Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.896
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(17): 4733-4750.e26, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-38971152

RESUMEN

We identify a population of Protogenin-positive (PRTG+ve) MYChigh NESTINlow stem cells in the four-week-old human embryonic hindbrain that subsequently localizes to the ventricular zone of the rhombic lip (RLVZ). Oncogenic transformation of early Prtg+ve rhombic lip stem cells initiates group 3 medulloblastoma (Gr3-MB)-like tumors. PRTG+ve stem cells grow adjacent to a human-specific interposed vascular plexus in the RLVZ, a phenotype that is recapitulated in Gr3-MB but not in other types of medulloblastoma. Co-culture of Gr3-MB with endothelial cells promotes tumor stem cell growth, with the endothelial cells adopting an immature phenotype. Targeting the PRTGhigh compartment of Gr3-MB in vivo using either the diphtheria toxin system or chimeric antigen receptor T cells constitutes effective therapy. Human Gr3-MBs likely arise from early embryonic RLVZ PRTG+ve stem cells inhabiting a specific perivascular niche. Targeting the PRTGhigh compartment and/or the perivascular niche represents an approach to treat children with Gr3-MB.


Asunto(s)
Meduloblastoma , Células Madre Neoplásicas , Humanos , Meduloblastoma/patología , Meduloblastoma/metabolismo , Animales , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Ratones , Rombencéfalo/metabolismo , Rombencéfalo/embriología , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Células Endoteliales/metabolismo , Nicho de Células Madre , Células Madre/metabolismo , Técnicas de Cocultivo , Estructuras Embrionarias , Metencéfalo/embriología
2.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38733989

RESUMEN

The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.


Asunto(s)
Cerebelo , Neuronas , Retina , Animales , Femenino , Masculino , Ratones , Cerebelo/metabolismo , Cerebelo/irrigación sanguínea , Cerebelo/citología , Canales Iónicos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Vasos Retinianos/metabolismo
3.
Cell ; 186(20): 4454-4471.e19, 2023 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-37703875

RESUMEN

Macrophages are heterogeneous and play critical roles in development and disease, but their diversity, function, and specification remain inadequately understood during human development. We generated a single-cell RNA sequencing map of the dynamics of human macrophage specification from PCW 4-26 across 19 tissues. We identified a microglia-like population and a proangiogenic population in 15 macrophage subtypes. Microglia-like cells, molecularly and morphologically similar to microglia in the CNS, are present in the fetal epidermis, testicle, and heart. They are the major immune population in the early epidermis, exhibit a polarized distribution along the dorsal-lateral-ventral axis, and interact with neural crest cells, modulating their differentiation along the melanocyte lineage. Through spatial and differentiation trajectory analysis, we also showed that proangiogenic macrophages are perivascular across fetal organs and likely yolk-sac-derived as microglia. Our study provides a comprehensive map of the heterogeneity and developmental dynamics of human macrophages and unravels their diverse functions during development.


Asunto(s)
Macrófagos , Humanos , Diferenciación Celular , Linaje de la Célula , Macrófagos/citología , Microglía , Especificidad de Órganos
4.
Immunity ; 55(5): 862-878.e8, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35508166

RESUMEN

Macrophage colony stimulating factor-1 (CSF-1) plays a critical role in maintaining myeloid lineage cells. However, congenital global deficiency of CSF-1 (Csf1op/op) causes severe musculoskeletal defects that may indirectly affect hematopoiesis. Indeed, we show here that osteolineage-derived Csf1 prevented developmental abnormalities but had no effect on monopoiesis in adulthood. However, ubiquitous deletion of Csf1 conditionally in adulthood decreased monocyte survival, differentiation, and migration, independent of its effects on bone development. Bone histology revealed that monocytes reside near sinusoidal endothelial cells (ECs) and leptin receptor (Lepr)-expressing perivascular mesenchymal stromal cells (MSCs). Targeted deletion of Csf1 from sinusoidal ECs selectively reduced Ly6C- monocytes, whereas combined depletion of Csf1 from ECs and MSCs further decreased Ly6Chi cells. Moreover, EC-derived CSF-1 facilitated recovery of Ly6C- monocytes and protected mice from weight loss following induction of polymicrobial sepsis. Thus, monocytes are supported by distinct cellular sources of CSF-1 within a perivascular BM niche.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos , Células Madre Mesenquimatosas , Animales , Médula Ósea , Células de la Médula Ósea , Células Endoteliales , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Monocitos
5.
Immunity ; 55(4): 606-622.e6, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35358427

RESUMEN

Lymph node (LN) stromal cells play a crucial role in LN development and in supporting adaptive immune responses. However, their origin, differentiation pathways, and transcriptional programs are still elusive. Here, we used lineage-tracing approaches and single-cell transcriptome analyses to determine origin, transcriptional profile, and composition of LN stromal and endothelial progenitors. Our results showed that all major stromal cell subsets and a large proportion of blood endothelial cells originate from embryonic Hoxb6+ progenitors of the lateral plate mesoderm (LPM), whereas lymphatic endothelial cells arise from Pax3+ progenitors of the paraxial mesoderm (PXM). Single-cell RNA sequencing revealed the existence of different Cd34+ and Cxcl13+ stromal cell subsets and showed that embryonic LNs contain proliferating progenitors possibly representing the amplifying populations for terminally differentiated cells. Taken together, our work identifies the earliest embryonic sources of LN stromal and endothelial cells and demonstrates that stromal diversity begins already during LN development.


Asunto(s)
Células Endoteliales , Células Endoteliales/metabolismo , Ganglios Linfáticos , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Células del Estroma , Factores de Transcripción/metabolismo
6.
Immunity ; 54(1): 176-190.e7, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33333014

RESUMEN

The developmental and molecular heterogeneity of tissue macrophages is unravelling, as are their diverse contributions to physiology and pathophysiology. Moreover, also given tissues harbor macrophages in discrete anatomic locations. Functional contributions of specific cell populations can in mice be dissected using Cre recombinase-mediated mutagenesis. However, single promoter-based Cre models show limited specificity for cell types. Focusing on macrophages in the brain, we establish here a binary transgenic system involving complementation-competent NCre and CCre fragments whose expression is driven by distinct promoters: Sall1ncre: Cx3cr1ccre mice specifically target parenchymal microglia and compound transgenic Lyve1ncre: Cx3cr1ccre animals target vasculature-associated macrophages, in the brain, as well as other tissues. We imaged the respective cell populations and retrieved their specific translatomes using the RiboTag in order to define them and analyze their differential responses to a challenge. Collectively, we establish the value of binary transgenesis to dissect tissue macrophage compartments and their functions.


Asunto(s)
Encéfalo/citología , Sistema Nervioso Central/fisiología , Integrasas/metabolismo , Macrófagos/fisiología , Microglía/fisiología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos
7.
Immunol Rev ; 324(1): 95-103, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38747455

RESUMEN

Fat is stored in distinct depots with unique features in both mice and humans and B cells reside in all adipose depots. We have shown that B cells modulate cardiometabolic disease through activities in two of these key adipose depots: visceral adipose tissue (VAT) and perivascular adipose tissue (PVAT). VAT refers to the adipose tissue surrounding organs, within the abdomen and thorax, and is comprised predominantly of white adipocytes. This depot has been implicated in mediating obesity-related dysmetabolism. PVAT refers to adipose tissue surrounding major arteries. It had long been thought to exist to provide protection and insulation for the vessel, yet recent work demonstrates an important role for PVAT in harboring immune cells, promoting their function and regulating the biology of the underlying vessel. The role of B-2 cells and adaptive immunity in adipose tissue biology has been nicely reviewed elsewhere. Given that, the predominance of B-1 cells in adipose tissue at homeostasis, and the emerging role of B-1 cells in a variety of disease states, we will focus this review on how B-1 cells function in VAT and PVAT depots to promote homeostasis and limit inflammation linked to cardiometabolic disease and factors that regulate this function.


Asunto(s)
Tejido Adiposo , Inmunidad Innata , Inflamación , Humanos , Animales , Inflamación/inmunología , Inflamación/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/etiología , Inmunomodulación
8.
Immunity ; 48(5): 1014-1028.e6, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752062

RESUMEN

Stromal cells (SCs) establish the compartmentalization of lymphoid tissues critical to the immune response. However, the full diversity of lymph node (LN) SCs remains undefined. Using droplet-based single-cell RNA sequencing, we identified nine peripheral LN non-endothelial SC clusters. Included are the established subsets, Ccl19hi T-zone reticular cells (TRCs), marginal reticular cells, follicular dendritic cells (FDCs), and perivascular cells. We also identified Ccl19lo TRCs, likely including cholesterol-25-hydroxylase+ cells located at the T-zone perimeter, Cxcl9+ TRCs in the T-zone and interfollicular region, CD34+ SCs in the capsule and medullary vessel adventitia, indolethylamine N-methyltransferase+ SCs in the medullary cords, and Nr4a1+ SCs in several niches. These data help define how transcriptionally distinct LN SCs support niche-restricted immune functions and provide evidence that many SCs are in an activated state.


Asunto(s)
Ganglios Linfáticos/inmunología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Células del Estroma/inmunología , Transcriptoma/inmunología , Animales , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Quimiocina CCL19/metabolismo , Células Dendríticas Foliculares/inmunología , Células Dendríticas Foliculares/metabolismo , Femenino , Ganglios Linfáticos/metabolismo , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Ratones Endogámicos C57BL , Células del Estroma/metabolismo
9.
Proc Natl Acad Sci U S A ; 121(42): e2407246121, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39374384

RESUMEN

The glymphatic pathway was defined in rodents as a network of perivascular spaces (PVSs) that facilitates organized distribution of cerebrospinal fluid (CSF) into the brain parenchyma. To date, perivascular CSF and cerebral interstitial fluid exchange has not been shown in humans. Using intrathecal gadolinium contrast-enhanced MRI, we show that contrast-enhanced CSF moves through the PVS into the parenchyma, supporting the existence of a glymphatic pathway in humans.


Asunto(s)
Líquido Cefalorraquídeo , Sistema Glinfático , Imagen por Resonancia Magnética , Humanos , Sistema Glinfático/fisiología , Sistema Glinfático/diagnóstico por imagen , Líquido Cefalorraquídeo/fisiología , Líquido Cefalorraquídeo/metabolismo , Imagen por Resonancia Magnética/métodos , Masculino , Medios de Contraste , Adulto , Femenino , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Gadolinio , Persona de Mediana Edad
10.
Development ; 150(19)2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37756588

RESUMEN

Perivascular fibroblasts (PVFs) are a fibroblast-like cell type that reside on large-diameter blood vessels in the adult meninges and central nervous system (CNS). PVFs contribute to fibrosis following injury but their homeostatic functions are not defined. PVFs were previously shown to be absent from most brain regions at birth and are only detected postnatally within the cerebral cortex. However, the origin, timing and cellular mechanisms of PVF development are not known. We used Col1a1-GFP and Col1a2-CreERT2 transgenic mice to track PVF development postnatally. Using lineage tracing and in vivo imaging we show that brain PVFs originate from the meninges and are first seen on parenchymal cerebrovasculature at postnatal day (P) 5. After P5, PVF coverage of the cerebrovasculature expands via local cell proliferation and migration from the meninges. Finally, we show that PVFs and perivascular macrophages develop concurrently. These findings provide the first complete timeline for PVF development in the brain, enabling future work into how PVF development is coordinated with cell types and structures in and around the perivascular spaces to support normal CNS vascular function.

11.
Proc Natl Acad Sci U S A ; 120(46): e2307480120, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37943835

RESUMEN

Ischemic diseases lead to considerable morbidity and mortality, yet conventional clinical treatment strategies for therapeutic angiogenesis fall short of being impactful. Despite the potential of biomaterials to deliver pro-angiogenic molecules at the infarct site to induce angiogenesis, their efficacy has been impeded by aberrant vascular activation and off-target circulation. Here, we present a semisynthetic low-molecular sulfated chitosan oligosaccharide (SCOS) that efficiently induces therapeutic arteriogenesis with a spontaneous generation of collateral circulation and blood reperfusion in rodent models of hind limb ischemia and myocardial infarction. SCOS elicits anti-inflammatory macrophages' (Mφs') differentiation into perivascular Mφs, which in turn directs artery formation via a cell-to-cell communication rather than secretory factor regulation. SCOS-mediated arteriogenesis requires a canonical Notch signaling pathway in Mφs via the glycosylation of protein O-glucosyltransferases 2, which results in promoting arterial differentiation and tissue repair in ischemia. Thus, this highly bioactive oligosaccharide can be harnessed to direct efficiently therapeutic arteriogenesis and perfusion for the treatment of ischemic diseases.


Asunto(s)
Neovascularización Fisiológica , Sulfatos , Ratones , Animales , Neovascularización Fisiológica/fisiología , Sulfatos/metabolismo , Ratones Noqueados , Músculo Esquelético/metabolismo , Isquemia/metabolismo , Macrófagos/metabolismo , Miembro Posterior/irrigación sanguínea , Modelos Animales de Enfermedad
12.
Proc Natl Acad Sci U S A ; 120(14): e2217744120, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36989300

RESUMEN

Quantifying the flow of cerebrospinal fluid (CSF) is crucial for understanding brain waste clearance and nutrient delivery, as well as edema in pathological conditions such as stroke. However, existing in vivo techniques are limited to sparse velocity measurements in pial perivascular spaces (PVSs) or low-resolution measurements from brain-wide imaging. Additionally, volume flow rate, pressure, and shear stress variation in PVSs are essentially impossible to measure in vivo. Here, we show that artificial intelligence velocimetry (AIV) can integrate sparse velocity measurements with physics-informed neural networks to quantify CSF flow in PVSs. With AIV, we infer three-dimensional (3D), high-resolution velocity, pressure, and shear stress. Validation comes from training with 70% of PTV measurements and demonstrating close agreement with the remaining 30%. A sensitivity analysis on the AIV inputs shows that the uncertainty in AIV inferred quantities due to uncertainties in the PVS boundary locations inherent to in vivo imaging is less than 30%, and the uncertainty from the neural net initialization is less than 1%. In PVSs of N = 4 wild-type mice we find mean flow speed 16.33 ± 11.09 µm/s, volume flow rate 2.22 ± 1.983 × 103 µm3/s, axial pressure gradient ( - 2.75 ± 2.01)×10-4 Pa/µm (-2.07 ± 1.51 mmHg/m), and wall shear stress (3.00 ± 1.45)×10-3 Pa (all mean ± SE). Pressure gradients, flow rates, and resistances agree with prior predictions. AIV infers in vivo PVS flows in remarkable detail, which will improve fluid dynamic models and potentially clarify how CSF flow changes with aging, Alzheimer's disease, and small vessel disease.


Asunto(s)
Inteligencia Artificial , Redes Neurales de la Computación , Animales , Ratones , Reología/métodos , Encéfalo , Física , Velocidad del Flujo Sanguíneo
13.
Development ; 149(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35217866

RESUMEN

In the developing mammalian brain, neuroepithelial cells interact with blood vessels to regulate angiogenesis, blood-brain barrier maturation and other key neurovascular functions. Genetic studies in mice have shown that neurovascular development is controlled, in part, by Itgb8, which encodes the neuroepithelial cell-expressed integrin ß8 subunit. However, these studies have involved complete loss-of-function Itgb8 mutations, and have not discerned the relative roles for the ß8 integrin extracellular matrix (ECM) binding region versus the intracellular signaling tail. Here, Cre/lox strategies have been employed to selectively delete the cytoplasmic tail of murine Itgb8 without perturbing its transmembrane and extracellular domains. We report that the ß8 integrin cytoplasmic domain is essential for inside-out modulation of adhesion, including activation of latent-TGFßs in the ECM. Quantitative sequencing of the brain endothelial cell transcriptome identifies TGFß-regulated genes with putative links to blood vessel morphogenesis, including several genes linked to Wnt/ß-catenin signaling. These results reveal that the ß8 integrin cytoplasmic domain is essential for the regulation of TGFß-dependent gene expression in endothelial cells and suggest that cross-talk between TGFßs and Wnt pathways is crucial for neurovascular development.


Asunto(s)
Células Endoteliales , Cadenas beta de Integrinas , Animales , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Integrinas/genética , Integrinas/metabolismo , Mamíferos/metabolismo , Ratones , Factor de Crecimiento Transformador beta/metabolismo
14.
Brain ; 147(9): 3059-3069, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39049445

RESUMEN

Perivascular macrophages (PVMs) and, to a lesser degree, microglia are targets and reservoirs of HIV and simian immunodeficiency virus (SIV) in the brain. Previously, we demonstrated that colony-stimulating factor 1 receptor (CSF1R) in PVMs was upregulated and activated in chronically SIV-infected rhesus macaques with encephalitis, correlating with SIV infection of PVMs. Herein, we investigated the role of CSF1R in the brain during acute SIV infection using BLZ945, a brain-penetrant CSF1R kinase inhibitor. Apart from three uninfected historic controls, nine Indian rhesus macaques were infected acutely with SIVmac251 and divided into three groups (n = 3 each): an untreated control and two groups treated for 20-30 days with low- (10 mg/kg/day) or high- (30 mg/kg/day) dose BLZ945. With the high-dose BLZ945 treatment, there was a significant reduction in cells expressing CD163 and CD206 across all four brain areas examined, compared with the low-dose treatment and control groups. In 9 of 11 tested regions, tissue viral DNA (vDNA) loads were reduced by 95%-99% following at least one of the two doses, and even to undetectable levels in some instances. Decreased numbers of CD163+ and CD206+ cells correlated significantly with lower levels of vDNA in all four corresponding brain areas. In contrast, BLZ945 treatment did not significantly affect the number of microglia. Our results indicate that doses as low as 10 mg/kg/day of BLZ945 are sufficient to reduce the tissue vDNA loads in the brain with no apparent adverse effect. This study provides evidence that infected PVMs are highly sensitive to CSF1R inhibition, opening new possibilities to achieve viral clearance.


Asunto(s)
Encéfalo , Macaca mulatta , Macrófagos , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/virología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Carga Viral/efectos de los fármacos , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Antígenos CD/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/virología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Receptores de Superficie Celular/metabolismo , Anisoles
15.
Brain ; 147(1): 39-55, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-37691438

RESUMEN

Brain perivascular macrophages are specialized populations of macrophages that reside in the space around cerebral vessels, such as penetrating arteries and venules. With the help of cutting-edge technologies, such as cell fate mapping and single-cell multi-omics, their multifaceted, pivotal roles in phagocytosis, antigen presentation, vascular integrity maintenance and metabolic regulation have more recently been further revealed under physiological conditions. Accumulating evidence also implies that perivascular macrophages are involved in the pathogenesis of neurodegenerative disease, cerebrovascular dysfunction, autoimmune disease, traumatic brain injury and epilepsy. They can act in either protective or detrimental ways depending on the disease course and stage. However, the underlying mechanisms of perivascular macrophages remain largely unknown. Therefore, we highlight potential future directions in research on perivascular macrophages, including the utilization of genetic mice and novel therapeutic strategies that target these unique immune cells for neuroprotective purposes. In conclusion, this review provides a comprehensive update on the current knowledge of brain perivascular macrophages, shedding light on their pivotal roles in central nervous system health and disease.


Asunto(s)
Enfermedades Neurodegenerativas , Ratones , Animales , Enfermedades Neurodegenerativas/patología , Encéfalo/patología , Macrófagos/metabolismo , Sistema Nervioso Central , Fagocitosis
16.
Brain ; 147(6): 2214-2229, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38802114

RESUMEN

Mild traumatic brain injury (mTBI) has emerged as a potential risk factor for the development of neurodegenerative conditions such as Alzheimer's disease and chronic traumatic encephalopathy. Blast mTBI, caused by exposure to a pressure wave from an explosion, is predominantly experienced by military personnel and has increased in prevalence and severity in recent decades. Yet the underlying pathology of blast mTBI is largely unknown. We examined the expression and localization of AQP4 in human post-mortem frontal cortex and observed distinct laminar differences in AQP4 expression following blast exposure. We also observed similar laminar changes in AQP4 expression and localization and delayed impairment of glymphatic function that emerged 28 days following blast injury in a mouse model of repetitive blast mTBI. In a cohort of veterans with blast mTBI, we observed that blast exposure was associated with an increased burden of frontal cortical MRI-visible perivascular spaces, a putative neuroimaging marker of glymphatic perivascular dysfunction. These findings suggest that changes in AQP4 and delayed glymphatic impairment following blast injury may render the post-traumatic brain vulnerable to post-concussive symptoms and chronic neurodegeneration.


Asunto(s)
Acuaporina 4 , Traumatismos por Explosión , Sistema Glinfático , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Acuaporina 4/metabolismo , Traumatismos por Explosión/complicaciones , Traumatismos por Explosión/patología , Traumatismos por Explosión/metabolismo , Conmoción Encefálica/metabolismo , Conmoción Encefálica/complicaciones , Conmoción Encefálica/patología , Conmoción Encefálica/fisiopatología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Lóbulo Frontal/diagnóstico por imagen , Sistema Glinfático/metabolismo , Sistema Glinfático/patología , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Veteranos
17.
Cell Mol Life Sci ; 81(1): 31, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38212566

RESUMEN

The intricate relationship between the central nervous system (CNS) and the immune system plays a crucial role in the pathogenesis of various neurological diseases. Understanding the interactions among the immunopathological processes at the brain borders is essential for advancing our knowledge of disease mechanisms and developing novel diagnostic and therapeutic approaches. In this review, we explore the emerging role of neuroimaging in providing valuable insights into brain barrier inflammation and brain fluid drainage in human neurological diseases. Neuroimaging techniques have enabled us not only to visualize and assess brain structures, but also to study the dynamics of the CNS in health and disease in vivo. By analyzing imaging findings, we can gain a deeper understanding of the immunopathology observed at the brain-immune interface barriers, which serve as critical gatekeepers that regulate immune cell trafficking, cytokine release, and clearance of waste products from the brain. This review explores the integration of neuroimaging data with immunopathological findings, providing valuable insights into brain barrier integrity and immune responses in neurological diseases. Such integration may lead to the development of novel diagnostic markers and targeted therapeutic approaches that can benefit patients with neurological disorders.


Asunto(s)
Sistema Glinfático , Enfermedades del Sistema Nervioso , Humanos , Sistema Glinfático/patología , Encéfalo/patología , Sistema Nervioso Central/patología , Enfermedades del Sistema Nervioso/diagnóstico por imagen , Enfermedades del Sistema Nervioso/terapia , Enfermedades del Sistema Nervioso/patología , Inflamación/diagnóstico por imagen , Inflamación/patología , Barrera Hematoencefálica/diagnóstico por imagen
18.
Cell Mol Life Sci ; 81(1): 239, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38801464

RESUMEN

The brain's network of perivascular channels for clearance of excess fluids and waste plays a critical role in the pathogenesis of several neurodegenerative diseases including cerebral amyloid angiopathy (CAA). CAA is the main cause of hemorrhagic stroke in the elderly, the most common vascular comorbidity in Alzheimer's disease and also implicated in adverse events related to anti-amyloid immunotherapy. Remarkably, the mechanisms governing perivascular clearance of soluble amyloid ß-a key culprit in CAA-from the brain to draining lymphatics and systemic circulation remains poorly understood. This knowledge gap is critically important to bridge for understanding the pathophysiology of CAA and accelerate development of targeted therapeutics. The authors of this review recently converged their diverse expertise in the field of perivascular physiology to specifically address this problem within the framework of a Leducq Foundation Transatlantic Network of Excellence on Brain Clearance. This review discusses the overarching goal of the consortium and explores the evidence supporting or refuting the role of impaired perivascular clearance in the pathophysiology of CAA with a focus on translating observations from rodents to humans. We also discuss the anatomical features of perivascular channels as well as the biophysical characteristics of fluid and solute transport.


Asunto(s)
Péptidos beta-Amiloides , Encéfalo , Angiopatía Amiloide Cerebral , Humanos , Encéfalo/metabolismo , Encéfalo/patología , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Animales , Péptidos beta-Amiloides/metabolismo , Sistema Glinfático/metabolismo , Sistema Glinfático/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología
19.
Bioessays ; 45(3): e2200121, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36707486

RESUMEN

The behavior of somatic stem cells is regulated by their niche. Interaction between hematopoietic stem cells (HSCs) and their niches are a representative model to understand stem cell-niche interplay. Here, we provide an overview of crosstalk between HSCs and their niches in bone marrow and extramedullary organs following the life journey of HSCs from emergence, development, maturation until aging. We highlight the unique differences of HSC niches in different life stages within various organs focusing on recent literature to propose new speculations and hypotheses.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Reacciones Cruzadas , Nicho de Células Madre
20.
Proc Natl Acad Sci U S A ; 119(17): e2120439119, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35412862

RESUMEN

Long-duration spaceflight induces changes to the brain and cerebrospinal fluid compartments and visual acuity problems known as spaceflight-associated neuro-ocular syndrome (SANS). The clinical relevance of these changes and whether they equally affect crews of different space agencies remain unknown. We used MRI to analyze the alterations occurring in the perivascular spaces (PVS) in NASA and European Space Agency astronauts and Roscosmos cosmonauts after a 6-mo spaceflight on the International Space Station (ISS). We found increased volume of basal ganglia PVS and white matter PVS (WM-PVS) after spaceflight, which was more prominent in the NASA crew than the Roscosmos crew. Moreover, both crews demonstrated a similar degree of lateral ventricle enlargement and decreased subarachnoid space at the vertex, which was correlated with WM-PVS enlargement. As all crews experienced the same environment aboard the ISS, the differences in WM-PVS enlargement may have been due to, among other factors, differences in the use of countermeasures and high-resistive exercise regimes, which can influence brain fluid redistribution. Moreover, NASA astronauts who developed SANS had greater pre- and postflight WM-PVS volumes than those unaffected. These results provide evidence for a potential link between WM-PVS fluid and SANS.


Asunto(s)
Astronautas , Líquido Cefalorraquídeo , Sistema Glinfático , Vuelo Espacial , Trastornos de la Visión , Líquido Cefalorraquídeo/diagnóstico por imagen , Sistema Glinfático/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Trastornos de la Visión/líquido cefalorraquídeo , Trastornos de la Visión/diagnóstico por imagen , Sustancia Blanca/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA