Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.263
Filtrar
1.
Mol Immunol ; 174: 41-46, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39182279

RESUMEN

Colony stimulating factor 1 receptor (CSF1R) is an essential receptor for both colony stimulating factor 1 (CSF1) and interleukin (IL) 34 signaling expressed on monocyte precursors and myeloid cells, including monocytes, dendritic cells (DC), and microglia. In humans, dominant heterozygous pathogenic variants in CSF1R cause a neurological condition known as CSF1R-related disorder (CSF1R-RD), typically with late onset, previously referred to as adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). CSF1R-RD is characterized by microglia reduction and altered monocyte function; however, the impact of pathogenic CSF1R variants on the human DC lineage remains largely unknown. We previously reported that cord blood CD34+ stem cell-derived DCs generated in vitro originate specifically from CSF1R expressing precursors. In this study, we examined the DC lineage of four unrelated patients with late-onset CSF1R-RD who carried heterozygous missense CSF1R variants (c.2330G>A, c.2375C>A, c.2329C>T, and c.2381T>C) affecting different amino acids in the protein tyrosine kinase domain of CSF1R. CD34+ stem cells and CD14+ monocytes were isolated from peripheral blood and subjected to an in vitro culture protocol to differentiate towards conventional DCs and monocyte-derived DCs, respectively. Flow cytometric analysis revealed that monocytes from patients with late-onset CSF1R-RD were still able to differentiate into monocyte-derived DCs in vitro, whereas the ability of CD34+ stem cells to differentiate into conventional DCs was impaired. Strikingly, the peripheral blood of patients contained all naturally occurring DC subsets. We conclude that the in vitro abrogation of DC-development in patients with heterozygous pathogenic missense CSF1R variants does not translate to an impairment in DC development in vivo and speculate that CSF1R signalling in vivo is compensated, which needs further study.


Asunto(s)
Antígenos CD34 , Diferenciación Celular , Células Dendríticas , Mutación Missense , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Células Dendríticas/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Antígenos CD34/metabolismo , Diferenciación Celular/genética , Femenino , Masculino , Persona de Mediana Edad , Heterocigoto , Adulto , Anciano , Monocitos/metabolismo , Células Cultivadas , Receptor de Factor Estimulante de Colonias de Macrófagos
2.
Acta Neuropathol Commun ; 12(1): 139, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39217398

RESUMEN

CSF1R-related disorder (CSF1R-RD) is a neurodegenerative condition that predominantly affects white matter due to genetic alterations in the CSF1R gene, which is expressed by microglia. We studied an elderly man with a hereditary, progressive dementing disorder of unclear etiology. Standard genetic testing for leukodystrophy and other neurodegenerative conditions was negative. Brain autopsy revealed classic features of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), including confluent white matter degeneration with axonal spheroids and pigmented glial cells in the affected white matter, consistent with CSF1R-RD. Subsequent long-read sequencing identified a novel deletion in CSF1R that was not detectable with short-read exome sequencing. To gain insight into potential mechanisms underlying white matter degeneration in CSF1R-RD, we studied multiple brain regions exhibiting varying degrees of white matter pathology. We found decreased CSF1R transcript and protein across brain regions, including intact white matter. Single nuclear RNA sequencing (snRNAseq) identified two disease-associated microglial cell states: lipid-laden microglia (expressing GPNMB, ATG7, LGALS1, LGALS3) and inflammatory microglia (expressing IL2RA, ATP2C1, FCGBP, VSIR, SESN3), along with a small population of CD44+ peripheral monocyte-derived macrophages exhibiting migratory and phagocytic signatures. GPNMB+ lipid-laden microglia with ameboid morphology represented the end-stage disease microglia state. Disease-associated oligodendrocytes exhibited cell stress signatures and dysregulated apoptosis-related genes. Disease-associated oligodendrocyte precursor cells (OPCs) displayed a failure in their differentiation into mature myelin-forming oligodendrocytes, as evidenced by upregulated LRP1, PDGFRA, SOX5, NFIA, and downregulated NKX2-2, NKX6.2, SOX4, SOX8, TCF7L2, YY1, ZNF488. Overall, our findings highlight microglia-oligodendroglia crosstalk in demyelination, with CSF1R dysfunction promoting phagocytic and inflammatory microglia states, an arrest in OPC differentiation, and oligodendrocyte depletion.


Asunto(s)
Neuroglía , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Masculino , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neuroglía/patología , Neuroglía/metabolismo , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Leucoencefalopatías/metabolismo , Anciano , Microglía/patología , Microglía/metabolismo , Perfilación de la Expresión Génica , Transcriptoma , Sustancia Blanca/patología , Sustancia Blanca/metabolismo , Encéfalo/patología , Encéfalo/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos
3.
Neuron ; 112(17): 2910-2921.e7, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39142282

RESUMEN

Microglia, the resident immune cells of the central nervous system, are intimately involved in the brain's most basic processes, from pruning neural synapses during development to preventing excessive neuronal activity throughout life. Studies have reported both helpful and harmful roles for microglia at the blood-brain barrier (BBB) in the context of disease. However, less is known about microglia-endothelial cell interactions in the healthy brain. To investigate the role of microglia at a healthy BBB, we used the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 to deplete microglia and analyzed the BBB ultrastructure, permeability, and transcriptome. Interestingly, we found that, despite their direct contact with endothelial cells, microglia are not necessary for the maintenance of BBB structure, function, or gene expression in the healthy brain. However, we found that PLX5622 treatment alters brain endothelial cholesterol metabolism. This effect was independent from microglial depletion, suggesting that PLX5622 has off-target effects on brain vasculature.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Colesterol , Células Endoteliales , Microglía , Microglía/metabolismo , Microglía/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Animales , Colesterol/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Ratones , Encéfalo/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Ratones Endogámicos C57BL , Masculino , Compuestos Orgánicos
4.
Nat Commun ; 15(1): 7097, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39154007

RESUMEN

Converging evidence indicates that extra-embryonic yolk sac is the source of both macrophages and endothelial cells in adult mouse tissues. Prevailing views are that these embryonically derived cells are maintained after birth by proliferative self-renewal in their differentiated states. Here we identify clonogenic endothelial-macrophage (EndoMac) progenitor cells in the adventitia of embryonic and postnatal mouse aorta, that are independent of Flt3-mediated bone marrow hematopoiesis and derive from an early embryonic CX3CR1+ and CSF1R+ source. These bipotent progenitors are proliferative and vasculogenic, contributing to adventitial neovascularization and formation of perfused blood vessels after transfer into ischemic tissue. We establish a regulatory role for angiotensin II, which enhances their clonogenic and differentiation properties and rapidly stimulates their proliferative expansion in vivo. Our findings demonstrate that embryonically derived EndoMac progenitors participate in local vasculogenic responses in the aortic wall by contributing to the expansion of endothelial cells and macrophages postnatally.


Asunto(s)
Aorta , Macrófagos , Animales , Macrófagos/citología , Macrófagos/metabolismo , Aorta/citología , Ratones , Receptor 1 de Quimiocinas CX3C/metabolismo , Receptor 1 de Quimiocinas CX3C/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Diferenciación Celular , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Angiotensina II , Proliferación Celular , Células Madre/citología , Células Madre/metabolismo , Ratones Endogámicos C57BL , Femenino , Neovascularización Fisiológica , Receptores de Quimiocina/metabolismo , Receptores de Quimiocina/genética , Masculino , Hematopoyesis/fisiología , Tirosina Quinasa 3 Similar a fms
5.
Transl Psychiatry ; 14(1): 338, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39179543

RESUMEN

Microglia, traditionally regarded as innate immune cells in the brain, drive neuroinflammation and synaptic dysfunctions in the early phases of Alzheimer disease (AD), acting upstream to Aß accumulation. Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its levels are significantly increased in neurodegenerative diseases, possibly contributing to the chronic inflammatory microglial response. On the other hand, CSF-1R inhibitors confer neuroprotection in preclinical models of neurodegenerative diseases. Here, we determined the effects of the CSF-1R inhibitor PLX3397 on the Aß-mediated synaptic alterations in ex vivo hippocampal slices. Electrophysiological findings show that PLX3397 rescues LTP impairment and neurotransmission changes induced by Aß. In addition, using confocal imaging experiments, we demonstrate that PLX3397 stimulates a microglial transition toward a phagocytic phenotype, which in turn promotes the clearance of Aß from glutamatergic terminals. We believe that the selective pruning of Aß-loaded synaptic terminals might contribute to the restoration of LTP and excitatory transmission alterations observed upon acute PLX3397 treatment. This result is in accordance with the mechanism proposed for CSF1R inhibitors, that is to eliminate responsive microglia and replace it with newly generated, homeostatic microglia, capable of promoting brain repair. Overall, our findings identify a connection between the rapid microglia adjustments and the early synaptic alterations observed in AD, possibly highlighting a novel disease-modifying target.


Asunto(s)
Aminopiridinas , Péptidos beta-Amiloides , Hipocampo , Potenciación a Largo Plazo , Microglía , Animales , Microglía/efectos de los fármacos , Microglía/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Péptidos beta-Amiloides/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aminopiridinas/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Pirroles/farmacología , Ratones , Fagocitosis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Ratones Endogámicos C57BL , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Ácido Glutámico/metabolismo
6.
Mol Biol Rep ; 51(1): 920, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39158794

RESUMEN

BACKGROUND: Liaoning cashmere goat is recognized as a valuable genetic resource breed, with restrictions on genetic outflow in China. Hair follicle development in the cashmere goat is influenced by melatonin and long non-coding RNAs (lncRNAs). However, the role of lncRNAs in facilitating melatonin-promoted cashmere growth remains poorly understood. Previous studies have identified a new lncRNA, lncRNA018392, which is involved in the melatonin-promoted proliferation of cashmere skin fibroblasts. METHOD: Flow cytometry and CCK-8 assays confirmed that silencing lncRNA018392 negates the effects of melatonin on cell proliferation, and that proliferation was reduced when the gene CSF1R, located near lncRNA018392, was inhibited. Further investigation using a dual-luciferase reporter assay showed that lncRNA018392 could positively regulate the promoter of CSF1R. RESULTS: Results from RNA-binding protein immunoprecipitation (RIP) and chromatin immunoprecipitation sequencing (ChIP-Seq) revealed that lncRNA018392 interacts with the transcription factor SPI1, with CSF1R being a downstream target gene regulated by SPI1. This interaction was confirmed by ChIP-PCR, which demonstrated SPI1's binding to CSF1R. CONCLUSIONS: This study found that the melatonin-responsive lncRNA018392 accelerates the cell cycle and promotes cell proliferation by recruiting SPI1 to upregulate the expression of the neighboring gene CSF1R. These findings provide a theoretical foundation for elucidating the molecular mechanisms of cashmere growth and for the molecular breeding of cashmere goats.


Asunto(s)
Proliferación Celular , Fibroblastos , Cabras , Melatonina , ARN Largo no Codificante , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cabras/genética , Fibroblastos/metabolismo , Proliferación Celular/genética , Melatonina/farmacología , Melatonina/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Piel/metabolismo , Piel/citología , Regulación hacia Arriba/genética , Regulación hacia Arriba/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Folículo Piloso/metabolismo , Transactivadores
7.
Eur Respir Rev ; 33(173)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39142709

RESUMEN

Pulmonary alveolar proteinosis (PAP) is a syndrome that results from the accumulation of lipoproteinaceous material in the alveolar space. According to the underlying pathogenetic mechanisms, three different forms have been identified, namely primary, secondary and congenital. Primary PAP is caused by disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling due to the presence of neutralising autoantibodies (autoimmune PAP) or GM-CSF receptor genetic defects (hereditary PAP), which results in dysfunctional alveolar macrophages with reduced phagocytic clearance of particles, cholesterol and surfactant. The serum level of GM-CSF autoantibody is the only disease-specific biomarker of autoimmune PAP, although it does not correlate with disease severity. In PAP patients with normal serum GM-CSF autoantibody levels, elevated serum GM-CSF levels is highly suspicious for hereditary PAP. Several biomarkers have been correlated with disease severity, although they are not specific for PAP. These include lactate dehydrogenase, cytokeratin 19 fragment 21.1, carcinoembryonic antigen, neuron-specific enolase, surfactant proteins, Krebs von Lungen 6, chitinase-3-like protein 1 and monocyte chemotactic proteins. Finally, increased awareness of the disease mechanisms has led to the development of pathogenesis-based treatments, such as GM-CSF augmentation and cholesterol-targeting therapies.


Asunto(s)
Autoanticuerpos , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Proteinosis Alveolar Pulmonar , Proteinosis Alveolar Pulmonar/terapia , Proteinosis Alveolar Pulmonar/diagnóstico , Proteinosis Alveolar Pulmonar/inmunología , Humanos , Autoanticuerpos/sangre , Resultado del Tratamiento , Biomarcadores/sangre , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Predisposición Genética a la Enfermedad , Animales , Transducción de Señal , Pulmón/inmunología
8.
J Neurol ; 271(9): 6025-6037, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39031193

RESUMEN

The CSF1R gene, located on chromosome 5, encodes a 108 kDa protein and plays a critical role in regulating myeloid cell function. Mutations in CSF1R have been identified as a cause of a rare white matter disease called adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP, also known as CSF1R-related leukoencephalopathy), characterized by progressive neurological dysfunction. This study aimed to broaden the genetic basis of ALSP by identifying novel CSF1R variants in patients with characteristic clinical and imaging features of ALSP. Genetic analysis was performed through whole-exome sequencing or panel analysis for leukodystrophy genes. Variant annotation and classification were conducted using computational tools, and the identified variants were categorized following the recommendations of the American College of Medical Genetics and Genomics (ACMG). To assess the evolutionary conservation of the novel variants within the CSF1R protein, amino acid sequences were compared across different species. The study identified six previously unreported CSF1R variants (c.2384G>T, c.2133_2919del, c.1837G>A, c.2304C>A, c.2517G>T, c.2642C>T) in seven patients with ALSP, contributing to the expanding knowledge of the genetic diversity underlying this rare disease. The analysis revealed considerable genetic and clinical heterogeneity among these patients. The findings emphasize the need for a comprehensive understanding of the genetic basis of rare diseases like ALSP and underscored the importance of genetic testing, even in cases with no family history of the disease. The study's contribution to the growing spectrum of ALSP genetics and phenotypes enhances our knowledge of this condition, which can be crucial for both diagnosis and potential future treatments.


Asunto(s)
Leucoencefalopatías , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Femenino , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Masculino , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Adulto , Persona de Mediana Edad , Mutación , Neuroglía/patología , Anciano , Receptor de Factor Estimulante de Colonias de Macrófagos
10.
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
11.
Clin Cancer Res ; 30(18): 3996-4004, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-38995311

RESUMEN

PURPOSE: Tenosynovial giant cell tumor (TGCT) is a locally aggressive neoplasm caused by dysregulation of the colony-stimulating factor 1 (CSF1) gene and overexpression of the CSF1 ligand. Surgery is the standard of care for most patients, but there are limited treatment options for patients with TGCT not amenable to surgery. This study evaluates vimseltinib, an investigational, oral, switch-control tyrosine kinase inhibitor designed to selectively and potently inhibit the CSF1 receptor. PATIENTS AND METHODS: This first-in-human, multicenter, open-label phase I/II study of vimseltinib in patients with malignant solid tumors (N = 37) or TGCT not amenable to surgery (N = 32) followed a pharmacologically guided 3 + 3 study design (NCT03069469). The primary objectives were to assess safety and tolerability, determine the recommended phase II dose, and characterize the pharmacokinetics; exploratory objectives included pharmacodynamics and efficacy. RESULTS: Vimseltinib was well tolerated; the majority of non-laboratory treatment-emergent adverse events were of grade 1/2 severity. There was no evidence of cholestatic hepatotoxicity or drug-induced liver injury. The recommended phase II dose was determined to be 30 mg twice weekly (no loading dose), and vimseltinib plasma exposure increased with the dose. In patients with TGCT, the median treatment duration was 25.1 months (range, 0.7-46.9), and the objective response rate as assessed by independent radiological review using RECIST version 1.1 was 72%. CONCLUSIONS: Vimseltinib demonstrated long-term tolerability, manageable safety, dose-dependent exposure, and robust antitumor activity in patients with TGCT not amenable to surgery.


Asunto(s)
Tumor de Células Gigantes de las Vainas Tendinosas , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Masculino , Femenino , Persona de Mediana Edad , Adulto , Anciano , Tumor de Células Gigantes de las Vainas Tendinosas/tratamiento farmacológico , Tumor de Células Gigantes de las Vainas Tendinosas/patología , Tumor de Células Gigantes de las Vainas Tendinosas/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Resultado del Tratamiento , Adulto Joven , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/administración & dosificación , Anilidas , Quinolinas , Receptor de Factor Estimulante de Colonias de Macrófagos
12.
J Neuroinflammation ; 21(1): 168, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961498

RESUMEN

BACKGROUND: The eye is a highly specialized sensory organ which encompasses the retina as a part of the central nervous system, but also non-neural compartments such as the transparent vitreous body ensuring stability of the eye globe and a clear optical axis. Hyalocytes are the tissue-resident macrophages of the vitreous body and are considered to play pivotal roles in health and diseases of the vitreoretinal interface, such as proliferative vitreoretinopathy or diabetic retinopathy. However, in contrast to other ocular macrophages, their embryonic origin as well as the extent to which these myeloid cells might be replenished by circulating monocytes remains elusive. RESULTS: In this study, we combine transgenic reporter mice, embryonic and adult fate mapping approaches as well as parabiosis experiments with multicolor immunofluorescence labeling and confocal laser-scanning microscopy to comprehensively characterize the murine hyalocyte population throughout development and in adulthood. We found that murine hyalocytes express numerous well-known myeloid cell markers, but concomitantly display a distinct immunophenotype that sets them apart from retinal microglia. Embryonic pulse labeling revealed a yolk sac-derived origin of murine hyalocytes, whose precursors seed the developing eye prenatally. Finally, postnatal labeling and parabiosis established the longevity of hyalocytes which rely on Colony Stimulating Factor 1 Receptor (CSF1R) signaling for their maintenance, independent of blood-derived monocytes. CONCLUSION: Our study identifies hyalocytes as long-living progeny of the yolk sac hematopoiesis and highlights their role as integral members of the innate immune system of the eye. As a consequence of their longevity, immunosenescence processes may culminate in hyalocyte dysfunction, thereby contributing to the development of vitreoretinal diseases. Therefore, myeloid cell-targeted therapies that convey their effects through the modification of hyalocyte properties may represent an interesting approach to alleviate the burden imposed by diseases of the vitreoretinal interface.


Asunto(s)
Macrófagos , Ratones Transgénicos , Cuerpo Vítreo , Saco Vitelino , Animales , Ratones , Cuerpo Vítreo/citología , Saco Vitelino/citología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Animales Recién Nacidos
14.
J Nanobiotechnology ; 22(1): 409, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38992688

RESUMEN

OBJECTIVE: This study aimed to investigate the critical role of MDSCs in CRC immune suppression, focusing on the CSF1R and JAK/STAT3 signaling axis. Additionally, it assessed the therapeutic efficacy of LNCs@CSF1R siRNA and anti-PD-1 in combination. METHODS: Single-cell transcriptome sequencing data from CRC and adjacent normal tissues identified MDSC-related differentially expressed genes. RNA-seq analysis comprehensively profiled MDSC gene expression in murine CRC tumors. LNCs@CSF1R siRNA nanocarriers effectively targeted and inhibited CSF1R. Flow cytometry quantified changes in MDSC surface markers post-CSF1R inhibition. RNA-seq and pathway enrichment analyses revealed the impact of CSF1R on MDSC metabolism and signaling. The effect of CSF1R inhibition on the JAK/STAT3 signaling axis was validated using Colivelin and metabolic assessments. Glucose and fatty acid uptake were measured via fluorescence-based flow cytometry. The efficacy of LNCs@CSF1R siRNA and anti-PD-1, alone and in combination, was evaluated in a murine CRC model with extensive tumor section analyses. RESULTS: CSF1R played a significant role in MDSC-mediated immune suppression. LNCs@CSF1R siRNA nanocarriers effectively targeted MDSCs and inhibited CSF1R. CSF1R regulated MDSC fatty acid metabolism and immune suppression through the JAK/STAT3 signaling axis. Inhibition of CSF1R reduced STAT3 activation and target gene expression, which was rescued by Colivelin. Combined treatment with LNCs@CSF1R siRNA and anti-PD-1 significantly slowed tumor growth and reduced MDSC abundance within CRC tumors. CONCLUSION: CSF1R via the JAK/STAT3 axis critically regulates MDSCs, particularly in fatty acid metabolism and immune suppression. Combined therapy with LNCs@CSF1R siRNA and anti-PD-1 enhances therapeutic efficacy in a murine CRC model, providing a strong foundation for future clinical applications.


Asunto(s)
Neoplasias Colorrectales , Células Supresoras de Origen Mieloide , ARN Interferente Pequeño , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Factor de Transcripción STAT3 , Animales , Células Supresoras de Origen Mieloide/metabolismo , Ratones , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor de Transcripción STAT3/metabolismo , Línea Celular Tumoral , Humanos , Transducción de Señal/efectos de los fármacos , Receptor de Muerte Celular Programada 1/metabolismo , Femenino , Ratones Endogámicos BALB C , Quinasas Janus/metabolismo , Inmunomodulación/efectos de los fármacos , Receptor de Factor Estimulante de Colonias de Macrófagos
15.
Nat Commun ; 15(1): 5654, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969669

RESUMEN

Hematopoietic stem cell transplantation can deliver therapeutic proteins to the central nervous system (CNS) through transplant-derived microglia-like cells. However, current conditioning approaches result in low and slow engraftment of transplanted cells in the CNS. Here we optimized a brain conditioning regimen that leads to rapid, robust, and persistent microglia replacement without adverse effects on neurobehavior or hematopoiesis. This regimen combines busulfan myeloablation and six days of Colony-stimulating factor 1 receptor inhibitor PLX3397. Single-cell analyses revealed unappreciated heterogeneity of microglia-like cells with most cells expressing genes characteristic of homeostatic microglia, brain-border-associated macrophages, and unique markers. Cytokine analysis in the CNS showed transient inductions of myeloproliferative and chemoattractant cytokines that help repopulate the microglia niche. Bone marrow transplant of progranulin-deficient mice conditioned with busulfan and PLX3397 restored progranulin in the brain and eyes and normalized brain lipofuscin storage, proteostasis, and lipid metabolism. This study advances our understanding of CNS repopulation by hematopoietic-derived cells and demonstrates its therapeutic potential for treating progranulin-dependent neurodegeneration.


Asunto(s)
Busulfano , Microglía , Progranulinas , Animales , Microglía/metabolismo , Microglía/efectos de los fármacos , Progranulinas/metabolismo , Progranulinas/genética , Ratones , Busulfano/farmacología , Trasplante de Células Madre Hematopoyéticas , Aminopiridinas/farmacología , Encéfalo/metabolismo , Pirroles/farmacología , Ratones Endogámicos C57BL , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Trasplante de Médula Ósea , Masculino , Sistema Nervioso Central/metabolismo , Ratones Noqueados , Acondicionamiento Pretrasplante/métodos , Análisis de la Célula Individual , Citocinas/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores
16.
Neuron ; 112(16): 2686-2707.e8, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-38897209

RESUMEN

Microglia replacement strategies are increasingly being considered for the treatment of primary microgliopathies like adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). However, available mouse models fail to recapitulate the diverse neuropathologies and reduced microglia numbers observed in patients. In this study, we generated a xenotolerant mouse model lacking the fms-intronic regulatory element (FIRE) enhancer within Csf1r, which develops nearly all the hallmark pathologies associated with ALSP. Remarkably, transplantation of human induced pluripotent stem cell (iPSC)-derived microglial (iMG) progenitors restores a homeostatic microglial signature and prevents the development of axonal spheroids, white matter abnormalities, reactive astrocytosis, and brain calcifications. Furthermore, transplantation of CRISPR-corrected ALSP-patient-derived iMG reverses pre-existing spheroids, astrogliosis, and calcification pathologies. Together with the accompanying study by Munro and colleagues, our results demonstrate the utility of FIRE mice to model ALSP and provide compelling evidence that iMG transplantation could offer a promising new therapeutic strategy for ALSP and perhaps other microglia-associated neurological disorders.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas , Leucoencefalopatías , Microglía , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Animales , Microglía/metabolismo , Leucoencefalopatías/genética , Humanos , Ratones , Células Madre Pluripotentes Inducidas/trasplante , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Gliosis , Receptor de Factor Estimulante de Colonias de Macrófagos
17.
CNS Neurosci Ther ; 30(6): e14815, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38922778

RESUMEN

AIMS: Colony stimulating factor 1 receptor (CSF1R)-related leukoencephalopathy is a rapidly progressing neurodegenerative disease caused by CSF1R gene mutations. This study aimed to identify and investigate the effect of a novel intronic mutation (c.1754-3C>G) of CSF1R on splicing. METHODS: A novel intronic mutation was identified using whole-exome sequencing. To investigate the impact of this mutation, we employed various bioinformatics tools to analyze the transcription of the CSF1R gene and the three-dimensional structure of its encoded protein. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) was performed to validate the findings. RESULTS: A novel mutation (c.1754-3C>G) in CSF1R was identified, which results in exon 13 skipping due to the disruption of the 3' splice site consensus sequence NYAG/G. This exon skipping event was further validated in the peripheral blood of the mutation carrier through RT-PCR and Sanger sequencing. Protein structure prediction indicated a disruption in the tyrosine kinase domain, with the truncated protein showing significant structural alterations. CONCLUSIONS: Our findings underscore the importance of intronic mis-splicing mutations in the diagnosis and management of CSF1R-related leukoencephalopathy.


Asunto(s)
Intrones , Leucoencefalopatías , Mutación , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Leucoencefalopatías/genética , Mutación/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Intrones/genética , Femenino , Masculino , Adulto , Empalme del ARN/genética , Receptor de Factor Estimulante de Colonias de Macrófagos
18.
Cell Rep ; 43(6): 114352, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38870011

RESUMEN

Addressing the mononuclear phagocyte system (MPS) and macrophage M1/M2 activation is important in diagnosing hematological disorders and inflammatory pathologies and designing therapeutic tools. CSF1R is a reliable marker to identify all circulating MPS cells and tissue macrophages in humans using a single surface protein. CSF1R permits the quantification and isolation of monocyte and dendritic cell (DC) subsets in conjunction with CD14, CD16, and CD1c and is stable across the lifespan and sexes in the absence of overt pathology. Beyond cell detection, measuring M1/M2 activation in humans poses challenges due to response heterogeneity, transient signaling, and multiple regulation steps for transcripts and proteins. MPS cells respond in a conserved manner to M1/M2 pathways such as interleukin-4 (IL-4), steroids, interferon-γ (IFNγ), and lipopolysaccharide (LPS), for which we propose an ad hoc modular gene expression tool. Signature analysis highlights macrophage activation mosaicism in experimental samples, an emerging concept that points to mixed macrophage activation states in pathology.


Asunto(s)
Activación de Macrófagos , Macrófagos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Activación de Macrófagos/genética , Macrófagos/metabolismo , Macrófagos/inmunología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Interferón gamma/metabolismo , Lipopolisacáridos/farmacología , Femenino , Mosaicismo , Masculino , Monocitos/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Interleucina-4/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/inmunología , Receptores de IgG/metabolismo , Receptores de IgG/genética , Antígenos CD1/metabolismo , Antígenos CD1/genética , Sistema Mononuclear Fagocítico/metabolismo , Glicoproteínas , Receptor de Factor Estimulante de Colonias de Macrófagos
19.
Neuron ; 112(16): 2732-2748.e8, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-38897208

RESUMEN

Microglia are brain-resident macrophages that contribute to central nervous system (CNS) development, maturation, and preservation. Here, we examine the consequences of permanent microglial deficiencies on brain aging using the Csf1rΔFIRE/ΔFIRE mouse model. In juvenile Csf1rΔFIRE/ΔFIRE mice, we show that microglia are dispensable for the transcriptomic maturation of other brain cell types. By contrast, with advancing age, pathologies accumulate in Csf1rΔFIRE/ΔFIRE brains, macroglia become increasingly dysregulated, and white matter integrity declines, mimicking many pathological features of human CSF1R-related leukoencephalopathy. The thalamus is particularly vulnerable to neuropathological changes in the absence of microglia, with atrophy, neuron loss, vascular alterations, macroglial dysregulation, and severe tissue calcification. We show that populating Csf1rΔFIRE/ΔFIRE brains with wild-type microglia protects against many of these pathological changes. Together with the accompanying study by Chadarevian and colleagues1, our results indicate that the lifelong absence of microglia results in an age-related neurodegenerative condition that can be counteracted via transplantation of healthy microglia.


Asunto(s)
Envejecimiento , Encéfalo , Microglía , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Microglía/patología , Microglía/metabolismo , Animales , Ratones , Envejecimiento/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Encéfalo/patología , Ratones Endogámicos C57BL , Masculino , Sustancia Blanca/patología , Leucoencefalopatías/patología , Tálamo/patología
20.
Mol Pharm ; 21(8): 3979-3991, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38935927

RESUMEN

Colony-stimulating factor 1 receptor (CSF1R) is a type III receptor tyrosine kinase that is crucial for immune cell activation, survival, proliferation, and differentiation. Its expression significantly increases in macrophages during inflammation, playing a crucial role in regulating inflammation resolution and termination. Consequently, CSF1R has emerged as a critical target for both therapeutic intervention and imaging of inflammatory diseases. Herein, we have developed a radiotracer, 1-[4-((7-(dimethylamino)quinazolin-4-yl)oxy)phenyl]-3-(4-[18F]fluorophenyl)urea ([18F]17), for in vivo positron emission tomography (PET) imaging of CSF1R. Compound 17 exhibits a comparable inhibitory potency against CSF1R as the well-known CSF1R inhibitor PLX647. The radiosynthesis of [18F]17 was successfully performed by radiofluorination of aryltrimethyltin precursor with a yield of approximately 12% at the end of synthesis, maintaining a purity exceeding 98%. In vivo stability and biodistribution studies demonstrate that [18F]17 remains >90% intact at 30 min postinjection, with no defluorination observed even at 60 min postinjection. The PET/CT imaging study in lipopolysaccharide-induced pulmonary inflammation mice indicates that [18F]17 offers a more sensitive characterization of pulmonary inflammation compared to traditional [18F]FDG. Notably, [18F]17 shows a higher discrepancy in uptake ratio between mice with pulmonary inflammation and the sham group. Furthermore, the variations in [18F]17 uptake ratio observed on day 7 and day 14 correspond to lung density changes observed in CT imaging. Moreover, the expression levels of CSF1R on day 7 and day 14 follow a trend similar to the uptake pattern of [18F]17, indicating its potential for accurately characterizing CSF1R expression levels and effectively monitoring the pulmonary inflammation progression. These results strongly suggest that [18F]17 has promising prospects as a CSF1R PET tracer, providing diagnostic opportunities for pulmonary inflammatory diseases.


Asunto(s)
Neumonía , Tomografía de Emisión de Positrones , Radiofármacos , Animales , Ratones , Neumonía/diagnóstico por imagen , Neumonía/metabolismo , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Distribución Tisular , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Radioisótopos de Flúor , Humanos , Masculino , Ratones Endogámicos C57BL , Pulmón/diagnóstico por imagen , Pulmón/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA