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













Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35054815

RESUMEN

Neurodegenerative diseases are debilitating impairments that affect millions of people worldwide and are characterized by progressive degeneration of structure and function of the central or peripheral nervous system. Effective biomarkers for neurodegenerative diseases can be used to improve the diagnostic workup in the clinic as well as facilitate the development of effective disease-modifying therapies. Progranulin (PGRN) has been reported to be involved in various neurodegenerative disorders. Hence, in the current study we systematically compared the inflammation and accumulation of typical neurodegenerative disease markers in the brain tissue between PGRN knockout (PGRN KO) and wildtype (WT) mice. We found that PGRN deficiency led to significant neuron loss as well as activation of microglia and astrocytes in aged mice. Several characteristic neurodegenerative markers, including α-synuclein, TAR DNA-binding protein 43 (TDP-43), Tau, and ß-amyloid, were all accumulated in the brain of PGRN-deficient mice as compared to WT mice. Moreover, higher aggregation of lipofuscin was observed in the brain tissue of PGRN-deficient mice compared with WT mice. In addition, the autophagy was also defective in the brain of PGRN-deficient mice, indicated by the abnormal expression level of autophagy marker LC3-II. Collectively, comprehensive assays support the idea that PGRN plays an important role during the development of neurodegenerative disease, indicating that PGRN might be a useful biomarker for neurodegenerative diseases in clinical settings.


Asunto(s)
Envejecimiento/patología , Biomarcadores/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Progranulinas/deficiencia , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Autofagia , Encéfalo/patología , Encéfalo/ultraestructura , Proteínas de Unión al ADN/metabolismo , Lipofuscina/metabolismo , Ratones Noqueados , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Progranulinas/metabolismo , Agregado de Proteínas , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
2.
EMBO J ; 41(4): e109108, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35019161

RESUMEN

Haploinsufficiency of the progranulin (PGRN)-encoding gene (GRN) causes frontotemporal lobar degeneration (GRN-FTLD) and results in microglial hyperactivation, TREM2 activation, lysosomal dysfunction, and TDP-43 deposition. To understand the contribution of microglial hyperactivation to pathology, we used genetic and pharmacological approaches to suppress TREM2-dependent transition of microglia from a homeostatic to a disease-associated state. Trem2 deficiency in Grn KO mice reduced microglia hyperactivation. To explore antibody-mediated pharmacological modulation of TREM2-dependent microglial states, we identified antagonistic TREM2 antibodies. Treatment of macrophages from GRN-FTLD patients with these antibodies led to reduced TREM2 signaling due to its enhanced shedding. Furthermore, TREM2 antibody-treated PGRN-deficient microglia derived from human-induced pluripotent stem cells showed reduced microglial hyperactivation, TREM2 signaling, and phagocytic activity, but lysosomal dysfunction was not rescued. Similarly, lysosomal dysfunction, lipid dysregulation, and glucose hypometabolism of Grn KO mice were not rescued by TREM2 ablation. Synaptic loss and neurofilament light-chain (NfL) levels, a biomarker for neurodegeneration, were further elevated in the Grn/Trem2 KO cerebrospinal fluid (CSF). These findings suggest that TREM2-dependent microglia hyperactivation in models of GRN deficiency does not promote neurotoxicity, but rather neuroprotection.


Asunto(s)
Degeneración Lobar Frontotemporal/patología , Glicoproteínas de Membrana/metabolismo , Microglía/fisiología , Monocitos/metabolismo , Progranulinas/deficiencia , Receptores Inmunológicos/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Degeneración Lobar Frontotemporal/metabolismo , Humanos , Lisosomas/metabolismo , Lisosomas/patología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/efectos de los fármacos , Monocitos/efectos de los fármacos , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Quinasa Syk/metabolismo
3.
Eur J Pharmacol ; 909: 174431, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34428436

RESUMEN

Recent advances have revealed that progranulin (PGRN) is related to the aetiology of psoriasis. Moreover, curcumin, a compound derived from turmeric, has been proposed as a potential therapeutic approach in psoriasis-like dermatitis, but it is still unclear whether curcumin affects the development of psoriasis-like skin lesions under PGRN-deficient conditions. Therefore, in this study, we developed a mouse model of psoriatic skin lesions using topical application of imiquimod (IMQ) in both wild type and PGRN-knockout mice to test this possibility. We observed that PGRN deficiency not only increased proinflammatory cytokine IL-17A levels and aggravated psoriasis-like damaged appearance and epidermal thickening but also directly mediated changes in keratinocyte proliferation (Krt 14, cyclinD1 and c-Myc) and differentiation (Krt 10 and Filaggrin) associated gene expression following IMQ challenge, compared to those in the control group. Furthermore, curcumin treatment (50 mg/kg and 200 mg/kg, intragastrically) for 21 consecutive days suppressed the IMQ exposure-induced increase in PGRN expression. Importantly, curcumin treatment significantly alleviated the PGRN deficiency-induced exacerbation of psoriatic appearance, histological features and keratinocyte proliferation after IMQ exposure. In summary, these results demonstrate the direct regulation of PGRN in keratinocyte proliferation and differentiation in psoriatic lesions and demonstrate the protective effect of curcumin on PGRN deficiency-induced psoriatic skin lesion exacerbation.


Asunto(s)
Curcumina/farmacología , Progranulinas/deficiencia , Psoriasis/tratamiento farmacológico , Animales , Proliferación Celular , Curcumina/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Imiquimod/administración & dosificación , Imiquimod/inmunología , Interleucina-17/sangre , Interleucina-17/metabolismo , Queratinocitos/inmunología , Queratinocitos/patología , Masculino , Ratones , Ratones Noqueados , Progranulinas/genética , Psoriasis/sangre , Psoriasis/genética , Psoriasis/inmunología
4.
Cell ; 184(18): 4651-4668.e25, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34450028

RESUMEN

GRN mutations cause frontotemporal dementia (GRN-FTD) due to deficiency in progranulin (PGRN), a lysosomal and secreted protein with unclear function. Here, we found that Grn-/- mice exhibit a global deficiency in bis(monoacylglycero)phosphate (BMP), an endolysosomal phospholipid we identified as a pH-dependent PGRN interactor as well as a redox-sensitive enhancer of lysosomal proteolysis and lipolysis. Grn-/- brains also showed an age-dependent, secondary storage of glucocerebrosidase substrate glucosylsphingosine. We investigated a protein replacement strategy by engineering protein transport vehicle (PTV):PGRN-a recombinant protein linking PGRN to a modified Fc domain that binds human transferrin receptor for enhanced CNS biodistribution. PTV:PGRN rescued various Grn-/- phenotypes in primary murine macrophages and human iPSC-derived microglia, including oxidative stress, lysosomal dysfunction, and endomembrane damage. Peripherally delivered PTV:PGRN corrected levels of BMP, glucosylsphingosine, and disease pathology in Grn-/- CNS, including microgliosis, lipofuscinosis, and neuronal damage. PTV:PGRN thus represents a potential biotherapeutic for GRN-FTD.


Asunto(s)
Productos Biológicos/uso terapéutico , Encéfalo/metabolismo , Enfermedades por Almacenamiento Lisosomal/terapia , Progranulinas/uso terapéutico , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Endosomas/metabolismo , Femenino , Demencia Frontotemporal/sangre , Demencia Frontotemporal/líquido cefalorraquídeo , Gliosis/complicaciones , Gliosis/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Inflamación/patología , Metabolismo de los Lípidos , Lipofuscina/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Degeneración Nerviosa/patología , Fenotipo , Progranulinas/deficiencia , Progranulinas/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Transferrina/metabolismo , Distribución Tisular
5.
Acta Neuropathol Commun ; 9(1): 57, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33795008

RESUMEN

Frontotemporal lobar degeneration (FTLD) causes a spectrum of clinical presentations of frontotemporal dementia (FTD), including progressive changes in behavior, personality, executive function, and language. Up to 20% of familial FTLD cases are caused by progranulin (GRN) haploinsufficiency (FTD-GRN), with one of the most common causal variant being a nonsense mutation at arginine 493 (R493X). Recently, a genetic knockin FTD-GRN mouse model was generated bearing this GrnR493X mutation, at the analogous arginine in murine Grn. Aged, homozygous GrnR493X mice (GrnR493X/R493X) have been shown to phenotypically replicate several neuropathological hallmarks previously demonstrated in Grn null mice. We conducted a comprehensive neuropathological and behavioral assessment of 18 month old GrnR493X/R493X mice, observing a striking lysosomal dysfunction and thalamic neurodegeneration not previously described in this model, as well as a male-specific increase in generalized anxiety. These findings provide additional phenotypic markers of pathogenesis in aged GrnR493X/R493X mice that will contribute to better defining mechanisms underlying FTD-GRN, and offer relevant outcome measures for preclinical efficacy testing of novel therapeutics that target nonsense mutations leading to this devastating disease.


Asunto(s)
Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Progranulinas/genética , Animales , Conducta Animal , Codón sin Sentido , Modelos Animales de Enfermedad , Femenino , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/patología , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Progranulinas/deficiencia
6.
Life Sci ; 264: 118687, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33181174

RESUMEN

Breast cancer is one of the most malignant diseases world-wide and ranks the first among female cancers. Progranulin (PGRN) plays a carcinogenic role in breast cancer, but its mechanisms are not clear. In addition, there are few reports on the relationship between PGRN and tumor-associated macrophages (TAMs). AIMS: To investigate the effects of exosomes derived from PGRN-/- TAMs on invasion and migration of breast cancer cells. MAIN METHODS: Mouse breast cancer xenograft model was constructed to explore the effect of PGRN-/- tumor environment (TME) on breast cancer. Flow cytometry was used to compare TAMs of wild type (WT) and PGRN-/- tumor tissue. Transwell assay, wound healing assay and western blot were used to explore the effect of WT and PGRN-/- TAMs and their exosomes on invasion, migration and epithelial-mesenchymal transition (EMT) of breast cancer cells. MicroRNA (miRNA) assay was used to find out the differentially expressed miRNA of negative control (NC) and siPGRN-TAMs exosomes. Quantitative PCR and luciferase report assay were used to explore the target gene. KEY FINDINGS: The lung metastasis of breast cancer of PGRN-/- mice was inhibited. PGRN-/- TAMs inhibited invasion, migration and EMT of breast cancer cells through their exosomes. MiR-5100 of PGRN-/- TAMs-derived exosomes was up-regulated, which might regulate expression of CXCL12, thereby inhibiting the CXCL12/CXCR4 axis, and ultimately inhibiting the invasion, migration and EMT of breast cancer cells. SIGNIFICANCE: Our study elucidates a new molecular mechanism of lung metastasis of breast cancer, so it may contribute to efficient prevention and therapeutic strategies.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Exosomas/metabolismo , Progranulinas/deficiencia , Macrófagos Asociados a Tumores/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/prevención & control , Exosomas/genética , Exosomas/patología , Femenino , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Invasividad Neoplásica/genética , Invasividad Neoplásica/prevención & control , Progranulinas/genética , Macrófagos Asociados a Tumores/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
7.
Ann Clin Transl Neurol ; 7(12): 2433-2449, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33197149

RESUMEN

OBJECTIVE: The goal of this study was to investigate the effect of progranulin insufficiency on extracellular vesicles (EVs), a heterogeneous population of vesicles that may contribute to progression of neurodegenerative disease. Loss-of-function mutations in progranulin (GRN) are a major cause of frontotemporal dementia (FTD), and brains from GRN carriers with FTD (FTD-GRN) exhibit signs of lysosomal dysfunction. Lysosomal dysfunction may induce compensatory increases in secretion of exosomes, EVs secreted from the endolysosomal system, so we hypothesized that progranulin insufficiency would increase EV levels in the brain. METHODS: We analyzed levels and protein contents of brain EVs from Grn-/- mice, which model the lysosomal abnormalities of FTD-GRN patients. We then measured brain EVs in FTD-GRN patients. To assess the relationship of EVs with symptomatic disease, we measured plasma EVs in presymptomatic and symptomatic GRN mutation carriers. RESULTS: Grn-/- mice had elevated brain EV levels and altered EV protein contents relative to wild-type mice. These changes were age-dependent, occurring only after the emergence of pathology in Grn-/- mice. FTD-GRN patients (n = 13) had elevated brain EV levels relative to controls (n = 5). Symptomatic (n = 12), but not presymptomatic (n = 7), GRN carriers had elevated plasma EV levels relative to controls (n = 8). INTERPRETATION: These data show that symptomatic FTD-GRN patients have elevated levels of brain and plasma EVs, and that this effect is modeled in the brain of Grn-/- mice after the onset of pathology. This increase in EVs could influence FTD disease progression, and provides further support for EVs as potential FTD biomarkers.


Asunto(s)
Vesículas Extracelulares/metabolismo , Lóbulo Frontal/metabolismo , Demencia Frontotemporal/metabolismo , Progranulinas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Progresión de la Enfermedad , Femenino , Demencia Frontotemporal/sangre , Demencia Frontotemporal/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Progranulinas/deficiencia , Progranulinas/genética , Proteómica , Método Simple Ciego
8.
Nature ; 588(7838): 459-465, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32866962

RESUMEN

Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.


Asunto(s)
Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Progranulinas/deficiencia , Proteinopatías TDP-43/metabolismo , Proteinopatías TDP-43/patología , Envejecimiento/genética , Envejecimiento/patología , Animales , Núcleo Celular/genética , Núcleo Celular/patología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Complemento C1q/antagonistas & inhibidores , Complemento C1q/inmunología , Complemento C3b/antagonistas & inhibidores , Complemento C3b/inmunología , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Poro Nuclear/metabolismo , Poro Nuclear/patología , Progranulinas/genética , RNA-Seq , Análisis de la Célula Individual , Proteinopatías TDP-43/tratamiento farmacológico , Proteinopatías TDP-43/genética , Tálamo/metabolismo , Tálamo/patología , Transcriptoma
9.
Sci Rep ; 10(1): 13688, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792571

RESUMEN

Patients with frontotemporal dementia (FTD) resulting from granulin (GRN) haploinsufficiency have reduced levels of progranulin and exhibit dysregulation in inflammatory and lysosomal networks. Microglia produce high levels of progranulin, and reduction of progranulin in microglia alone is sufficient to recapitulate inflammation, lysosomal dysfunction, and hyperproliferation in a cell-autonomous manner. Therefore, targeting microglial dysfunction caused by progranulin insufficiency represents a potential therapeutic strategy to manage neurodegeneration in FTD. Limitations of current progranulin-enhancing strategies necessitate the discovery of new targets. To identify compounds that can reverse microglial defects in Grn-deficient mouse microglia, we performed a compound screen coupled with high throughput sequencing to assess key transcriptional changes in inflammatory and lysosomal pathways. Positive hits from this initial screen were then further narrowed down based on their ability to rescue cathepsin activity, a critical biochemical readout of lysosomal capacity. The screen identified nor-binaltorphimine dihydrochloride (nor-BNI) and dibutyryl-cAMP, sodium salt (DB-cAMP) as two phenotypic modulators of progranulin deficiency. In addition, nor-BNI and DB-cAMP also rescued cell cycle abnormalities in progranulin-deficient cells. These data highlight the potential of a transcription-based platform for drug screening, and advance two novel lead compounds for FTD.


Asunto(s)
Bucladesina/farmacología , Proteasas de Cisteína/metabolismo , Demencia Frontotemporal/genética , Perfilación de la Expresión Génica/métodos , Microglía/citología , Naltrexona/análogos & derivados , Progranulinas/deficiencia , Animales , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Demencia Frontotemporal/tratamiento farmacológico , Demencia Frontotemporal/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Modelos Biológicos , Naltrexona/farmacología , Análisis de Secuencia de ARN , Bibliotecas de Moléculas Pequeñas/farmacología
10.
Ann N Y Acad Sci ; 1460(1): 43-56, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31423598

RESUMEN

Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by insulin deficiency, and patients with diabetes have an increased risk of bone fracture and significantly impaired fracture healing. Proinflammatory cytokine tumor necrosis factor-alpha is significantly upregulated in diabetic fractures and is believed to underlie delayed fracture healing commonly observed in diabetes. Our previous genetic screen for the binding partners of progranulin (PGRN), a growth factor-like molecule that induces chondrogenesis, led to the identification of tumor necrosis factor receptors (TNFRs) as the PGRN-binding receptors. In this study, we employed several in vivo models to ascertain whether PGRN has therapeutic effects in diabetic fracture healing. Here, we report that deletion of PGRN significantly delayed bone fracture healing and aggravated inflammation in the fracture models of mice with T1DM. In contrast, recombinant PGRN effectively promoted diabetic fracture healing by inhibiting inflammation and enhancing chondrogenesis. In addition, both TNFR1 proinflammatory and TNFR2 anti-inflammatory signaling pathways are involved in PGRN-stimulated diabetic fracture healing. Collectively, these findings illuminate a novel understanding concerning the role of PGRN in diabetic fracture healing and may have an application in the development of novel therapeutic intervention strategies for diabetic and other types of impaired fracture healing.


Asunto(s)
Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/patología , Curación de Fractura/efectos de los fármacos , Progranulinas/farmacología , Animales , Condrogénesis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Eliminación de Gen , Humanos , Inflamación/patología , Ratones , Progranulinas/deficiencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Cell Mol Immunol ; 17(10): 1077-1091, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31467413

RESUMEN

Progranulin is a secreted neurotrophin that assists in the autophagolysosomal pathways that contribute to MHC-mediated antigen processing, pathogen removal, and autoimmunity. We showed that patients with multiple sclerosis (MS) have high levels of circulating progranulin and that its depletion in a mouse model by a monoclonal antibody aggravates MS-like experimental autoimmune encephalomyelitis (EAE). However, unexpectedly, progranulin-deficient mice (Grn-/-) were resistant to EAE, and this resistance was fully restored by wild-type bone marrow transplantation. FACS analyses revealed a loss of MHC-II-positive antigen-presenting cells in Grn-/- mice and a reduction in the number of CD8+ and CD4+ T-cells along with a strong increase in the number of scavenger receptor class B (CD36+) phagocytes, suggesting defects in antigen presentation along with a compensatory increase in phagocytosis. Indeed, bone marrow-derived dendritic cells from Grn-/- mice showed stronger uptake of antigens but failed to elicit antigen-specific T-cell proliferation. An increase in the number of CD36+ phagocytes was associated with increased local inflammation at the site of immunization, stronger stimulation-evoked morphological transformation of bone marrow-derived macrophages to phagocytes, an increase in the phagocytosis of E. coli particles and latex beads and defects in the clearance of the material. Hence, the outcomes in the EAE model reflect the dichotomy of progranulin-mediated immune silencing and autoimmune mechanisms of antigen recognition and presentation, and our results reveal a novel progranulin-dependent pathway in autoimmune encephalomyelitis.


Asunto(s)
Resistencia a la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Progranulinas/deficiencia , Adolescente , Adulto , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos/metabolismo , Médula Ósea/patología , Trasplante de Médula Ósea , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/sangre , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Recuento de Linfocitos , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Esclerosis Múltiple/sangre , Esclerosis Múltiple/patología , Células Mieloides/patología , Ovalbúmina , Fagocitos/metabolismo , Fagocitosis , Progranulinas/sangre , Progranulinas/metabolismo , Receptores Depuradores de Clase B/metabolismo , Linfocitos T/inmunología , Adulto Joven
12.
J Interferon Cytokine Res ; 40(1): 33-42, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31804874

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease caused by the disorders of immune regulation but its pathogenesis is poorly understood. Progranulin (PGRN) is an immunomodulatory protein that is upregulated in SLE patients. However, the factors involved in regulating the pathogenesis of SLE by PGRN are largely unknown. We sought to investigate the role and molecular mechanisms of PGRN in SLE to develop a novel therapeutic target. We used an animal model of SLE that was induced in PGRN-deficient and normal wild type (WT) mice using pristane. PGRN concentrations were measured in SLE and the impact of PGRN deficiency was examined by measuring tissue injury and immune responses of T cells (Th1, Th2, Th17, and Treg) and B cells. SLE patients and mice showed elevated PGRN levels. Compared with WT SLE mice, inflammatory cell infiltration, tissue edema, and necrosis were alleviated in PGRN-/- SLE mice and the levels of serum chemistry markers of tissue damage and the presence of anti-double-stranded DNA and anti-ribosomal protein P0 antibodies were all significantly decreased. We further discovered that PGRN deficiency could disturb the immune responses of T cell (Th1, Th2, Th17, and Treg) and B cell responses, leading to the decrease of inflammatory cytokines including interferon-γ and interleukin-17A and increased levels of regulatory B cells. PGRN plays a proinflammatory role in the development of SLE partially through promoting the production of autoantibodies and enhancing Th1 and Th17 cell responses. This may provide new therapeutic options for patients with SLE.


Asunto(s)
Inflamación/inmunología , Inflamación/patología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Progranulinas/inmunología , Adolescente , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/terapia , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/terapia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Progranulinas/deficiencia , Terpenos
13.
J Mol Cell Cardiol ; 138: 197-211, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31866375

RESUMEN

AIMS: Age-related cardiac hypertrophy and subsequent heart failure are predicted to become increasingly serious problems in aging populations. Progranulin (PGRN) deficiency is known to be associated with accelerated aging in the brain. We aimed to evaluate the effects of PGRN deficiency on cardiac aging, including left ventricular hypertrophy. METHODS AND RESULTS: Echocardiography was performed on wild-type (WT) and PGRN-knockout (KO) mice every 3 months from 3 to 18 months of age. Compared to that of WT mice, PGRN KO mice exhibited age-dependent cardiac hypertrophy and cardiac dysfunction at 18 months. Morphological analyses showed that the heart weight to tibia length ratio and cross-sectional area of cardiomyocytes at 18 months were significantly increased in PGRN KO mice relative to those in WT mice. Furthermore, accumulation of lipofuscin and increases in senescence markers were observed in the hearts of PGRN KO mice, suggesting that PGRN deficiency led to enhanced aging of the heart. Enhanced complement C1q (C1q) and activated ß-catenin protein expression levels were also observed in the hearts of aged PGRN KO mice. Treatment of PGRN-deficient cardiomyocytes with C1q caused ß-catenin activation and cardiac hypertrophy. Blocking C1q-induced ß-catenin activation in PGRN-depleted cardiomyocytes attenuated hypertrophic changes. Finally, we showed that C1 inhibitor treatment reduced cardiac hypertrophy and dysfunction in old KO mice, possibly by reducing ß-catenin activation. These results suggest that C1q is a crucial regulator of cardiac hypertrophy induced by PGRN ablation. CONCLUSION: The present study demonstrates that PGRN deficiency enhances age-related cardiac hypertrophy via C1q-induced ß-catenin activation. PGRN is a potential therapeutic target to prevent cardiac hypertrophy and dysfunction.


Asunto(s)
Envejecimiento/metabolismo , Cardiomegalia/metabolismo , Complemento C1q/metabolismo , Progranulinas/deficiencia , beta Catenina/metabolismo , Animales , Aorta/patología , Biomarcadores/metabolismo , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Constricción Patológica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Miocitos Cardíacos/metabolismo , Fenotipo , Presión , Progranulinas/metabolismo , Ratas , Transducción de Señal
14.
Cell Death Dis ; 10(7): 524, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285425

RESUMEN

Mitochondrial dysfunction is considered as a key mediator in the pathogenesis of diabetic nephropathy (DN). Therapeutic strategies targeting mitochondrial dysfunction hold considerable promise for the treatment of DN. In this study, we investigated the role of progranulin (PGRN), a secreted glycoprotein, in mediating mitochondrial homeostasis and its therapeutic potential in DN. We found that the level of PGRN was significantly reduced in the kidney from STZ-induced diabetic mice and patients with biopsy-proven DN compared with healthy controls. In DN model, PGRN-deficient mice aggravated podocyte injury and proteinuria versus wild-type mice. Functionally, PGRN deficiency exacerbated mitochondrial damage and dysfunction in podocytes from diabetic mice. In vitro, treatment with recombinant human PGRN (rPGRN) attenuated high glucose-induced mitochondrial dysfunction in podocytes accompanied by enhanced mitochondrial biogenesis and mitophagy. Inhibition of mitophagy disturbed the protective effects of PGRN in high glucose-induced podocytotoxicity. Mechanistically, we demonstrated that PGRN maintained mitochondrial homeostasis via PGRN-Sirt1-PGC-1α/FoxO1 signaling-mediated mitochondrial biogenesis and mitophagy. Finally, we provided direct evidence for therapeutic potential of PGRN in mice with DN. This study provides new insights into the novel role of PGRN in maintaining mitochondrial homeostasis, suggesting that PGRN may be an innovative therapeutic strategy for treating patients with DN.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Homeostasis , Mitocondrias/metabolismo , Mitofagia , Biogénesis de Organelos , Podocitos/patología , Progranulinas/metabolismo , Acetilación , Animales , Biopsia , Muerte Celular , Citoprotección , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Proteína Forkhead Box O1/metabolismo , Glucosa/toxicidad , Humanos , Riñón/metabolismo , Riñón/patología , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Podocitos/metabolismo , Podocitos/ultraestructura , Progranulinas/deficiencia , Ratas , Sirtuina 1/metabolismo
15.
PLoS One ; 14(7): e0212382, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31291241

RESUMEN

Mutation in the GRN gene, encoding the progranulin (PGRN) protein, shows a dose-dependent disease correlation, wherein haploinsufficiency results in frontotemporal lobar degeneration (FTLD) and complete loss results in neuronal ceroid lipofuscinosis (NCL). Although the exact function of PGRN is unknown, it has been increasingly implicated in lysosomal physiology. Here we report that PGRN interacts with the lysosomal enzyme, glucocerebrosidase (GCase), and is essential for proper GCase activity. GCase activity is significantly reduced in tissue lysates from PGRN-deficient mice. This is further evidence that reduced lysosomal hydrolase activity may be a pathological mechanism in cases of GRN-related FTLD and NCL.


Asunto(s)
Glucosilceramidasa/metabolismo , Progranulinas/deficiencia , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Glucosilceramidasa/genética , Células HEK293 , Haploinsuficiencia , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Progranulinas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
16.
EMBO Mol Med ; 11(6)2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31122931

RESUMEN

Microglia adopt numerous fates with homeostatic microglia (HM) and a microglial neurodegenerative phenotype (MGnD) representing two opposite ends. A number of variants in genes selectively expressed in microglia are associated with an increased risk for neurodegenerative diseases such as Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD). Among these genes are progranulin (GRN) and the triggering receptor expressed on myeloid cells 2 (TREM2). Both cause neurodegeneration by mechanisms involving loss of function. We have now isolated microglia from Grn-/- mice and compared their transcriptomes to those of Trem2-/-mice Surprisingly, while loss of Trem2 enhances the expression of genes associated with a homeostatic state, microglia derived from Grn-/- mice showed a reciprocal activation of the MGnD molecular signature and suppression of gene characteristic for HM The opposite mRNA expression profiles are associated with divergent functional phenotypes. Although loss of TREM2 and progranulin resulted in opposite activation states and functional phenotypes of microglia, FDG (fluoro-2-deoxy-d-glucose)-µPET of brain revealed reduced glucose metabolism in both conditions, suggesting that opposite microglial phenotypes result in similar wide spread brain dysfunction.


Asunto(s)
Cerebelo , Glucosa/metabolismo , Glicoproteínas de Membrana/deficiencia , Microglía/metabolismo , Tomografía de Emisión de Positrones , Progranulinas/deficiencia , Receptores Inmunológicos/deficiencia , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Cerebelo/diagnóstico por imagen , Cerebelo/metabolismo , Degeneración Lobar Frontotemporal/diagnóstico por imagen , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Ratones , Ratones Noqueados
17.
Mol Ther ; 27(2): 465-478, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30559071

RESUMEN

Adeno-associated virus-mediated gene replacement is emerging as a safe and effective means of correcting single-gene mutations affecting the CNS. AAV-mediated progranulin gene (GRN) delivery has been proposed as a treatment for GRN-deficient frontotemporal dementia and neuronal ceroid lipofuscinosis, and recent studies using intraparenchymal AAV-Grn delivery to brain have shown moderate success in histopathologic and behavioral rescue in mouse models. Here, we used AAV9 to deliver GRN to the lateral ventricle to achieve widespread expression in the Grn null mouse brain. We found that, despite a global increase in progranulin, overexpression resulted in dramatic and selective hippocampal toxicity and degeneration affecting neurons and glia. Hippocampal degeneration was preceded by T cell infiltration and perivascular cuffing. GRN delivery with an ependymal-targeting AAV for selective secretion of progranulin into the cerebrospinal fluid similarly resulted in T cell infiltration, as well as ependymal hypertrophy. Interestingly, overexpression of GRN in wild-type animals also provoked T cell infiltration. These results call into question the safety of GRN overexpression in the CNS, with evidence for both a region-selective immune response and cellular proliferative response. Our results highlight the importance of careful consideration of target gene biology and cellular response to overexpression prior to progressing to the clinic.


Asunto(s)
Dependovirus/genética , Progranulinas/metabolismo , Linfocitos T/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Demencia Frontotemporal/genética , Demencia Frontotemporal/terapia , Terapia Genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Modelos Teóricos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/terapia , Progranulinas/líquido cefalorraquídeo , Progranulinas/deficiencia , Progranulinas/genética
18.
Neurobiol Aging ; 72: 186.e9-186.e12, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30057241

RESUMEN

Loss-of-function mutations in progranulin (PGRN) gene cause frontotemporal lobar degeneration. Here, we report a case of a 63-year-old woman with a 2-year history of speech impairment, diagnosed with a nonfluent variant of primary progressive aphasia, a subtype of frontotemporal lobar degeneration. In this patient, a novel heterozygous frameshift mutation, c.77delG, in exon 2 of PGRN gene, introducing premature stop codon, p.(C26SfsX28), has been identified. Cultured fibroblasts derived from the patient and her asymptomatic first-degree relative with c.77delG mutation had decreased levels of PGRN messenger RNA (mRNA) and protein. However, PGRN mRNA levels did not recover upon incubation with inhibitors of nonsense-mediated mRNA decay (cycloheximide or puromycin), suggesting involvement of other mRNA degradation pathways. In addition, we observed upregulated wingless-type mouse mammary tumor virus integration site (WNT) signaling pathway gene, WNT3A, in fibroblasts of the patient and her asymptomatic first-degree relative with c.77delG mutation. As reported previously, this is an early hallmark of PGRN deficiency.


Asunto(s)
Fibroblastos/metabolismo , Afasia Progresiva Primaria no Fluente/genética , Progranulinas/genética , Proteína Wnt3/genética , Células Cultivadas , Femenino , Mutación del Sistema de Lectura , Haploinsuficiencia , Humanos , Persona de Mediana Edad , Linaje , Progranulinas/deficiencia , ARN Mensajero/metabolismo
19.
Methods Mol Biol ; 1806: 233-253, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29956280

RESUMEN

Currently one of the few molecules that equally excites a neuroscientist, a cancer biologist, an immunologist, and a developmental biologist is progranulin (GRN/Grn)-a pluripotent growth factor that plays key roles in cell survival, proliferation, development, tissue regeneration, inflammation, wound healing, and angiogenesis. However, the molecular pathways associated with GRN signaling involved in these varied physiological processes are not understood. Gene inactivation has been considered as one of the best methods to delineate the biological role of a protein, and gene targeting is a direct means to disrupt a gene's open reading frame and block its expression, for instance, in a mouse. Such a gene knockout animal model also served as an in vivo disease model where loss of gene or its function is thought to be the primary disease mechanism, as is the case with progranulin loss of function in frontotemporal lobar degeneration (FTLD). It is estimated that up to half of the cases of familial, dominant FTLD might be due to GRN haploinsufficiency. To understand the molecular pathways associated with GRN loss, constitutive and conditional progranulin knockout (Grn-/-) mice have also been constructed in several laboratories, including ours. These mice show several disease-characteristic features and suggest that continued studies on the Grn-/- mice would be instructive in the understanding of complex GRN biology in health and disease.


Asunto(s)
Conducta Animal , Encéfalo/metabolismo , Biología Molecular/métodos , Progranulinas/metabolismo , Envejecimiento , Animales , Aprendizaje por Laberinto , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa , Progranulinas/deficiencia , Progranulinas/aislamiento & purificación
20.
Sci Rep ; 7(1): 1679, 2017 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-28490764

RESUMEN

Astrocytes are glial cells that support and protect neurons in the central nervous systems including the retina. Retinal ganglion cells (RGCs) are in contact with the astrocytes and our earlier findings showed the reduction of the number of cells in the ganglion cell layer in adult progranulin deficient mice. In the present study, we focused on the time of activation of the astrocytes and the alterations in the number of RGCs in the retina and optic nerve in progranulin deficient mice. Our findings showed that the number of Brn3a-positive cells was reduced and the expression of glial fibrillary acidic protein (GFAP) was increased in progranulin deficient mice. The progranulin deficient mice had a high expression of GFAP on postnatal day 9 (P9) but not on postnatal day 1. These mice also had a decrease in the number of the Brn3a-positive cells on P9. Taken together, these findings indicate that the absence of progranulin can affect the survival of RGCs subsequent the activation of astrocytes during retinal development.


Asunto(s)
Crecimiento y Desarrollo , Progranulinas/deficiencia , Células Ganglionares de la Retina/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Nervio Óptico/metabolismo , Progranulinas/metabolismo , Factor de Transcripción Brn-3A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA