumors. Thus, loss of TGFBR2 in intestinal epithelial cells promotes the invasion and malignant transformation of tumors initiated by Apc mutation, providing evidence that Wnt signaling deregulation and TGF-beta signaling inactivation cooperate to drive the initiation and progression, respectively, of intestinal cancers in vivo.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Genes APC , Animales , Procesos de Crecimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Transgénicos , Mutación , Invasividad Neoplásica , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
9.
Oncogene ; 24(18): 3028-41, 2005 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-15735717

RESUMEN

The transforming growth factor beta (TGF-beta) signaling pathway, which is activated by the TGF-beta receptor complex consisting of type I and type II TGF-beta receptors (TGFBR1 and TGFBR2), regulates cell growth and death. TGF-beta and components of its signaling pathway, particularly TGFBR2, have been implicated as tumor suppressor genes and important antimitogenic factors in the gastrointestinal tract and liver. An in vivo approach to study these effects has been hindered by the embryonic lethality of Tgfbr2(-/-) mice and poor viability of the Tgfb1(-/-) mice. Consequently, we have developed a hepatocyte-specific Tgfbr2 knockout mouse, the Alb-cre Tgfbr2(flx/flx) mouse, to study the physiologically relevant effects of TGF-beta signaling on epithelial cell proliferation in vivo. After 70% hepatectomy, we observed increased proliferation and an increased liver mass : body weight ratio in the Alb-cre Tgfbr2(flx/flx) mice compared to Tgfbr2(flx/flx) mice. We also observed decreased expression and increased phosphorylation of p130 in the livers from the Alb-cre Tgfbr2(flx/flx) mice as well as increased expression of cyclin E, which is transcriptionally regulated, in part, by p130:E2F4. Consistent with these results, in a hepatocyte cell line derived from the Tgfbr2(flx/flx) mice, we found that TGF-beta increases the nuclear localization of E2F4, and presumably the transcriptional repression of the p130:E2F4 complex. Thus, we have demonstrated that TGF-beta signaling in vivo regulates the mitogenic response in the regenerating liver, affecting the liver mass : body weight ratio after partial hepatectomy, and that these mitogenic responses are accompanied by alterations in p130 expression and phosphorylation, implicating p130 as one of the proteins regulated in vivo by TGF-beta during liver regeneration.


Asunto(s)
División Celular/fisiología , Hepatocitos/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Hepatectomía , Hepatocitos/citología , Regeneración Hepática/fisiología , Ratones , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Smad , Transactivadores/metabolismo
10.
J Vis Exp ; (109)2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-27023377

RESUMEN

Stroke affecting white matter accounts for up to 25% of clinical stroke presentations, occurs silently at rates that may be 5-10 fold greater, and contributes significantly to the development of vascular dementia. Few models of focal white matter stroke exist and this lack of appropriate models has hampered understanding of the neurobiologic mechanisms involved in injury response and repair after this type of stroke. The main limitation of other subcortical stroke models is that they do not focally restrict the infarct to the white matter or have primarily been validated in non-murine species. This limits the ability to apply the wide variety of murine research tools to study the neurobiology of white matter stroke. Here we present a methodology for the reliable production of a focal stroke in murine white matter using a local injection of an irreversible eNOS inhibitor. We also present several variations on the general protocol including two unique stereotactic variations, retrograde neuronal tracing, as well as fresh tissue labeling and dissection that greatly expand the potential applications of this technique. These variations allow for multiple approaches to analyze the neurobiologic effects of this common and understudied form of stroke.


Asunto(s)
Axones/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/toxicidad , Degeneración Nerviosa/patología , Ornitina/análogos & derivados , Accidente Cerebrovascular/patología , Sustancia Blanca/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/inducido químicamente , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ornitina/toxicidad , Accidente Cerebrovascular/inducido químicamente , Sustancia Blanca/patología
11.
Neurotherapeutics ; 9(2): 349-58, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22362423

RESUMEN

Stroke is a devastating neurological disease with limited functional recovery. Stroke affects all cellular elements of the brain and impacts areas traditionally classified as both gray matter and white matter. In fact, stroke in subcortical white matter regions of the brain accounts for approximately 30% of all stroke subtypes, and white matter injury is a component of most classes of stroke damage. However, most basic scientific information in stroke cell death and neural repair relates principally to neuronal cell death and repair. Despite an emerging biological understanding of white matter development, adult function, and reorganization in inflammatory diseases, such as multiple sclerosis, little is known of the specific molecular and cellular events in white matter ischemia. This limitation stems in part from the difficulty in generating animal models of white matter stroke. This review will discuss recent progress in studies of animal models of white matter stroke, and the emerging principles of cell death and repair in oligodendrocytes, axons, and astrocytes in white matter ischemic injury.


Asunto(s)
Modelos Animales de Enfermedad , Fibras Nerviosas Mielínicas/patología , Accidente Cerebrovascular/patología , Animales , Ensayos Clínicos como Asunto/métodos , Humanos , Vaina de Mielina/genética , Vaina de Mielina/patología , Especificidad de la Especie , Accidente Cerebrovascular/genética
12.
J Neurosci Methods ; 180(2): 261-72, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19439360

RESUMEN

Subcortical white matter stroke is a common stroke subtype but has had limited pre-clinical modeling. Recapitulating this disease process in mice has been impeded by the relative inaccessibility of the subcortical white matter arterial supply to induce white matter ischemia in isolation. In this report, we detail a subcortical white matter stroke model developed in the mouse and its characterization with a comprehensive set of MRI, immunohistochemical, neuronal tract tracing and electron microscopic studies. Focal injection of the vasoconstrictor endothelin-1 into the subcortical white matter produces an infarct core that develops a maximal MRI signal by day 2, which is comparable in relative size and location to human subcortical stroke. Immunohistochemical studies indicate that oligodendrocyte apoptosis is maximal at day 1 and apoptotic cells extend away from the stroke core into the peri-infarct white matter. The amount of myelin loss exceeds axonal fiber loss in this peri-infarct region. Activation of microglia/macrophages takes place at 1 day after injection near injured axons. Neuronal tract tracing demonstrates that subcortical white matter stroke disconnects a large region of bilateral sensorimotor cortex. There is a robust glial response after stroke by BrdU pulse-labeling, and oligodendrocyte precursor cells are initiated to proliferate and differentiate within the first week of injury. These results demonstrate the utility of the endothelin-1 mediated subcortical stroke in the mouse to study post-stroke repair mechanisms, as the infarct core extends through the partially damaged peri-infarct white matter and induces an early glial progenitor response.


Asunto(s)
Demencia Vascular/patología , Fibras Nerviosas Mielínicas/patología , Regeneración Nerviosa/fisiología , Células Madre/fisiología , Accidente Cerebrovascular/patología , Degeneración Walleriana/patología , Animales , Apoptosis/fisiología , Proliferación Celular , Demencia Vascular/inducido químicamente , Demencia Vascular/fisiopatología , Modelos Animales de Enfermedad , Endotelina-1/farmacología , Gliosis/etiología , Gliosis/patología , Gliosis/fisiopatología , Macrófagos/patología , Imagen por Resonancia Magnética/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/patología , Vías Nerviosas/patología , Vías Nerviosas/fisiopatología , Oligodendroglía/citología , Oligodendroglía/fisiología , Recuperación de la Función/fisiología , Accidente Cerebrovascular/inducido químicamente , Accidente Cerebrovascular/fisiopatología , Vasoconstrictores/farmacología , Degeneración Walleriana/etiología , Degeneración Walleriana/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA