Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
1.
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
2.
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
3.
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
4.
Int Immunopharmacol ; 138: 112473, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38943977

RESUMEN

As the resident immune cells in the central nervous system, microglia exhibit a 'sensitized' or 'primed' phenotype with dystrophic morphology and dysregulated functions in aged brains. Although studies have demonstrated the inflammatory profile of aged microglia in several neurological diseases, this issue is largely uncertain in stroke. Consequently, this study investigated the effects of primed and repopulated microglia on post-ischemic brain injury in aged mice. We replaced primed microglia with newly repopulated microglia through pharmacological administration and withdrawal of the colony-stimulating factor 1 receptor (CSF1R) inhibitor, PLX3397. Further, we performed a series of behavioral tests and flow cytometry in mouse models of middle cerebral artery occlusion (MCAO) to study the effects of microglial replacement on ischemic injury in the aged brain. With depletion and subsequent repopulation of microglia in MCAO mice, microglial replacement in aged mice improved neurological function and decreased brain infarction. This protective effect was accompanied by the reduction of peripheral immune cells infiltrating into brains. We showed that the repopulated microglia expressed elevated neuroprotective factors (including Cluster of Differentiation 206, transforming growth factor-ß, and interleukin-10) and diminished expression of inflammatory markers (including Cluster of Differentiation 86, interleukin-6, and tumor necrosis factor α). Moreover, microglial replacement protected the blood-brain barrier and relieved neuronal death in aged mice subjected to 60 min of MCAO. These results imply that the replacement of microglia in the aged brain may alleviate brain damage and neuroinflammation, and therefore, ischemic brain damage. Thus, targeting microglia could be a promising therapeutic strategy for ischemic stroke.


Asunto(s)
Envejecimiento , Infarto de la Arteria Cerebral Media , Ratones Endogámicos C57BL , Microglía , Enfermedades Neuroinflamatorias , Fármacos Neuroprotectores , Animales , Microglía/efectos de los fármacos , Microglía/inmunología , Masculino , Ratones , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/farmacología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/patología , Aminopiridinas/farmacología , Aminopiridinas/uso terapéutico , Pirroles/farmacología , Pirroles/uso terapéutico , Isquemia Encefálica/tratamiento farmacológico , Encéfalo/patología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Modelos Animales de Enfermedad , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Barrera Hematoencefálica/efectos de los fármacos
5.
Front Immunol ; 15: 1366841, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38711521

RESUMEN

Introduction: Age-related macular degeneration (AMD) is a prevalent, chronic and progressive retinal degenerative disease characterized by an inflammatory response mediated by activated microglia accumulating in the retina. In this study, we demonstrate the therapeutically effects and the underlying mechanisms of microglial repopulation in the laser-induced choroidal neovascularization (CNV) model of exudative AMD. Methods: The CSF1R inhibitor PLX3397 was used to establish a treatment paradigm for microglial repopulation in the retina. Neovascular leakage and neovascular area were examined by fundus fluorescein angiography (FFA) and immunostaining of whole-mount RPE-choroid-sclera complexes in CNV mice receiving PLX3397. Altered cellular senescence was measured by beta-galactosidase (SA-ß-gal) activity and p16INK4a expression. The effect and mechanisms of repopulated microglia on leukocyte infiltration and the inflammatory response in CNV lesions were analyzed. Results: We showed that ten days of the CSF1R inhibitor PLX3397 treatment followed by 11 days of drug withdrawal was sufficient to stimulate rapid repopulation of the retina with new microglia. Microglial repopulation attenuated pathological choroid neovascularization and dampened cellular senescence in CNV lesions. Repopulating microglia exhibited lower levels of activation markers, enhanced phagocytic function and produced fewer cytokines involved in the immune response, thereby ameliorating leukocyte infiltration and attenuating the inflammatory response in CNV lesions. Discussion: The microglial repopulation described herein are therefore a promising strategy for restricting inflammation and choroidal neovascularization, which are important players in the pathophysiology of AMD.


Asunto(s)
Aminopiridinas , Neovascularización Coroidal , Modelos Animales de Enfermedad , Microglía , Animales , Neovascularización Coroidal/etiología , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Microglía/metabolismo , Microglía/efectos de los fármacos , Ratones , Aminopiridinas/farmacología , Aminopiridinas/uso terapéutico , Ratones Endogámicos C57BL , Degeneración Macular/patología , Degeneración Macular/metabolismo , Degeneración Macular/tratamiento farmacológico , Inflamación , 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 , Pirroles/uso terapéutico , Senescencia Celular/efectos de los fármacos
6.
Glia ; 72(8): 1484-1500, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38780213

RESUMEN

Microglia are innate immune cells in the brain and show exceptional heterogeneity. They are key players in brain physiological development regulating synaptic plasticity and shaping neuronal networks. In pathological disease states, microglia-induced synaptic pruning mediates synaptic loss and targeting microglia was proposed as a promising therapeutic strategy. However, the effect of microglia depletion and subsequent repopulation on dendritic spine density and neuronal function in the adult brain is largely unknown. In this study, we investigated whether pharmacological microglia depletion affects dendritic spine density after long-term permanent microglia depletion and after short-term microglia depletion with subsequent repopulation. Long-term microglia depletion using colony-stimulating-factor-1 receptor (CSF1-R) inhibitor PLX5622 resulted in increased overall spine density, especially of mushroom spines, and increased excitatory postsynaptic current amplitudes. Short-term PLX5622 treatment with subsequent repopulation of microglia had an opposite effect resulting in activated microglia with increased synaptic phagocytosis and consequently decreased spine density and reduced excitatory neurotransmission, while Barnes maze and elevated plus maze testing was unaffected. Moreover, RNA sequencing data of isolated repopulated microglia showed an activated and proinflammatory phenotype. Long-term microglia depletion might be a promising therapeutic strategy in neurological diseases with pathological microglial activation, synaptic pruning, and synapse loss. However, repopulation after depletion induces activated microglia and results in a decrease of dendritic spines possibly limiting the therapeutic application of microglia depletion. Instead, persistent modulation of pathological microglia activity might be beneficial in controlling synaptic damage.


Asunto(s)
Encéfalo , Espinas Dendríticas , Ratones Endogámicos C57BL , Microglía , Animales , Microglía/efectos de los fármacos , Microglía/metabolismo , Espinas Dendríticas/efectos de los fármacos , Masculino , Ratones , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Fagocitosis/fisiología , Fagocitosis/efectos de los fármacos , Plasticidad Neuronal/fisiología , Plasticidad Neuronal/efectos de los fármacos , Ratones Transgénicos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Compuestos Orgánicos
7.
Sci Rep ; 14(1): 12531, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822100

RESUMEN

Binding affinity is an important factor in drug design to improve drug-target selectivity and specificity. In this study, in silico techniques based on molecular docking followed by molecular dynamics (MD) simulations were utilized to identify the key residue(s) for CSF1R binding affinity among 14 pan-tyrosine kinase inhibitors and 15 CSF1R-specific inhibitors. We found tryptophan at position 550 (W550) on the CSF1R binding site interacted with the inhibitors' aromatic ring in a π-π way that made the ligands better at binding. Upon W550-Alanine substitution (W550A), the binding affinity of trans-(-)-kusunokinin and imatinib to CSF1R was significantly decreased. However, in terms of structural features, W550 did not significantly affect overall CSF1R structure, but provided destabilizing effect upon mutation. The W550A also did not either cause ligand to change its binding site or conformational changes due to ligand binding. As a result of our findings, the π-π interaction with W550's aromatic ring could be still the choice for increasing binding affinity to CSF1R. Nevertheless, our study showed that the increasing binding to W550 of the design ligand may not ensure CSF1R specificity and inhibition since W550-ligand bound state did not induce significantly conformational change into inactive state.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Triptófano , Triptófano/química , Triptófano/metabolismo , Ligandos , Sitios de Unión , Humanos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/química , Receptor de Factor Estimulante de Colonias de Macrófagos
8.
Alzheimers Res Ther ; 16(1): 101, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711159

RESUMEN

BACKGROUND: In Alzheimer's disease (AD), microglia surround extracellular plaques and mount a sustained inflammatory response, contributing to the pathogenesis of the disease. Identifying approaches to specifically target plaque-associated microglia (PAMs) without interfering in the homeostatic functions of non-plaque associated microglia would afford a powerful tool and potential therapeutic avenue. METHODS: Here, we demonstrated that a systemically administered nanomedicine, hydroxyl dendrimers (HDs), can cross the blood brain barrier and are preferentially taken up by PAMs in a mouse model of AD. As proof of principle, to demonstrate biological effects in PAM function, we treated the 5xFAD mouse model of amyloidosis for 4 weeks via systemic administration (ip, 2x weekly) of HDs conjugated to a colony stimulating factor-1 receptor (CSF1R) inhibitor (D-45113). RESULTS: Treatment resulted in significant reductions in amyloid-beta (Aß) and a stark reduction in the number of microglia and microglia-plaque association in the subiculum and somatosensory cortex, as well as a downregulation in microglial, inflammatory, and synaptic gene expression compared to vehicle treated 5xFAD mice. CONCLUSIONS: This study demonstrates that systemic administration of a dendranib may be utilized to target and modulate PAMs.


Asunto(s)
Enfermedad de Alzheimer , Dendrímeros , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía , Placa Amiloide , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Microglía/efectos de los fármacos , Microglía/metabolismo , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/patología , Ratones , Péptidos beta-Amiloides/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Humanos
9.
J Med Chem ; 67(8): 6854-6879, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38593344

RESUMEN

Blocking CSF-1/CSF-1R pathway has emerged as a promising strategy to remodel tumor immune microenvironment (TME) by reprogramming tumor-associated macrophages (TAMs). In this work, a novel CSF-1R inhibitor C19 with a highly improved pharmacokinetic profile and in vivo anticolorectal cancer (CRC) efficiency was successfully discovered. C19 could effectively reprogram M2-like TAMs to M1 phenotype and reshape the TME by inducing the recruitment of CD8+ T cells into tumors and reducing the infiltration of immunosuppressive Tregs/MDSCs. Deeper mechanistic studies revealed that C19 facilitated the infiltration of CD8+ T cells by enhancing the secretion of chemokine CXCL9, thus significantly potentiating the anti-CRC efficiency of PD-1 blockade. More importantly, C19 combined with PD-1 mAb could induce durable antitumor immune memory, effectively overcoming the recurrence of CRC. Taken together, our findings suggest that C19 is a promising therapeutic option for sensitizing CRC to anti-PD-1 therapy.


Asunto(s)
Neoplasias Colorrectales , Inmunoterapia , Receptor de Factor Estimulante de Colonias de Macrófagos , Neoplasias Colorrectales/tratamiento farmacológico , Animales , Humanos , Ratones , Inmunoterapia/métodos , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/síntesis química , Antineoplásicos/química , Microambiente Tumoral/efectos de los fármacos , Ratones Endogámicos BALB C , Línea Celular Tumoral , Femenino , Descubrimiento de Drogas , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Masculino , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/inmunología , Ratones Endogámicos C57BL , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología
10.
J Pharmacol Sci ; 155(2): 29-34, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677783

RESUMEN

Microglia are the residential immune cells in the central nervous system. Their roles as innate immune cells and regulators of synaptic remodeling are critical to the development and the maintenance of the brain. Numerous studies have depleted microglia to elucidate their involvement in healthy and pathological conditions. PLX3397, a blocker of colony stimulating factor 1 receptor (CSF1R), is widely used to deplete mouse microglia due to its non-invasiveness and convenience. Recently, other small rodents, including Syrian hamsters (Mesocricetus auratus) and Mongolian gerbils (Meriones unguiculatus), have been recognized as valuable animal models for studying brain functions and diseases. However, whether microglia depletion via PLX3397 is feasible in these species remains unclear. Here, we administered PLX3397 orally via food pellets to hamsters and gerbils. PLX3397 successfully depleted gerbil microglia but had no effect on microglial density in hamsters. Comparative analysis of the CSF1R amino acid sequence in different species hints that amino acid substitutions in the juxtamembrane domain may potentially contribute to the inefficacy of PLX3397 in hamsters.


Asunto(s)
Aminopiridinas , Encéfalo , Gerbillinae , Microglía , Pirroles , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Animales , Cricetinae , Administración Oral , Aminopiridinas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/citología , Mesocricetus , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Animales , Pirroles/farmacología , Pirrolidinas/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Especificidad de la Especie
11.
J Neuroinflammation ; 21(1): 108, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664840

RESUMEN

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by the presence of proteinaceous alpha-synuclein (α-syn) inclusions (Lewy bodies), markers of neuroinflammation and the progressive loss of nigrostriatal dopamine (DA) neurons. These pathological features can be recapitulated in vivo using the α-syn preformed fibril (PFF) model of synucleinopathy. We have previously determined that microglia proximal to PFF-induced nigral α-syn inclusions increase in soma size, upregulate major-histocompatibility complex-II (MHC-II) expression, and increase expression of a suite of inflammation-associated transcripts. This microglial response is observed months prior to degeneration, suggesting that microglia reacting to α-syn inclusion may contribute to neurodegeneration and could represent a potential target for novel therapeutics. The goal of this study was to determine whether colony stimulating factor-1 receptor (CSF1R)-mediated microglial depletion impacts the magnitude of α-syn aggregation, nigrostriatal degeneration, or the response of microglial in the context of the α-syn PFF model. METHODS: Male Fischer 344 rats were injected intrastriatally with either α-syn PFFs or saline. Rats were continuously administered Pexidartinib (PLX3397B, 600 mg/kg), a CSF1R inhibitor, to deplete microglia for a period of either 2 or 6 months. RESULTS: CSF1R inhibition resulted in significant depletion (~ 43%) of ionized calcium-binding adapter molecule 1 immunoreactive (Iba-1ir) microglia within the SNpc. However, CSF1R inhibition did not impact the increase in microglial number, soma size, number of MHC-II immunoreactive microglia or microglial expression of Cd74, Cxcl10, Rt-1a2, Grn, Csf1r, Tyrobp, and Fcer1g associated with phosphorylated α-syn (pSyn) nigral inclusions. Further, accumulation of pSyn and degeneration of nigral neurons was not impacted by CSF1R inhibition. Paradoxically, long term CSF1R inhibition resulted in increased soma size of remaining Iba-1ir microglia in both control and PFF rats, as well as expression of MHC-II in extranigral regions. CONCLUSIONS: Collectively, our results suggest that CSF1R inhibition does not impact the microglial response to nigral pSyn inclusions and that CSF1R inhibition is not a viable disease-modifying strategy for PD.


Asunto(s)
Microglía , Ratas Endogámicas F344 , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , alfa-Sinucleína , Animales , Microglía/metabolismo , Microglía/efectos de los fármacos , alfa-Sinucleína/metabolismo , Ratas , Masculino , 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 , Aminopiridinas/farmacología , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Sustancia Negra/metabolismo , Sustancia Negra/patología , Sustancia Negra/efectos de los fármacos , Modelos Animales de Enfermedad
12.
Int Immunopharmacol ; 133: 112071, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38636374

RESUMEN

Microglia play a pivotal role in the neuroinflammatory response after brain injury, and their proliferation is dependent on colony-stimulating factors. In the present study, we investigated the effect of inhibiting microglia proliferation on neurological damage post intracerebral hemorrhage (ICH) in a mouse model, an aspect that has never been studied before. Using a colony-stimulating factor-1 receptor antagonist (GW2580), we observed that inhibition of microglia proliferation significantly ameliorated neurobehavioral deficits, attenuated cerebral edema, and reduced hematoma volume after ICH. This intervention was associated with a decrease in pro-inflammatory factors in microglia and an increased infiltration of peripheral regulatory CD8 + CD122+ T cells into the injured brain tissue. The CXCR3/CXCL10 axis is the mechanism of brain homing of regulatory CD8 + CD122+ T cells, and the high expression of IL-10 is the hallmark of their synergistic anti-inflammatory effect with microglia. And activated astrocytes around the insult site are a prominent source of CXCL10. Thus, inhibition of microglial proliferation offers a new perspective for clinical translation. The cross-talk between multiple cells involved in the regulation of the inflammatory response highlights the comprehensive nature of neuroimmunomodulation.


Asunto(s)
Encéfalo , Linfocitos T CD8-positivos , Hemorragia Cerebral , Microglía , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Animales , Masculino , Ratones , Anisoles , Encéfalo/patología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/inmunología , Quimiocina CXCL10/metabolismo , Modelos Animales de Enfermedad , Interleucina-10/metabolismo , Subunidad beta del Receptor de Interleucina-2/metabolismo , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Pirimidinas , Receptores CXCR3/metabolismo , Receptores CXCR3/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
14.
Bioorg Med Chem ; 100: 117628, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38330850

RESUMEN

Although neuroinflammation is a significant pathogenic feature of many neurologic disorders, its precise function in-vivo is still not completely known. PET imaging enables the longitudinal examination, quantification, and tracking of different neuroinflammation biomarkers in living subjects. Particularly, PET imaging of Microglia, specialised dynamic immune cells crucial for maintaining brain homeostasis in central nervous system (CNS), is crucial for staging the neuroinflammation. Colony Stimulating Factor- 1 Receptor (CSF-1R) PET imaging is a novel method for the quantification of neuroinflammation. CSF-1R is mainly expressed on microglia, and neurodegenerative disorders greatly up-regulate its expression. The present review primarily focuses on the development, pros and cons of all the CSF-1R PET tracers reported for neuroinflammation imaging. Apart from neuroinflammation imaging, CSF-1R inhibitors are also reported for the therapy of neurodegenerative diseases such as Alzheimer's disease (AD). AD is a prevalent, advancing, and fatal neurodegenerative condition that have the characteristic feature of persistent neuroinflammation and primarily affects the elderly. The aetiology of AD is profoundly influenced by amyloid-beta (Aß) plaques, intracellular neurofibrillary tangles, and microglial dysfunction. Increasing evidence suggests that CSF-1R inhibitors (CSF-1Ri) can be helpful in preclinical models of neurodegenerative diseases. This review article also summarises the most recent developments of CSF-1Ri-based therapy for AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Anciano , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Factores Estimulantes de Colonias/metabolismo , Microglía/metabolismo , Enfermedades Neurodegenerativas/diagnóstico por imagen , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neuroinflamatorias , Tomografía de Emisión de Positrones/métodos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
15.
Eur J Ophthalmol ; 34(4): 1165-1173, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38099815

RESUMEN

OBJECTIVE: This study analyzed how high glucose affects CSF1R and p-ERK1/2 expression in RF/6A cells. METHODS: The cells were cultured as high glucose (HG) and normal control (C) groups, and CSF1R shRNA was introduced. Real time PCR was used to detect the expression of CSF1R and p-ERK1/2 mRNA. Western blot was used to detect the expression of CSF1R and p-ERK1/2 proteins. Cell Counting Kit 8 (CCK-8) method was used to detect cell proliferation, while flow cytometry was used to detect apoptosis in HREC. RESULTS: Real-time PCR showed significantly raised CSF1R mRNA expression in HG. CSF1R inhibition lowered HG + LV shCSF1R CSF1R mRNA levels. Western blotting revealed higher CSF1R and p-ERK1/2 protein expression in HG than in C. Their expression level dropped after CSF1R inhibition. The number of tube-forming cells was higher in HG than in C, which reduced after CSF1R suppression. Inhibiting CSF1R also decreased cell proliferation and raised apoptosis. CONCLUSION: Overall, under high glucose, CSF1R and p-ERK1/2 were highly expressed, leading to reduced cellular activity, and CSF1R inhibition helped alleviate this effect.


Asunto(s)
Apoptosis , Western Blotting , Proliferación Celular , Glucosa , Sistema de Señalización de MAP Quinasas , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Glucosa/farmacología , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , 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 , Sistema de Señalización de MAP Quinasas/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Citometría de Flujo , Animales , Regulación de la Expresión Génica , Línea Celular , Microglía/metabolismo , Microglía/efectos de los fármacos , Ratas , Transducción de Señal , ARN Interferente Pequeño/genética
16.
Glia ; 72(2): 375-395, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37909242

RESUMEN

White matter abnormalities, related to poor cerebral perfusion, are a core feature of small vessel cerebrovascular disease, and critical determinants of vascular cognitive impairment and dementia. Despite this importance there is a lack of treatment options. Proliferation of microglia producing an expanded, reactive population and associated neuroinflammatory alterations have been implicated in the onset and progression of cerebrovascular white matter disease, in patients and in animal models, suggesting that targeting microglial proliferation may exert protection. Colony-stimulating factor-1 receptor (CSF1R) is a key regulator of microglial proliferation. We found that the expression of CSF1R/Csf1r and other markers indicative of increased microglial abundance are significantly elevated in damaged white matter in human cerebrovascular disease and in a clinically relevant mouse model of chronic cerebral hypoperfusion and vascular cognitive impairment. Using the mouse model, we investigated long-term pharmacological CSF1R inhibition, via GW2580, and demonstrated that the expansion of microglial numbers in chronic hypoperfused white matter is prevented. Transcriptomic analysis of hypoperfused white matter tissue showed enrichment of microglial and inflammatory gene sets, including phagocytic genes that were the predominant expression modules modified by CSF1R inhibition. Further, CSF1R inhibition attenuated hypoperfusion-induced white matter pathology and rescued spatial learning impairments and to a lesser extent cognitive flexibility. Overall, this work suggests that inhibition of CSF1R and microglial proliferation mediates protection against chronic cerebrovascular white matter pathology and cognitive deficits. Our study nominates CSF1R as a target for the treatment of vascular cognitive disorders with broader implications for treatment of other chronic white matter diseases.


Asunto(s)
Trastornos Cerebrovasculares , Trastornos del Conocimiento , Disfunción Cognitiva , Leucoencefalopatías , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Sustancia Blanca , Animales , Ratones , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Receptores del Factor Estimulante de Colonias/metabolismo , Sustancia Blanca/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
17.
Nat Commun ; 14(1): 118, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36624100

RESUMEN

Microglia are central to pathogenesis in many neurological conditions. Drugs targeting colony-stimulating factor-1 receptor (CSF1R) to block microglial proliferation in preclinical disease models have shown mixed outcomes, thus the therapeutic potential of this approach remains unclear. Here, we show that CSF1R inhibitors given by multiple dosing paradigms in the Tg2541 tauopathy mouse model cause a sex-independent reduction in pathogenic tau and reversion of non-microglial gene expression patterns toward a normal wild type signature. Despite greater drug exposure in male mice, only female mice have functional rescue and extended survival. A dose-dependent upregulation of immediate early genes and neurotransmitter dysregulation are observed in the brains of male mice only, indicating that excitotoxicity may preclude functional benefits. Drug-resilient microglia in male mice exhibit morphological and gene expression patterns consistent with increased neuroinflammatory signaling, suggesting a mechanistic basis for sex-specific excitotoxicity. Complete microglial ablation is neither required nor desirable for neuroprotection and therapeutics targeting microglia must consider sex-dependent effects.


Asunto(s)
Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Tauopatías , Animales , Femenino , Masculino , Ratones , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Microglía/metabolismo , Fenotipo , Receptores del Factor Estimulante de Colonias/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Tauopatías/metabolismo
18.
Eur J Med Chem ; 245(Pt 1): 114884, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36335744

RESUMEN

Colony-Stimulating Factor-1 Receptor (CSF1R) is a receptor tyrosine kinase that controls the differentiation and maintenance of most tissue-resident macrophages and bone-resorbing osteoclasts. Mutations of CSF1R have been implicated in neurodegeneration, skeletal anomalies, and cancers. Activation of CSF1R by endogenous cytokine ligation to the ectodomain triggers the autophosphorylation of the intracellular tyrosine kinase domain, and thereafter, activation of several downstream pro-survival kinase cascades, including PI3K, ERK1/2, and JNK. The immunological role of CSF1R in regulating tumor-associate macrophages (TAMs) have been well-documented. TAMs harboring activated CSF1R release tumorigenic cytokines, which further deconditioning tumor microenvironment to a protumoral phenotype. Pharmacological inhibition of CSF1R has emerged as a promising antitumor strategy, with PLX3397 (pexidartinib) been approved by the FDA for the treatment of tenosynovial giant cell tumor in 2019. Research around developing novel small-molecule CSF1R inhibitors, as well as expanding their potential indications, have drawn numerous attentions thenceforward. Herein, we've comprehensively reviewed the latest progression of CSF1R inhibitors under clinical and preclinical studies. Key findings of CSF1R targeted therapies either as monotherapy or combinatorial therapy have also been discussed.


Asunto(s)
Antineoplásicos , Inmunoterapia , Neoplasias , Proteínas Tirosina Quinasas Receptoras , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Citocinas/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , /farmacología , /uso terapéutico
19.
Cell Death Dis ; 13(10): 859, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-36209194

RESUMEN

Sustained oxidative stress in castration-resistant prostate cancer (CRPC) cells potentiates the overall tumor microenvironment (TME). Targeting the TME using colony-stimulating factor 1 receptor (CSF1R) inhibition is a promising therapy for CRPC. However, the therapeutic response to sustained CSF1R inhibition (CSF1Ri) is limited as a monotherapy. We hypothesized that one of the underlying causes for the reduced efficacy of CSF1Ri and increased oxidation in CRPC is the upregulation and uncoupling of endothelial nitric oxide synthase (NOS3). Here we show that in high-grade PCa human specimens, NOS3 abundance positively correlates with CSF1-CSF1R signaling and remains uncoupled. The uncoupling diminishes NOS3 generation of sufficient nitric oxide (NO) required for S-nitrosylation of CSF1R at specific cysteine sites (Cys 224, Cys 278, and Cys 830). Exogenous S-nitrosothiol administration (with S-nitrosoglutathione (GSNO)) induces S-nitrosylation of CSF1R and rescues the excess oxidation in tumor regions, in turn suppressing the tumor-promoting cytokines which are ineffectively suppressed by CSF1R blockade. Together these results suggest that NO administration could act as an effective combinatorial partner with CSF1R blockade against CRPC. In this context, we further show that exogenous NO treatment with GSNOR successfully augments the anti-tumor ability of CSF1Ri to effectively reduce the overall tumor burden, decreases the intratumoral percentage of anti-inflammatory macrophages, myeloid-derived progenitor cells and increases the percentage of pro-inflammatory macrophages, cytotoxic T lymphocytes, and effector T cells, respectively. Together, these findings support the concept that the NO-CSF1Ri combination has the potential to act as a therapeutic agent that restores control over TME, which in turn could improve the outcomes of PCa patients.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Receptor de Factor Estimulante de Colonias de Macrófagos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Cisteína , Humanos , Factor Estimulante de Colonias de Macrófagos , Masculino , Óxido Nítrico , Óxido Nítrico Sintasa de Tipo III , S-Nitrosoglutatión , Microambiente Tumoral
20.
Sci Transl Med ; 14(651): eabg7504, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35767650

RESUMEN

The role of tissue-resident macrophages during tissue regeneration or fibrosis is not well understood, mainly due to the lack of a specific marker for their identification. Here, we identified three populations of skeletal muscle-resident myelomonocytic cells: a population of macrophages positive for lymphatic vessel endothelial receptor 1 (LYVE1) and T cell membrane protein 4 (TIM4 or TIMD4), a population of LYVE1-TIM4- macrophages, and a population of cells likely representing dendritic cells that were positive for CD11C and major histocompatibility complex class II (MHCII). Using a combination of parabiosis and lineage-tracing experiments, we found that, at steady state, TIM4- macrophages were replenished from the blood, whereas TIM4+ macrophages locally self-renewed [self-renewing resident macrophages (SRRMs)]. We further showed that Timd4 could be reliably used to distinguish SRRMs from damage-induced infiltrating macrophages. Using a colony-stimulating factor 1 receptor (CSF1R) inhibition/withdrawal approach to specifically deplete SRRMs, we found that SRRMs provided a nonredundant function in clearing damage-induced apoptotic cells early after extensive acute injury. In contrast, in chronic mild injury as seen in a mouse model of Duchenne muscular dystrophy, depletion of both TIM4-- and TIM4+-resident macrophage populations through long-term CSF1R inhibition changed muscle fiber composition from damage-sensitive glycolytic fibers toward damage-resistant glycolytic-oxidative fibers, thereby protecting muscle against contraction-induced injury both ex vivo and in vivo. This work reveals a previously unidentified role for resident macrophages in modulating tissue metabolism and may have therapeutic potential given the ongoing clinical testing of CSF1R inhibitors.


Asunto(s)
Macrófagos , Músculo Esquelético , Distrofias Musculares , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Animales , Macrófagos/metabolismo , Macrófagos/patología , Proteínas de la Membrana/metabolismo , Ratones , Monocitos/metabolismo , Monocitos/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/tratamiento farmacológico , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas de Transporte Vesicular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA