Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
Cell Rep Med ; 5(6): 101606, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38866016

RESUMEN

Leptomeningeal disease (LMD) remains a rapidly lethal complication for late-stage melanoma patients. Here, we characterize the tumor microenvironment of LMD and patient-matched extra-cranial metastases using spatial transcriptomics in a small number of clinical specimens (nine tissues from two patients) with extensive in vitro and in vivo validation. The spatial landscape of melanoma LMD is characterized by a lack of immune infiltration and instead exhibits a higher level of stromal involvement. The tumor-stroma interactions at the leptomeninges activate tumor-promoting signaling, mediated through upregulation of SERPINA3. The meningeal stroma is required for melanoma cells to survive in the cerebrospinal fluid (CSF) and promotes MAPK inhibitor resistance. Knocking down SERPINA3 or inhibiting the downstream IGR1R/PI3K/AKT axis results in tumor cell death and re-sensitization to MAPK-targeting therapy. Our data provide a spatial atlas of melanoma LMD, identify the tumor-promoting role of meningeal stroma, and demonstrate a mechanism for overcoming microenvironment-mediated drug resistance in LMD.


Asunto(s)
Melanoma , Neoplasias Meníngeas , Células del Estroma , Microambiente Tumoral , Melanoma/genética , Melanoma/patología , Humanos , Microambiente Tumoral/genética , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Animales , Línea Celular Tumoral , Ratones , Regulación Neoplásica de la Expresión Génica , Transcriptoma/genética , Perfilación de la Expresión Génica , Meninges/patología , Meninges/metabolismo , Resistencia a Antineoplásicos/genética , Transducción de Señal , Femenino
2.
Adv Sci (Weinh) ; 11(26): e2402059, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38704728

RESUMEN

White matter hyperintensity (WMH) represents a critical global medical concern linked to cognitive decline and dementia, yet its underlying mechanisms remain poorly understood. Here, humans are directly demonstrated that high WMH burden correlates with delayed drainage of meningeal lymphatic vessels (mLVs) and glymphatic pathway. Additionally, a longitudinal cohort study reveals that glymphatic dysfunction predicts WMH progression. Next, in a rat model of WMH, the presence of impaired lymphangiogenesis and glymphatic drainage is confirmed, followed by elevated microglial activation and white matter demyelination. Notably, enhancing meningeal lymphangiogenesis through adeno-associated virus delivery of vascular endothelial growth factor-C (VEGF-C) mitigates microglial gliosis and white matter demyelination. Conversely, blocking the growth of mLVs with a VEGF-C trap strategy exacerbates these changes. The findings highlight the role of mLVs and glymphatic pathway dysfunction in aggravating brain white matter injury, providing a potential novel strategy for WMH prevention and treatment.


Asunto(s)
Sistema Glinfático , Meninges , Sustancia Blanca , Sistema Glinfático/metabolismo , Animales , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Humanos , Masculino , Ratas , Femenino , Meninges/metabolismo , Modelos Animales de Enfermedad , Vasos Linfáticos/metabolismo , Anciano , Imagen por Resonancia Magnética/métodos , Estudios Longitudinales
3.
Nature ; 628(8009): 863-871, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38570687

RESUMEN

Vertebrate organs require locally adapted blood vessels1,2. The gain of such organotypic vessel specializations is often deemed to be molecularly unrelated to the process of organ vascularization. Here, opposing this model, we reveal a molecular mechanism for brain-specific angiogenesis that operates under the control of Wnt7a/b ligands-well-known blood-brain barrier maturation signals3-5. The control mechanism relies on Wnt7a/b-dependent expression of Mmp25, which we find is enriched in brain endothelial cells. CRISPR-Cas9 mutagenesis in zebrafish reveals that this poorly characterized glycosylphosphatidylinositol-anchored matrix metalloproteinase is selectively required in endothelial tip cells to enable their initial migration across the pial basement membrane lining the brain surface. Mechanistically, Mmp25 confers brain invasive competence by cleaving meningeal fibroblast-derived collagen IV α5/6 chains within a short non-collagenous region of the central helical part of the heterotrimer. After genetic interference with the pial basement membrane composition, the Wnt-ß-catenin-dependent organotypic control of brain angiogenesis is lost, resulting in properly patterned, yet blood-brain-barrier-defective cerebrovasculatures. We reveal an organ-specific angiogenesis mechanism, shed light on tip cell mechanistic angiodiversity and thereby illustrate how organs, by imposing local constraints on angiogenic tip cells, can select vessels matching their distinctive physiological requirements.


Asunto(s)
Encéfalo , Neovascularización Fisiológica , Animales , Membrana Basal/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/citología , Encéfalo/citología , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Movimiento Celular , Colágeno Tipo IV/metabolismo , Sistemas CRISPR-Cas/genética , Células Endoteliales/metabolismo , Células Endoteliales/citología , Meninges/citología , Meninges/irrigación sanguínea , Meninges/metabolismo , Especificidad de Órganos , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
4.
Cell Prolif ; 57(8): e13627, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38421110

RESUMEN

The central nervous system (CNS) is surrounded by three membranes called meninges. Specialised fibroblasts, originating from the mesoderm and neural crest, primarily populate the meninges and serve as a binding agent. Our goal was to compare fibroblasts from meninges and skin obtained from the same human-aged donors, exploring their molecular and cellular characteristics related to CNS functions. We isolated meningeal fibroblasts (MFs) from brain donors and skin fibroblasts (SFs) from the same subjects. A functional analysis was performed measuring cell appearance, metabolic activity, and cellular orientation. We examined fibronectin, serpin H1, ß-III-tubulin, and nestin through qPCR and immunofluorescence. A whole transcriptome analysis was also performed to characterise the gene expression of MFs and SFs. MFs appeared more rapidly in the post-tissue processing, while SFs showed an elevated cellular metabolism and a well-defined cellular orientation. The four markers were mostly similar between the MFs and SFs, except for nestin, more expressed in MFs. Transcriptome analysis reveals significant differences, particularly in cyclic adenosine monophosphate (cAMP) metabolism and response to forskolin, both of which are upregulated in MFs. This study highlights MFs' unique characteristics, including the timing of appearance, metabolic activity, and gene expression patterns, particularly in cAMP metabolism and response to forskolin. These findings contribute to a deeper understanding of non-neuronal cells' involvement in CNS activities and potentially open avenues for therapeutic exploration.


Asunto(s)
Fibroblastos , Meninges , Piel , Transcriptoma , Humanos , Fibroblastos/metabolismo , Fibroblastos/citología , Piel/metabolismo , Piel/citología , Meninges/metabolismo , Meninges/citología , Perfilación de la Expresión Génica , Anciano , Células Cultivadas , Nestina/metabolismo , Nestina/genética , AMP Cíclico/metabolismo , Persona de Mediana Edad , Femenino , Masculino , Colforsina/farmacología
5.
Adv Healthc Mater ; 11(8): e2102153, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35007407

RESUMEN

Cognitive deficits (chemobrain) and peripheral neuropathy occur in ∼75% of patients treated for cancer with chemotherapy and persist long-term in >30% of survivors. Without preventive or curative interventions and with increasing survivorship rates, the population debilitated by these neurotoxicities is rising. Platinum-based chemotherapeutics, including cisplatin, induce neuronal mitochondrial defects leading to chemobrain and neuropathic pain. This study investigates the capacity of nasally administered mesenchymal stem cell-derived mitochondria coated with dextran-triphenylphosphonium polymer (coated mitochondria) to reverse these neurotoxicities. Nasally administered coated mitochondria are rapidly detectable in macrophages in the brain meninges but do not reach the brain parenchyma. The coated mitochondria change expression of >2400 genes regulating immune, neuronal, endocrine and vascular pathways in the meninges of mice treated with cisplatin. Nasal administration of coated mitochondria reverses cisplatin-induced cognitive deficits and resolves neuropathic pain at a >55-times lower dose compared to uncoated mitochondria. Reversal of these neuropathologies is associated with resolution of cisplatin-induced deficits in myelination, synaptosomal mitochondrial integrity and neurogenesis. These findings demonstrate that nasally administered coated mitochondria promote resolution of chemobrain and peripheral neuropathy, thereby identifying a novel facile strategy for clinical application of mitochondrial donation and treating central and peripheral nervous system pathologies by targeting the brain meninges.


Asunto(s)
Antineoplásicos , Deterioro Cognitivo Relacionado con la Quimioterapia , Neuralgia , Animales , Antineoplásicos/metabolismo , Cisplatino/farmacología , Humanos , Meninges/metabolismo , Ratones , Mitocondrias
6.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34479995

RESUMEN

Ectopic lymphoid tissue containing B cells forms in the meninges at late stages of human multiple sclerosis (MS) and when neuroinflammation is induced by interleukin (IL)-17 producing T helper (Th17) cells in rodents. B cell differentiation and the subsequent release of class-switched immunoglobulins have been speculated to occur in the meninges, but the exact cellular composition and underlying mechanisms of meningeal-dominated inflammation remain unknown. Here, we performed in-depth characterization of meningeal versus parenchymal Th17-induced rodent neuroinflammation. The most pronounced cellular and transcriptional differences between these compartments was the localization of B cells exhibiting a follicular phenotype exclusively to the meninges. Correspondingly, meningeal but not parenchymal Th17 cells acquired a B cell-supporting phenotype and resided in close contact with B cells. This preferential B cell tropism for the meninges and the formation of meningeal ectopic lymphoid tissue was partially dependent on the expression of the transcription factor Bcl6 in Th17 cells that is required in other T cell lineages to induce isotype class switching in B cells. A function of Bcl6 in Th17 cells was only detected in vivo and was reflected by the induction of B cell-supporting cytokines, the appearance of follicular B cells in the meninges, and of immunoglobulin class switching in the cerebrospinal fluid. We thus identify the induction of a B cell-supporting meningeal microenvironment by Bcl6 in Th17 cells as a mechanism controlling compartment specificity in neuroinflammation.


Asunto(s)
Enfermedades Neuroinflamatorias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Células Th17/metabolismo , Animales , Linfocitos B/inmunología , Comunicación Celular , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Centro Germinal/inmunología , Inflamación/metabolismo , Activación de Linfocitos , Masculino , Meninges/inmunología , Meninges/metabolismo , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/fisiopatología , Tejido Parenquimatoso/inmunología , Tejido Parenquimatoso/metabolismo , Proteínas Proto-Oncogénicas c-bcl-6/fisiología , Células Th17/inmunología , Células Th17/fisiología
7.
J Tissue Eng Regen Med ; 15(11): 948-963, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34463042

RESUMEN

Decellularization of extracellular matrices offers an alternative source of regenerative biomaterials that preserve biochemical structure and matrix components of native tissues. In this study, decellularized bovine spinal cord meninges (dSCM)-derived extracellular matrix hydrogel (MeninGEL) is fabricated by employing a protocol that involves physical, chemical, and enzymatic processing of spinal meninges tissue and preserves the biochemical structure of meninges. The success of decellularization is characterized by measuring the contents of residual DNA, glycosaminoglycans, and hydroxyproline, while a proteomics analysis is applied to reveal the composition of MeninGEL. Frequency and temperature sweep rheometry show that dSCM forms self-supporting hydrogel at physiological temperature. The MeninGEL possesses excellent cytocompatibility. Moreover, it is evidenced with immuno/histochemistry and gene expression studies that the hydrogel induces growth-factor free differentiation of human mesenchymal stem cells into neural-lineage cells. Furthermore, MeninGEL instructs human umbilical vein endothelial cells to form vascular branching. With its innate bioactivity and low batch-to-batch variation property, the MeninGEL has the potential to be an off-the-shelf product in nerve tissue regeneration and restoration.


Asunto(s)
Diferenciación Celular , Hidrogeles/farmacología , Meninges/metabolismo , Neovascularización Fisiológica , Neurogénesis , Médula Espinal/metabolismo , Ingeniería de Tejidos/métodos , Animales , Bovinos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colágeno/farmacología , ADN/metabolismo , Combinación de Medicamentos , Matriz Extracelular , Glicosaminoglicanos/metabolismo , Humanos , Hidroxiprolina/metabolismo , Cinética , Laminina/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Porosidad , Proteoglicanos/farmacología , Proteómica , Reología , Temperatura , Humectabilidad
8.
Cell Mol Life Sci ; 78(16): 6033-6049, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34274976

RESUMEN

Melanocytes are pigmented cells residing mostly in the skin and hair follicles of vertebrates, where they contribute to colouration and protection against UV-B radiation. However, the spectrum of their functions reaches far beyond that. For instance, these pigment-producing cells are found inside the inner ear, where they contribute to the hearing function, and in the heart, where they are involved in the electrical conductivity and support the stiffness of cardiac valves. The embryonic origin of such extracutaneous melanocytes is not clear. We took advantage of lineage-tracing experiments combined with 3D visualizations and gene knockout strategies to address this long-standing question. We revealed that Schwann cell precursors are recruited from the local innervation during embryonic development and give rise to extracutaneous melanocytes in the heart, brain meninges, inner ear, and other locations. In embryos with a knockout of the EdnrB receptor, a condition imitating Waardenburg syndrome, we observed only nerve-associated melanoblasts, which failed to detach from the nerves and to enter the inner ear. Finally, we looked into the evolutionary aspects of extracutaneous melanocytes and found that pigment cells are associated mainly with nerves and blood vessels in amphibians and fish. This new knowledge of the nerve-dependent origin of extracutaneous pigment cells might be directly relevant to the formation of extracutaneous melanoma in humans.


Asunto(s)
Encéfalo/fisiología , Oído Interno/fisiología , Corazón/fisiología , Meninges/fisiología , Sistema Nervioso/fisiopatología , Células de Schwann/fisiología , Anfibios/metabolismo , Anfibios/fisiología , Animales , Encéfalo/metabolismo , Linaje de la Célula/fisiología , Oído Interno/metabolismo , Desarrollo Embrionario/fisiología , Femenino , Peces/metabolismo , Peces/fisiología , Melanocitos/metabolismo , Melanocitos/fisiología , Meninges/metabolismo , Ratones , Sistema Nervioso/metabolismo , Embarazo , Receptor de Endotelina B/metabolismo , Células de Schwann/metabolismo
9.
Front Immunol ; 11: 571964, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33240262

RESUMEN

The potent costimulatory effect of CD137 has been implicated in several murine autoimmune disease models. CD137 costimulates and polarizes antigen-specific T cells toward a potent Th1/Tc1 response, and is essential for the development of experimental autoimmune encephalomyelitis (EAE), a murine model of Multiple Sclerosis (MS). This study aimed to investigate a role of CD137 in MS. Immunohistochemical and immunofluorescence staining of MS brain tissues was used to identify expression of CD137. CD137+ cells were identified in MS brain samples, with active lesions having the highest frequency of CD137+ cells. CD137 expression was found on several leukocyte subsets, including T cells, B cells and endothelial cells. In particular, CD137+ B cells were found in meningeal infiltrates. In vitro experiments showed that CD137 engagement on activated B cells increased early TNF and persistent IL-6 secretion with increased cell proliferation. These CD137+ B cells could interact with CD137L-expressing cells, secrete pro-inflammatory cytokines and accumulate in the meningeal infiltrate. This study demonstrates CD137 expression by activated B cells, enhancement of the inflammatory activity of B cells upon CD137 engagement, and provides evidence for a pathogenic role of CD137+ B cells in MS.


Asunto(s)
Ligando 4-1BB/metabolismo , Linfocitos B/inmunología , Interleucina-6/metabolismo , Meninges/metabolismo , Esclerosis Múltiple/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , Femenino , Humanos , Activación de Linfocitos , Masculino , Meninges/patología , Persona de Mediana Edad , Transducción de Señal , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
10.
Biomed Res Int ; 2020: 4616308, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33015166

RESUMEN

BACKGROUND: The brain is in many ways an immunologically and pharmacologically privileged site because of the blood-brain barrier (BBB). But for chronic peripheral inflammation, inflammatory signals can be transmitted from the peripheral system into the central nervous system (CNS) through multiple channels and result in neuroinflammation. Leptomeningeal cells that form the BBB can trigger one signaling pathway by releasing cytokines to transmit inflammatory signals. Besides, the Janus kinase (JAK) family may have a certain function in the activation of leptomeninges. In the present study, we try to use coniferyl aldehyde (CA), a natural anti-inflammatory phenolic compound, to inhibit this inflammatory process and elucidate the underlying molecular mechanisms. RESULTS: Secretion of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6) significantly increased after incubation with P. gingivalis. Moreover, TNF-α, IL-1ß, and IL-6 levels were upregulated, and the JAK2 signaling was enhanced in leptomeningeal cells in a conditioned medium from activated macrophages, which leads to the immune response in microglia. However, this inflammatory effect of leptomeningeal cells was reversed by CA administration, accompanied by the decreased immune response in microglia. The western blot assay revealed that JAK2 phosphorylation was suppressed in leptomeningeal cells treated with CA. CONCLUSIONS: This study demonstrates that activated macrophages by P. gingivalis markedly induce the release of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6) from leptomeningeal cells, thereby activating the JAK2 signaling pathway and subsequently enhancing immune responses in microglia in the CNS. CA effectively inhibits the inflammatory effect of leptomeningeal cells via suppressing the JAK2 signaling pathway.


Asunto(s)
Acroleína/análogos & derivados , Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Janus Quinasa 2/metabolismo , Meninges/efectos de los fármacos , Acroleína/farmacología , Animales , Línea Celular , Citocinas/metabolismo , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Meninges/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
11.
Nat Commun ; 11(1): 3866, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32737287

RESUMEN

Upon severe head injury (HI), blood vessels of the meninges and brain parenchyma are inevitably damaged. While limited vascular regeneration of the injured brain has been studied extensively, our understanding of meningeal vascular regeneration following head injury is quite limited. Here, we identify key pathways governing meningeal vascular regeneration following HI. Rapid and complete vascular regeneration in the meninges is predominantly driven by VEGFR2 signaling. Substantial increase of VEGFR2 is observed in both human patients and mouse models of HI, and endothelial cell-specific deletion of Vegfr2 in the latter inhibits meningeal vascular regeneration. We further identify the facilitating, stabilizing and arresting roles of Tie2, PDGFRß and Dll4 signaling, respectively, in meningeal vascular regeneration. Prolonged inhibition of this angiogenic process following HI compromises immunological and stromal integrity of the injured meninges. These findings establish a molecular framework for meningeal vascular regeneration after HI, and may guide development of wound healing therapeutics.


Asunto(s)
Traumatismos Craneocerebrales/genética , Células Endoteliales/metabolismo , Neovascularización Fisiológica/genética , Regeneración/genética , Transducción de Señal/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Circulación Cerebrovascular , Traumatismos Craneocerebrales/metabolismo , Traumatismos Craneocerebrales/patología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Regulación de la Expresión Génica/genética , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Meninges/lesiones , Meninges/metabolismo , Ratones , Ratones Noqueados , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/genética
12.
Acta Neuropathol Commun ; 8(1): 66, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32398070

RESUMEN

Analysis of isolated meninges and cerebrospinal fluid (CSF) of post-mortem MS cases has shown increased gene and protein expression for the pro-inflammatory cytokines: tumour necrosis factor (TNF) and interferon-γ (IFNγ). Here we tested the hypothesis that persistent production of these cytokines in the meningeal compartment and diffusion into underlying GM can drive chronic MS-like GM pathology. Lentiviral transfer vectors were injected into the sagittal sulcus of DA rats to deliver continuous expression of TNF + IFNγ transgenes in the meninges and the resulting neuropathology analysed after 1 and 2 months. Injection of TNF + IFNγ viral vectors, with or without prior MOG immunisation, induced extensive immune cell infiltration (CD4+ and CD8+ T-cells, CD79a + B-cells and macrophages) in the meninges by 28 dpi, which remained at 2 months. Control GFP viral vector did not induce infiltration. Subpial demyelination was seen underlying these infiltrates, which was partly dependant on prior myelin oligodendrocyte glycoprotein (MOG) immunisation. A significant decrease in neuronal numbers was seen at 28 and 56 days in cortical layers II-V that was independent of MOG immunisation. RNA analysis at 28 dpi showed an increase in expression of necroptotic pathway genes, including RIP3, MLKL, cIAP2 and Nox2. PhosphoRIP3+ and phosphoMLKL+ neurons were present in TNF + IFNγ vector injected animals, indicating activation of necroptosis. Our results suggest that persistent expression of TNF in the presence of IFNγ is a potent inducer of meningeal inflammation and can activate TNF signalling pathways in cortical cells leading to neuronal death and subpial demyelination and thus may contribute to clinical progression in MS.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Interferón gamma/metabolismo , Degeneración Nerviosa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Citocinas , Enfermedades Desmielinizantes/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/inmunología , Meninges/inmunología , Meninges/metabolismo , Meninges/patología , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Degeneración Nerviosa/inmunología , Ratas , Factor de Necrosis Tumoral alfa/inmunología
13.
J Comp Neurol ; 528(12): 2021-2032, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32003471

RESUMEN

Meningeal immunity along with its associated lymphatic vasculatures is widely discussed recently. Lymphatic vessels in meninges drain interstitial fluid into the deep-cervical lymph nodes. The vessels are composed of cells that express the cardinal marker for lymphatic endothelium-the lymphatic vessel hyaluronan receptor-1 (Lyve-1). However, studies also show the presence of nonendothelial Lyve-1 expressing cells in certain tissues. Therefore, we were curious if nonendothelial Lyve-1+ cells are also present in dura mater of meninges. We show that Lyve-1+ endothelial cells are distributed adjacent to the blood vessels in the brain dura mater of rats. We did not observe any lymphatic vessels in spinal dura mater. Interestingly, we also observed isolated population of nonlymphatic Lyve-1+ cells in both brain and spinal dura mater. Morphologically, the Lyve-1+ cells were extensively pleomorphic, sometimes elongated or round. Surprisingly, the thoracolumbal meningeal Lyve-1+ cells were predominantly round in morphology. Using endothelial specific marker VEGFR3 and macrophage markers CD68 and CD169, we observed that the isolated Lyve-1+ cells lacked endothelial cell signature, but were either CD68+ or CD169+ macrophages. Moreover, we observed that the Lyve-1+ cells colocalized with collagen fibers in the meninges, and some of Lyve-1+ cells had intracellular collagen. The study for the first time demonstrates the presence of Lyve-1 positive macrophages in the lymphatic and nonlymphatic regions in the meninges of rats.


Asunto(s)
Macrófagos/citología , Macrófagos/metabolismo , Meninges/citología , Meninges/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Femenino , Ratas , Ratas Endogámicas WKY , Médula Espinal/citología , Médula Espinal/metabolismo
14.
Eur J Pharmacol ; 875: 173035, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32097657

RESUMEN

Brain microglia cells are responsible for recognizing foreign bodies and act by activating other immune cells. Microglia react against infectious agents that cross the blood-brain barrier and release pro-inflammatory cytokines including interleukin (IL)-1ß, IL-33 and tumor necrosis factor (TNF). Mast cells (MCs) are immune cells also found in the brain meninges, in the perivascular spaces where they create a protective barrier and release pro-inflammatory compounds, such as IL-1ß, IL-33 and TNF. IL-1ß binds to the IL-1R1 receptor and activates a cascade of events that leads to the production of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and activation of the immune system. IL-33 is a member of the IL-1 family expressed by several immune cells including microglia and MCs and is involved in innate and adaptive immunity. IL-33 is a pleiotropic cytokine which binds the receptor ST2 derived from TLR/IL-1R super family and is released after cellular damage (also called "alarmin"). These cytokines are responsible for a number of brain inflammatory disorders. Activated IL-1ß in the brain stimulates microglia, MCs, and perivascular endothelial cells, mediating various inflammatory brain diseases. IL-37 also belongs to the IL-1 family and has the capacity to suppress IL-1ß with an anti-inflammatory property. IL-37 deficiency could activate and enhance myeloid differentiation (MyD88) and p38-dependent protein-activated mitogenic kinase (MAPK) with an increase in IL-1ß and IL-33 exacerbating neurological pathologies. In this article we report for the first time that microglia communicate and collaborate with MCs to produce pro-inflammatory cytokines that can be suppressed by IL-37 having a therapeutic potentiality.


Asunto(s)
Encefalopatías/inmunología , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1/metabolismo , Mastocitos/inmunología , Microglía/inmunología , Inmunidad Adaptativa , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encefalopatías/patología , Humanos , Inmunidad Innata , Mastocitos/metabolismo , Meninges/citología , Meninges/inmunología , Meninges/metabolismo , Meninges/patología , Microglía/metabolismo
15.
Acta Neuropathol Commun ; 8(1): 16, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32059751

RESUMEN

Subdural haematomas (SDHs) are characterized by rapidly or gradually accumulated haematomas between the arachnoid and dura mater. The mechanism of haematoma clearance has not been clearly elucidated until now. The meningeal lymphatic vessel (mLV) drainage pathway is a novel system that takes part in the clearance of waste products in the central nervous system (CNS). This study aimed to explore the roles of the mLV drainage pathway in SDH clearance and its impacting factors. We injected FITC-500D, A488-fibrinogen and autologous blood into the subdural space of mice/rats and found that these substances drained into deep cervical lymph nodes (dCLNs). FITC-500D was also observed in the lymphatic vessels (LYVE+) of the meninges and the dCLNs in mice. The SDH clearance rate in SDH rats that received deep cervical lymph vessel (dCLV) ligation surgery was significantly lower than that in the control group, as evaluated by haemoglobin quantification and MRI scanning. The drainage rate of mLVs was significantly slower after the SDH model was established, and the expression of lymphangiogenesis-related proteins, including LYVE1, FOXC2 and VEGF-C, in meninges was downregulated. In summary, our findings proved that SDH was absorbed through the mLV drainage pathway and that haematomas could inhibit the function of mLVs.


Asunto(s)
Hematoma Subdural/metabolismo , Ganglios Linfáticos/metabolismo , Sistema Linfático/metabolismo , Vasos Linfáticos/metabolismo , Meninges/metabolismo , Animales , Drenaje , Fluoresceína/administración & dosificación , Masculino , Ratones Endogámicos ICR , Ratas Sprague-Dawley
16.
World Neurosurg ; 137: e554-e563, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32068173

RESUMEN

BACKGROUND: MIR17 host gene (MIR17HG) is a potential therapeutic target for some cancer types. The aim of this study was to assess MIR17HG protein levels in patients with meningioma who had not been reported previously in the literature and comparing with normal meninges tissues. METHODS: MIR17HG protein levels were measured in 46 samples including 25 meningioma tissues procured during surgery and 21 normal meninges tissues obtained within 4 hours of death during autopsy procedures. Each sample was stored at -80°C until the evaluation of MIR17HG protein using a sandwich enzyme-linked immunoassay principle. Results were compared between the groups. RESULTS: MIR17HG protein levels were significantly higher in meningioma tissues compared with controls and difference was statistically significant (P = 0.012). Both World Health Organization grade I and grade II meningiomas had higher MIR17HG protein levels compared with controls and differences were statistically significant (P = 0.026 for grade I and P = 0.042 for grade II). Receiver operating characteristic curve analysis was performed to determine the cutoff of MIR17HG protein value in differentiating meningioma and control groups. At the cutoff value for MIR17HG protein of >0.0998 ng/mL, the sensitivity was 73.91%, 71.43%, and 77.78% and area under the curve was 0.756, 0.753, and 0.761 for meningioma group, grade I, and grade II subgroups, respectively, and specificity was 69.23% for each group. CONCLUSIONS: MIR17HG protein expression was found to have a higher level in meningiomas than in normal meninges tissues in our study. Considering the recurrence and irresectability for some meningiomas, which require further treatment, MIR17HG may be a new target for treatment in meningiomas and our study will shed light on further studies.


Asunto(s)
Neoplasias Meníngeas/metabolismo , Meninges/metabolismo , Meningioma/metabolismo , MicroARNs/metabolismo , Adulto , Anciano , Femenino , Humanos , Masculino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Meninges/patología , Meningioma/genética , Meningioma/patología , MicroARNs/genética , Persona de Mediana Edad
17.
Neurocrit Care ; 33(1): 140-151, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31768758

RESUMEN

BACKGROUND: How inflammatory cells are recruited into the central nervous system is a topic of interest in a number of neurological injuries. In aneurysmal subarachnoid hemorrhage (SAH), neutrophil accumulation in the central nervous system 3 days after the hemorrhage is a critical step in the development of delayed cerebral injury (DCI). The mechanism by which neutrophils enter the central nervous system is still unclear. METHODS AND RESULTS: To identify human effectors of neutrophil recruitment, cerebrospinal fluid (CSF) samples were taken from a small, selected sample of SAH patients with external ventricular drainage devices (10 patients). Among a battery of CSF cytokines tested 3 days after SAH, five cytokines were associated with poor 90-day outcome (modified Rankin Score 3-6). A parallel study in a mouse model of mild SAH showed elevation in three cytokines in the CNS compared to sham. IL-17 and IL-2 were increased in both patients and the mouse model. IL-17 was investigated further because of its known role in neutrophil recruitment. Inhibition of RAR-Related Orphan Receptor Gamma T, the master transcription factor of IL-17, with the inverse agonist GSK805 suppressed neutrophils entry into the CNS after SAH compared to control. Using an IL-17 reporter mouse, we investigated the source of IL-17 and found that myeloid cells were a common IL-17-producing cell type in the meninges after SAH, suggesting an autocrine role for neutrophil recruitment. CONCLUSIONS: Taken together, IL-17 appears to be in important factor in the recruitment of neutrophils into the meninges after SAH and could be an important target for therapies to ameliorate DCI.


Asunto(s)
Encéfalo/inmunología , Citocinas/inmunología , Interleucina-17/inmunología , Meninges/inmunología , Infiltración Neutrófila/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Hemorragia Subaracnoidea/inmunología , Animales , Velocidad del Flujo Sanguíneo , Encéfalo/metabolismo , Quimiocina CXCL2/inmunología , Quimiocina CXCL2/metabolismo , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Interleucina-17/metabolismo , Interleucina-1alfa/inmunología , Interleucina-1alfa/metabolismo , Interleucina-2/inmunología , Interleucina-2/metabolismo , Meninges/metabolismo , Ratones , Células Mieloides/inmunología , Infiltración Neutrófila/efectos de los fármacos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Hemorragia Subaracnoidea/metabolismo , Hemorragia Subaracnoidea/fisiopatología , Receptor Activador Expresado en Células Mieloides 1/inmunología , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Ultrasonografía Doppler Transcraneal , Vasoespasmo Intracraneal/diagnóstico por imagen , Vasoespasmo Intracraneal/fisiopatología
18.
Mol Neurobiol ; 57(3): 1484-1501, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31773411

RESUMEN

The leptomeninges, referring to the arachnoid and pia mater and their projections into the perivascular compartments in the central nervous system, actively participate in diverse biological processes including fluid homeostasis, immune cell infiltrations, and neurogenesis, yet their detailed cellular and molecular identities remain elusive. This study aimed to characterize platelet-derived growth factor beta (PDGFR-ß)-expressing cells in the leptomeninges in the adult rat brain using light and electron microscopy. PDGFR-ß+ cells were observed in the inner arachnoid, arachnoid trabeculae, pia mater, and leptomeningeal sheath of the subarachnoid vessels, thereby forming a cellular network throughout the leptomeninges. Leptomeningeal PDGFR-ß+ cells were commonly characterized by large euchromatic nuclei, thin branching processes forming web-like network, and the expression of the intermediate filaments nestin and vimentin. These cells were typical of active fibroblasts with a well-developed rough endoplasmic reticulum and close spatial correlation with collagen fibrils. Leptomeningeal PDGFR-ß+ cells ensheathing the vasculature in the subarachnoid space joined with pial PDGFR-ß+ cells upon entering the cortical parenchyma, yet perivascular PDGFR-ß+ cells in these penetrating vessels underwent abrupt changes in their morphological and molecular characteristics: they became more flattened with loss of immunoreactivity for nestin and vimentin and deficient collagen deposition, which was indicative of inactive fibroblasts termed fibrocytes. In the cortical parenchyma, PDGFR-ß immunoreactivity was almost exclusively localized to larger caliber vessels, and significantly decreased in capillary-like microvessels. Collectively, our data identify PDGFR-ß as a novel cellular marker for leptomeningeal fibroblasts comprising the leptomeninges and perivascular adventitial cells of the subarachnoid and penetrating large-sized cortical vasculatures.


Asunto(s)
Aracnoides/metabolismo , Encéfalo/ultraestructura , Meninges/metabolismo , Meninges/ultraestructura , Animales , Aracnoides/ultraestructura , Encéfalo/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Microscopía Electrónica/métodos , Piamadre/patología , Piamadre/ultraestructura , Proteínas Proto-Oncogénicas c-sis/metabolismo , Ratas , Vimentina/metabolismo , Vimentina/ultraestructura
19.
J Neuroinflammation ; 16(1): 259, 2019 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-31810488

RESUMEN

BACKGROUND: Recent studies of cortical pathology in secondary progressive multiple sclerosis have shown that a more severe clinical course and the presence of extended subpial grey matter lesions with significant neuronal/glial loss and microglial activation are associated with meningeal inflammation, including the presence of lymphoid-like structures in the subarachnoid space in a proportion of cases. METHODS: To investigate the molecular consequences of pro-inflammatory and cytotoxic molecules diffusing from the meninges into the underlying grey matter, we carried out gene expression profiling analysis of the motor cortex from 20 post-mortem multiple sclerosis brains with and without substantial meningeal inflammation and 10 non-neurological controls. RESULTS: Gene expression profiling of grey matter lesions and normal appearing grey matter not only confirmed the substantial pathological cell changes, which were greatest in multiple sclerosis cases with increased meningeal inflammation, but also demonstrated the upregulation of multiple genes/pathways associated with the inflammatory response. In particular, genes involved in tumour necrosis factor (TNF) signalling were significantly deregulated in MS cases compared with controls. Increased meningeal inflammation was found to be associated with a shift in the balance of TNF signalling away from TNFR1/TNFR2 and NFkB-mediated anti-apoptotic pathways towards TNFR1- and RIPK3-mediated pro-apoptotic/pro-necroptotic signalling in the grey matter, which was confirmed by RT-PCR analysis. TNFR1 was found expressed preferentially on neurons and oligodendrocytes in MS cortical grey matter, whereas TNFR2 was predominantly expressed by astrocytes and microglia. CONCLUSIONS: We suggest that the inflammatory milieu generated in the subarachnoid space of the multiple sclerosis meninges by infiltrating immune cells leads to increased demyelinating and neurodegenerative pathology in the underlying grey matter due to changes in the balance of TNF signalling.


Asunto(s)
Corteza Cerebral/metabolismo , Sustancia Gris/metabolismo , Meninges/metabolismo , Esclerosis Múltiple/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Corteza Cerebral/patología , Femenino , Sustancia Gris/patología , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Masculino , Meninges/patología , Persona de Mediana Edad , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Transcriptoma/fisiología , Factor de Necrosis Tumoral alfa/genética
20.
Ann Neurol ; 84(6): 829-842, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30362156

RESUMEN

OBJECTIVE: Cortical gray matter (GM) pathology, involving demyelination and neurodegeneration, associated with meningeal inflammation, could be important in determining disability progression in multiple sclerosis (MS). However, we need to know more about how cortical demyelination, neurodegeneration, and meningeal inflammation contribute to pathology at early stages of MS to better predict long-term outcome. METHODS: Tissue blocks from short disease duration MS (n = 12, median disease duration = 2 years), progressive MS (n = 21, disease duration = 25 years), non-diseased controls (n = 11), and other neurological inflammatory disease controls (n = 6) were quantitatively analyzed by immunohistochemistry, immunofluorescence, and in situ hybridization. RESULTS: Cortical GM demyelination was extensive in some cases of acute MS (range = 1-48% of total cortical GM), and subpial lesions were the most common type (62%). The numbers of activated (CD68+ ) microglia/macrophages were increased in cases with subpial lesions, and the density of neurons was significantly reduced in acute MS normal appearing and lesion GM, compared to controls (p < 0.005). Significant meningeal inflammation and lymphoid-like structures were seen in 4 of 12 acute MS cases. The extent of meningeal inflammation correlated with microglial/macrophage activation (p < 0.05), but not the area of cortical demyelination, reflecting the finding that lymphoid-like structures were seen adjacent to GM lesions as well as areas of partially demyelinated/remyelinated, cortical GM. INTERPRETATION: Our findings demonstrate that cortical demyelination, neuronal loss, and meningeal inflammation are notable pathological hallmarks of acute MS and support the need to identify early biomarkers of this pathology to better predict outcome. Ann Neurol 2018;84:829-842.


Asunto(s)
Corteza Cerebral/patología , Inflamación/complicaciones , Meninges/patología , Esclerosis Múltiple/complicaciones , Vaina de Mielina/patología , Adulto , Anciano , Antígenos CD/metabolismo , Corteza Cerebral/metabolismo , Estudios de Cohortes , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Femenino , Sustancia Gris/metabolismo , Sustancia Gris/patología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Meninges/metabolismo , Microglía/metabolismo , Microglía/patología , Persona de Mediana Edad , Vaina de Mielina/metabolismo , Factores de Transcripción/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA