Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.419
Filtrar
1.
Nat Med ; 30(5): 1406-1415, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38745011

RESUMEN

GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .


Asunto(s)
Dependovirus , Demencia Frontotemporal , Terapia Genética , Progranulinas , Humanos , Demencia Frontotemporal/genética , Demencia Frontotemporal/terapia , Demencia Frontotemporal/líquido cefalorraquídeo , Progranulinas/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Dependovirus/genética , Persona de Mediana Edad , Femenino , Masculino , Anciano , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/líquido cefalorraquídeo , Vectores Genéticos , Animales , Resultado del Tratamiento , Investigación Biomédica Traslacional , Ratones , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Proteínas de Neurofilamentos/sangre
2.
Reprod Biol Endocrinol ; 22(1): 38, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38575956

RESUMEN

The present study aimed to examine the effects of progranulin and omentin on basic ovarian cell functions. For this purpose, we investigated the effects of the addition of progranulin and omentin (0, 0.1, 1, or 10 ng/ml) on the viability, proliferation, apoptosis and steroidogenesis of cultured rabbit ovarian granulosa cells. To determine the importance of the interrelationships between granulosa cells and theca cells, we compared the influence of progranulin and omentin on progesterone and estradiol release in cultured granulosa cells and ovarian fragments containing both granulosa cells and theca cells. Cell viability, proliferation, cytoplasmic apoptosis and release of progesterone and estradiol were measured by Cell Counting Kit-8 (CCK-8), BrdU incorporation, cell death detection, and ELISA. Both progranulin and omentin increased granulosa cell viability and proliferation and decreased apoptosis. Progranulin increased progesterone release by granulosa cells but reduced progesterone output by ovarian fragments. Progranulin decreased estradiol release by granulosa cells but increased it in ovarian fragments. Omentin reduced progesterone release in both models. Omentin reduced estradiol release by granulosa cells but promoted this release in ovarian fragments. The present observations are the first to demonstrate that progranulin and omentin can be direct regulators of basic ovarian cell functions. Furthermore, the differences in the effects of these adipokines on steroidogenesis via granulosa and ovarian fragments indicate that these peptides could target both granulosa and theca cells.


Asunto(s)
Adipoquinas , Progesterona , Femenino , Animales , Conejos , Progesterona/metabolismo , Progranulinas/metabolismo , Progranulinas/farmacología , Adipoquinas/metabolismo , Adipoquinas/farmacología , Ovario/metabolismo , Células de la Granulosa/metabolismo , Estradiol/metabolismo , Apoptosis , Células Cultivadas , Proliferación Celular
3.
Sci Rep ; 14(1): 9064, 2024 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-38643236

RESUMEN

Frontotemporal dementia (FTD) is a debilitating neurodegenerative disorder with currently no disease-modifying treatment options available. Mutations in GRN are one of the most common genetic causes of FTD, near ubiquitously resulting in progranulin (PGRN) haploinsufficiency. Small molecules that can restore PGRN protein to healthy levels in individuals bearing a heterozygous GRN mutation may thus have therapeutic value. Here, we show that epigenetic modulation through bromodomain and extra-terminal domain (BET) inhibitors (BETi) potently enhance PGRN protein levels, both intracellularly and secreted forms, in human central nervous system (CNS)-relevant cell types, including in microglia-like cells. In terms of potential for disease modification, we show BETi treatment effectively restores PGRN levels in neural cells with a GRN mutation known to cause PGRN haploinsufficiency and FTD. We demonstrate that BETi can rapidly and durably enhance PGRN in neural progenitor cells (NPCs) in a manner dependent upon BET protein expression, suggesting a gain-of-function mechanism. We further describe a CNS-optimized BETi chemotype that potently engages endogenous BRD4 and enhances PGRN expression in neuronal cells. Our results reveal a new epigenetic target for treating PGRN-deficient forms of FTD and provide mechanistic insight to aid in translating this discovery into therapeutics.


Asunto(s)
Demencia Frontotemporal , Humanos , Progranulinas/metabolismo , Demencia Frontotemporal/tratamiento farmacológico , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Mutación , Epigénesis Genética , Proteínas que Contienen Bromodominio , Proteínas de Ciclo Celular/metabolismo
4.
Alzheimers Dement ; 20(5): 3525-3542, 2024 May.
Artículo en Italiano | MEDLINE | ID: mdl-38623902

RESUMEN

INTRODUCTION: Effective longitudinal biomarkers that track disease progression are needed to characterize the presymptomatic phase of genetic frontotemporal dementia (FTD). We investigate the utility of cerebral perfusion as one such biomarker in presymptomatic FTD mutation carriers. METHODS: We investigated longitudinal profiles of cerebral perfusion using arterial spin labeling magnetic resonance imaging in 42 C9orf72, 70 GRN, and 31 MAPT presymptomatic carriers and 158 non-carrier controls. Linear mixed effects models assessed perfusion up to 5 years after baseline assessment. RESULTS: Perfusion decline was evident in all three presymptomatic groups in global gray matter. Each group also featured its own regional pattern of hypoperfusion over time, with the left thalamus common to all groups. Frontal lobe regions featured lower perfusion in those who symptomatically converted versus asymptomatic carriers past their expected age of disease onset. DISCUSSION: Cerebral perfusion is a potential biomarker for assessing genetic FTD and its genetic subgroups prior to symptom onset. HIGHLIGHTS: Gray matter perfusion declines in at-risk genetic frontotemporal dementia (FTD). Regional perfusion decline differs between at-risk genetic FTD subgroups . Hypoperfusion in the left thalamus is common across all presymptomatic groups. Converters exhibit greater right frontal hypoperfusion than non-converters past their expected conversion date. Cerebral hypoperfusion is a potential early biomarker of genetic FTD.


Asunto(s)
Proteína C9orf72 , Circulación Cerebrovascular , Demencia Frontotemporal , Imagen por Resonancia Magnética , Proteínas tau , Humanos , Demencia Frontotemporal/genética , Demencia Frontotemporal/fisiopatología , Demencia Frontotemporal/diagnóstico por imagen , Femenino , Masculino , Persona de Mediana Edad , Estudios Longitudinales , Circulación Cerebrovascular/fisiología , Circulación Cerebrovascular/genética , Proteína C9orf72/genética , Proteínas tau/genética , Sustancia Gris/diagnóstico por imagen , Sustancia Gris/patología , Progranulinas/genética , Biomarcadores , Progresión de la Enfermedad , Encéfalo/diagnóstico por imagen , Heterocigoto , Mutación , Anciano , Marcadores de Spin , Adulto
5.
J Transl Med ; 22(1): 407, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689292

RESUMEN

BACKGROUND AND OBJECTIVE: Progranulin (PGRN), a multifunctional growth factor, plays indispensable roles in the regulation of cancer, inflammation, metabolic diseases, and neurodegenerative diseases. Nevertheless, its immune regulatory role in periodontitis is insufficiently understood. This study attempts to explore the regulatory effects of PGRN on macrophage polarization in periodontitis microenvironment. METHODS: Immunohistochemical (IHC) and multiplex immunohistochemical (mIHC) stainings were performed to evaluate the expression of macrophage-related markers and PGRN in gingival samples from periodontally healthy subjects and periodontitis subjects. RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were polarized towards M1 or M2 macrophages by the addition of LPS or IL-4, respectively, and were treated with or without PGRN. Real-time fluorescence quantitative PCR (qRT-PCR), immunofluorescence staining (IF), enzyme-linked immunosorbent assay (ELISA), and flow cytometry were used to determine the expressions of M1 and M2 macrophage-related markers. Co-immunoprecipitation was performed to detect the interaction between PGRN and tumor necrosis factor receptor 2 (TNFR2). Neutralizing antibody was used to block TNFR2 to confirm the role of TNFR2 in PGRN-mediated macrophage polarization. RESULTS: The IHC and mIHC staining of human gingival slices showed a significant accumulation of macrophages in the microenvironment of periodontitis, with increased expressions of both M1 and M2 macrophage markers. Meanwhile, PGRN was widely expressed in the gingival tissue of periodontitis and co-expressed mainly with M2 macrophages. In vitro experiments showed that in RAW264.7 cells and BMDMs, M1 markers (CD86, TNF-α, iNOS, and IL-6) substantially decreased and M2 markers (CD206, IL-10, and Arg-1) significantly increased when PGRN was applied to LPS-stimulated macrophages relatively to LPS stimulation alone. Besides, PGRN synergistically promoted IL-4-induced M2 markers expression, such as CD206, IL-10, and Arg1. In addition, the co-immunoprecipitation result showed the direct interaction of PGRN with TNFR2. mIHC staining further revealed the co-localization of PGRN and TNFR2 on M2 macrophages (CD206+). Blocking TNFR2 inhibited the regulation role of PGRN on macrophage M2 polarization. CONCLUSIONS: In summary, PGRN promotes macrophage M2 polarization through binding to TNFR2 in both pro- and anti-inflammatory periodontal microenvironments.


Asunto(s)
Polaridad Celular , Macrófagos , Periodontitis , Progranulinas , Receptores Tipo II del Factor de Necrosis Tumoral , Periodontitis/metabolismo , Periodontitis/patología , Macrófagos/metabolismo , Humanos , Animales , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Progranulinas/metabolismo , Ratones , Células RAW 264.7 , Encía/metabolismo , Encía/patología , Masculino , Femenino , Adulto , Activación de Macrófagos , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL
6.
Cancer Immunol Immunother ; 73(5): 76, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38554213

RESUMEN

BACKGROUND: Tumor microenvironment actually reduces antitumor effect against the immune attack by exclusion of CD8+T cells. Progranulin (PGRN) is a multifunctional growth factor with significant pathological effects in multiple tumors; however, its role in immunity evasion of breast cancer (BCa) is not completely understood. METHODS: We depleted GRN (PGRN gene) genetically in mice or specifically in PY8119 murine BCa cell line, and mouse models of orthotopic or subcutaneous transplantation were used. Chimeric mice-deficient of PGRN (Grn-/-) in bone marrow (BM) compartment was also generated. Association of PGRN expression with chemokine production or BCa development was investigated by histological and immunological assays. RESULTS: We found PGRN was involved in exhaustion of cytotoxic CD8+T cell in BCa with the increasing expressions of M2 markers and intercellular cell adhesion molecule-1 (ICAM-1) on macrophages. Specifically, ablation of PGRN in PY8119 cells reduced tumor burden, accompanied by the infiltrating of cytotoxic CD8+T cells into tumor nests. Moreover, our result revealed that blockade of PD-1 in PGRN-depleted tumors exhibited better antitumor effect in vivo and significantly decreased tumor burden. CONCLUSION: These findings suggest that inhibition of PGRN may act as a potential immune-therapeutic strategy by recovering infiltration of CD8+T cell in BCa tissue and thereby enhancing the response to anti-PD-1 therapy.


Asunto(s)
Molécula 1 de Adhesión Intercelular , Neoplasias , Animales , Ratones , Linfocitos T CD8-positivos , Línea Celular Tumoral , Molécula 1 de Adhesión Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Progranulinas/genética , Microambiente Tumoral
7.
Aging Clin Exp Res ; 36(1): 55, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38441695

RESUMEN

BACKGROUND: Progranulin is an anti-inflammatory protein that plays an essential role in the synapse function and the maintenance of neurons in the central nervous system (CNS). It has been shown that the CSF level of progranulin increases in Alzheimer's disease (AD) patients and is associated with the deposition of amyloid-beta (Aß) and tau in the brain tissue. In this study, we aimed to assess the longitudinal changes in cerebrospinal fluid (CSF) progranulin levels during different pathophysiological stages of AD and investigate associated AD pathologic features. METHODS: We obtained the CSF and neuroimaging data of 1001 subjects from the ADNI database. The participants were classified into four groups based on the A/T/N framework: A + /TN + , A + /TN-, A-/TN + , and A-/TN-. RESULTS: Based on our analysis there was a significant difference in CSF progranulin (P = 0.001) between ATN groups. Further ANOVA analysis revealed that there was no significant difference in the rate of change of CSF-progranulin ATN groups. We found that the rate of change of CSF progranulin was associated with baseline Aß-PET only in the A-/TN + group. A significant association was found between the rate of change of CSF progranulin and the Aß-PET rate of change only in A-/TN + CONCLUSION: Our findings revealed that an increase in CSF progranulin over time is associated with faster formation of Aß plaques in patients with only tau pathology based on the A/T/N classification (suspected non-Alzheimer's pathology). Together, our findings showed that the role of progranulin-related microglial activity on AD pathology can be stage-dependent, complicated, and more prominent in non-AD pathologic changes. Thus, there is a need for further studies to consider progranulin-based therapies for AD treatment.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides , Bases de Datos Factuales , Estudios Longitudinales , Progranulinas
8.
J Clin Periodontol ; 51(6): 774-786, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38462847

RESUMEN

AIM: To evaluate the effect of subgingival delivery of progranulin (PGRN)/gelatin methacryloyl (GelMA) complex as an adjunct to scaling and root planing (SRP) on an experimental periodontitis dog model with Class II furcation involvement (FI). MATERIALS AND METHODS: A Class II FI model was established, and the defects were divided into four treatment groups: (a) no treatment (control); (b) SRP; (c) SRP + GelMA; (d) SRP + PGRN/GelMA. Eight weeks after treatment, periodontal parameters were recorded, gingival crevicular fluid and gingival tissue were collected for ELISA and RT-qPCR, respectively, and mandibular tissue blocks were collected for micro computed tomography (micro-CT) scanning and hematoxylin and eosin (H&E) staining. RESULTS: The SRP + PGRN/GelMA group showed significant improvement in all periodontal parameters compared with those in the other groups. The expression of markers related to M1 macrophage and Th17 cell significantly decreased, and the expression of markers related to M2 macrophage and Treg cell significantly increased in the SRP + PGRN/GelMA group compared with those in the other groups. The volume, quality and area of new bone and the length of new cementum in the root furcation defects of the PGRN/GelMA group were significantly increased compared to those in the other groups. CONCLUSIONS: Subgingival delivery of the PGRN/GelMA complex could be a promising non-surgical adjunctive therapy for anti-inflammation, immunomodulation and periodontal regeneration.


Asunto(s)
Raspado Dental , Defectos de Furcación , Hidrogeles , Progranulinas , Animales , Perros , Defectos de Furcación/terapia , Hidrogeles/uso terapéutico , Raspado Dental/métodos , Inmunomodulación , Aplanamiento de la Raíz/métodos , Modelos Animales de Enfermedad , Periodontitis/terapia , Periodontitis/inmunología , Gelatina , Masculino , Microtomografía por Rayos X
9.
Alzheimers Res Ther ; 16(1): 66, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38539243

RESUMEN

BACKGROUND: Pathogenic heterozygous mutations in the progranulin gene (GRN) are a key cause of frontotemporal dementia (FTD), leading to significantly reduced biofluid concentrations of the progranulin protein (PGRN). This has led to a number of ongoing therapeutic trials aiming to treat this form of FTD by increasing PGRN levels in mutation carriers. However, we currently lack a complete understanding of factors that affect PGRN levels and potential variation in measurement methods. Here, we aimed to address this gap in knowledge by systematically reviewing published literature on biofluid PGRN concentrations. METHODS: Published data including biofluid PGRN concentration, age, sex, diagnosis and GRN mutation were collected for 7071 individuals from 75 publications. The majority of analyses (72%) had focused on plasma PGRN concentrations, with many of these (56%) measured with a single assay type (Adipogen) and so the influence of mutation type, age at onset, sex, and diagnosis were investigated in this subset of the data. RESULTS: We established a plasma PGRN concentration cut-off between pathogenic mutation carriers and non-carriers of 74.8 ng/mL using the Adipogen assay based on 3301 individuals, with a CSF concentration cut-off of 3.43 ng/mL. Plasma PGRN concentration varied by GRN mutation type as well as by clinical diagnosis in those without a GRN mutation. Plasma PGRN concentration was significantly higher in women than men in GRN mutation carriers (p = 0.007) with a trend in non-carriers (p = 0.062), and there was a significant but weak positive correlation with age in both GRN mutation carriers and non-carriers. No significant association was seen with weight or with TMEM106B rs1990622 genotype. However, higher plasma PGRN levels were seen in those with the GRN rs5848 CC genotype in both GRN mutation carriers and non-carriers. CONCLUSIONS: These results further support the usefulness of PGRN concentration for the identification of the large majority of pathogenic mutations in the GRN gene. Furthermore, these results highlight the importance of considering additional factors, such as mutation type, sex and age when interpreting PGRN concentrations. This will be particularly important as we enter the era of trials for progranulin-associated FTD.


Asunto(s)
Demencia Frontotemporal , Masculino , Humanos , Femenino , Progranulinas/genética , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Virulencia , Mutación/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética
10.
Clin Immunol ; 261: 109940, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38365048

RESUMEN

As the aging population increases, the focus on elderly patients with acute respiratory distress syndrome (ARDS) is also increasing. In this article, we found progranulin (PGRN) differential expression in ARDS patients and healthy controls, even in young and old ARDS patients. Its expression strongly correlates with several cytokines in both young and elderly ARDS patients. PGRN has comparable therapeutic effects in young and elderly mice with lipopolysaccharide-induced acute lung injury, manifesting as lung injury, apoptosis, inflammation, and regulatory T cells (Tregs) differentiation. Considering that Tregs differentiation relies on metabolic reprogramming, we discovered that Tregs differentiation was mediated by mitochondrial function, especially in the aged population. Furthermore, we demonstrated that PGRN alleviated the mitochondrial damage during Tregs differentiation through the AMPK/PGC-1α pathway in T cells. Collectively, PGRN may regulate mitochondria function to promote Tregs differentiation through the AMPK/PGC-1α pathway to improve ARDS.


Asunto(s)
Lesión Pulmonar Aguda , Síndrome de Dificultad Respiratoria , Humanos , Anciano , Ratones , Animales , Progranulinas/metabolismo , Progranulinas/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/farmacología , Linfocitos T Reguladores/metabolismo , Mitocondrias/metabolismo , Lesión Pulmonar Aguda/metabolismo
11.
Sci Total Environ ; 921: 171101, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38387595

RESUMEN

Recent epidemiological and animal studies have indicated that ambient fine particulate matter (PM2.5) exposure during pregnancy is closely associated with intrauterine growth restriction (IUGR). However, the underlying mechanisms remain to be revealed. In this study, we found that gestational exposure to PM2.5 significantly decreased fetal weight and crown-rump length in mice, accompanied by insufficient placental trophoblast syncytialization and increased expression of progranulin (PGRN) in mice placenta. Administering PGRN neutralizing antibody to pregnant mice alleviated growth restriction and insufficient placental trophoblast syncytialization caused by PM2.5, accompanied with suppressed activation of the mTOR signaling pathway. Furthermore, in vitro experiments using human placental BeWo cells showed that 10 µg·mL-1 PM2.5 activated PGRN/mTOR signaling and suppressed forskolin-induced cell fusion, which was blocked by knockdown of PGRN. Taken together, our results demonstrated that PM2.5 exposure during pregnancy inhibited placental trophoblast syncytialization by activating PGRN/mTOR signaling, leading to abnormal placental development and IUGR. This study reveals a novel mechanism underlying the developmental toxicity of PM2.5 exposure during pregnancy.


Asunto(s)
Placenta , Trofoblastos , Embarazo , Femenino , Humanos , Animales , Ratones , Placenta/metabolismo , Progranulinas/toxicidad , Progranulinas/metabolismo , Trofoblastos/metabolismo , Transducción de Señal , Desarrollo Fetal , Retardo del Crecimiento Fetal , Serina-Treonina Quinasas TOR/toxicidad , Serina-Treonina Quinasas TOR/metabolismo
12.
Neuroreport ; 35(5): 320-327, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38305117

RESUMEN

Sortilin is a single-pass type I transmembrane protein which can bind to various cargo proteins, regulating their surface location, secretion, or degradation in lysosomes. In our previous study, we found that sortilin can regulate progranulin expression by transporting it to lysosomes and reduce neuronal cell injury in hypoxia-ischemia, but the expression and function of sortilin in microglial cells during hypoxia-ischemia are unknown. The purpose of this study was to further investigate the function of sortilin in microglial cells and its effect on neuron cells. In rat BV2 microglial cells, sortilin was knocked down by lentivirus. After oxygen-glucose deprivation/reperfusion (OGD/R), expression of sortilin, progranulin (PGRN) and JNK pathway was detected by western blot, immunofluorescence was used to show the localization of PGRN, secretion of TNFα/IL-6 was measured by Elisa. Then co-culture microglial cells with neuron cells during hypoxia-ischemia and detected the neuron injury by CCK-8 and TUNEL. The expression of sortilin, mature and cleaved PGRN were all increased after OGD/R in microglial cells. Furthermore, sortilin inhibition accompany with less PGRN localization in lysosomes and more mature and less cleaved PGRN expression in microglial cells. Sortilin inhibition also can reduce the inflammatory response in microglial cells, but it does not alleviate neuronal injury in co-culture. This study demonstrated that sortilin can regulate the expression of PGRN and reduce the inflammatory response in microglial cells. However, only inhibiting sortilin in microglial cells did not have an impact on the survival of neurons during ischemia-hypoxia.


Asunto(s)
Microglía , Daño por Reperfusión , Ratas , Animales , Progranulinas/metabolismo , Técnicas de Cocultivo , Hipoxia/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Neuronas/metabolismo , Isquemia/metabolismo , Oxígeno/metabolismo , Glucosa/metabolismo , Daño por Reperfusión/metabolismo
13.
J Alzheimers Dis ; 98(2): 425-432, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38393901

RESUMEN

Background: Behavioral variant frontotemporal dementia (bvFTD) typically involves subtle changes in personality that can delay a timely diagnosis. Objective: Here, we report the case of a patient diagnosed of GRN-positive bvFTD at the age of 52 presenting with a 7-year history of narcissistic personality disorder, accordingly to DSM-5 criteria. Methods: The patient was referred to neurological and neuropsychological examination. She underwent 3 Tesla magnetic resonance imaging (MRI) and genetic studies. Results: The neuropsychological examination revealed profound deficits in all cognitive domains and 3T brain MRI showed marked fronto-temporal atrophy. A mutation in the GRN gene further confirmed the diagnosis. Conclusions: The present case documents an unusual onset of bvFTD and highlights the problematic nature of the differential diagnosis between prodromal psychiatric features of the disease and primary psychiatric disorders. Early recognition and diagnosis of bvFTD can lead to appropriate management and support for patients and their families. This case highlights the importance of considering neurodegenerative diseases, such as bvFTD, in the differential diagnosis of psychiatric disorders, especially when exacerbations of behavioral traits manifest in adults.


Asunto(s)
Demencia Frontotemporal , Femenino , Humanos , Demencia Frontotemporal/diagnóstico por imagen , Demencia Frontotemporal/genética , Trastorno de Personalidad Narcisista , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Imagen por Resonancia Magnética , Pruebas Neuropsicológicas , Progranulinas
14.
Ann Neurol ; 95(5): 917-928, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38356322

RESUMEN

OBJECTIVE: Triggering receptor expressed on myeloid cells-2 (TREM2) and progranulin (PGRN) are critical regulators of microglia activation and can be detected in cerebrospinal fluid (CSF). However, whether microglial reactivity is detrimental or neuroprotective for Alzheimer disease (AD) is still debatable. METHODS: We identified 663 participants with baseline ß-amyloid (Aß) positron emission tomography (PET) and CSF biomarker data, including phosphorylated tau181 (p-Tau181), soluble TREM2 (sTREM2), PGRN, and growth-associated protein-43 (GAP-43). Among them, 254 participants had concurrent longitudinal CSF biomarkers. We used multivariate regression analysis to study the associations of CSF microglial biomarkers with Aß PET, CSF p-Tau181, and CSF GAP-43 cross-sectionally and longitudinally. A Chinese aging cohort's independent CSF samples (n = 65) were analyzed as a validation. RESULTS: Higher baseline levels of CSF microglial biomarkers were related to faster rates of CSF sTREM2 increase and CSF PGRN decrease. Elevated CSF p-Tau181 was associated with higher levels of CSF microglial biomarkers and faster rates of CSF sTREM2 increase and CSF PGRN decrease. In both cohorts, higher Aß burden was associated with attenuated CSF p-Tau181 effects on CSF microglial biomarker increases. Independent of Aß PET and CSF p-Tau181 pathologies, higher levels of CSF sTREM2 but not CSF PGRN were related to elevated CSF GAP-43 levels and faster rates of CSF GAP-43 increase. INTERPRETATION: These findings suggest that higher Aß burden may attenuate the p-Tau-associated microglial responses, and TREM2-related microglial reactivity may independently correlate with GAP-43-related presynaptic loss. This study highlights the two-edged role of microglial reactivity in AD and other neurodegenerative diseases. ANN NEUROL 2024;95:917-928.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Glicoproteínas de Membrana , Microglía , Tomografía de Emisión de Positrones , Progranulinas , Receptores Inmunológicos , Proteínas tau , Humanos , Microglía/metabolismo , Masculino , Femenino , Péptidos beta-Amiloides/líquido cefalorraquídeo , Péptidos beta-Amiloides/metabolismo , Anciano , Proteínas tau/líquido cefalorraquídeo , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/diagnóstico por imagen , Persona de Mediana Edad , Receptores Inmunológicos/metabolismo , Progranulinas/líquido cefalorraquídeo , Glicoproteínas de Membrana/líquido cefalorraquídeo , Biomarcadores/líquido cefalorraquídeo , Anciano de 80 o más Años , Estudios Longitudinales , Estudios Transversales
15.
Clin Chim Acta ; 556: 117830, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38354999

RESUMEN

Protease inhibitors (PIs) are associated with an incidence of lipodystrophy among people living with HIV(PLHIV). Lipodystrophiesare characterised by the loss of adipose tissue. Evidence suggests that a patient's lipodystrophy phenotype is influenced by genetic mutation, age, gender, and environmental and genetic factors, such as single-nucleotide variants (SNVs). Pathogenic variants are considered to cause a more significant loss of adipose tissue compared to non-pathogenic. Lipid metabolising enzymes and transporter genes have a role in regulating lipoprotein metabolism and have been associated with lipodystrophy in HIV-infected patients (LDHIV). The long-term effect of the lipodystrophy syndrome is related to cardiovascular diseases (CVDs). Hence, we determined the SNVs of lipid metabolising enzymes and transporter genes in a total of 48 patient samples, of which 24 were with and 24 were without HIV-associated lipodystrophy (HIVLD) using next-generation sequencing. A panel of lipid metabolism, transport and elimination genes were sequenced. Three novel heterozygous non-synonymous variants at exon 8 (c.C1400A:p.S467Y, c.G1385A:p.G462E, and c.T1339C:p.S447P) in the ABCB6 gene were identified in patients with lipodystrophy. One homozygous non-synonymous SNV (exon5:c.T358C:p.S120P) in the GRN gene was identified in patients with lipodystrophy. One novelstop-gain SNV (exon5:c.C373T:p.Q125X) was found in the GRN gene among patients without lipodystrophy. Patients without lipodystrophy had one homozygous non-synonymous SNV (exon9:c.G1462T:p.G488C) in the ABCB6 gene. Our findings suggest that novel heterozygous non-synonymous variants in the ABCB6 gene may contribute to defective protein production, potentially intensifying the severity of lipodystrophy. Additionally, identifying a stop-gain SNV in the GRN gene among patients without lipodystrophy implies a potential role in the development of HIVLD.


Asunto(s)
Infecciones por VIH , Síndrome de Lipodistrofia Asociada a VIH , Lipodistrofia , Humanos , Síndrome de Lipodistrofia Asociada a VIH/genética , Síndrome de Lipodistrofia Asociada a VIH/complicaciones , Lipodistrofia/genética , Lipodistrofia/complicaciones , Lipodistrofia/epidemiología , Mutación , Tejido Adiposo , Lípidos , Infecciones por VIH/complicaciones , Infecciones por VIH/genética , Transportadoras de Casetes de Unión a ATP/genética , Progranulinas/genética
16.
J Neuroinflammation ; 21(1): 47, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38347588

RESUMEN

BACKGROUND: Progranulin (PGRN) haploinsufficiency due to progranulin gene (GRN) variants can cause frontotemporal dementia (FTD) with aberrant TAR DNA-binding protein 43 (TDP-43) accumulation. Despite microglial burden with TDP-43-related pathophysiology, direct microglial TDP-43 pathology has not been clarified yet, only emphasized in neuronal pathology. Thus, the objective of this study was to investigate TDP-43 pathology in microglia of patients with PGRN haploinsufficiency. METHODS: To design a human microglial cell model with PGRN haploinsufficiency, monocyte-derived microglia (iMGs) were generated from FTD-GRN patients carrying pathogenic or likely pathogenic variants (p.M1? and p.W147*) and three healthy controls. RESULTS: iMGs from FTD-GRN patients with PGRN deficiency exhibited severe neuroinflammation phenotype and failure to maintain their homeostatic molecular signatures, along with impaired phagocytosis. In FTD-GRN patients-derived iMGs, significant cytoplasmic TDP-43 aggregation and accumulation of lipid droplets with profound lysosomal abnormalities were observed. These pathomechanisms were mediated by complement C1q activation and upregulation of pro-inflammatory cytokines. CONCLUSIONS: Our study provides considerable cellular and molecular evidence that loss-of-function variants of GRN in human microglia can cause microglial dysfunction with abnormal TDP-43 aggregation induced by inflammatory milieu as well as the impaired lysosome. Elucidating the role of microglial TDP-43 pathology in intensifying neuroinflammation in individuals with FTD due to PGRN deficiency and examining consequential effects on microglial dysfunction might yield novel insights into the mechanisms underlying FTD and neurodegenerative disorders.


Asunto(s)
Demencia Frontotemporal , Enfermedad de Pick , Humanos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/genética , Haploinsuficiencia , Lisosomas/metabolismo , Microglía/patología , Enfermedades Neuroinflamatorias , Enfermedad de Pick/metabolismo , Progranulinas/genética , Progranulinas/metabolismo
17.
Nat Commun ; 15(1): 1434, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365772

RESUMEN

Comorbid proteinopathies are observed in many neurodegenerative disorders including Alzheimer's disease (AD), increase with age, and influence clinical outcomes, yet the mechanisms remain ill-defined. Here, we show that reduction of progranulin (PGRN), a lysosomal protein associated with TDP-43 proteinopathy, also increases tau inclusions, causes concomitant accumulation of α-synuclein and worsens mortality and disinhibited behaviors in tauopathy mice. The increased inclusions paradoxically protect against spatial memory deficit and hippocampal neurodegeneration. PGRN reduction in male tauopathy attenuates activity of ß-glucocerebrosidase (GCase), a protein previously associated with synucleinopathy, while increasing glucosylceramide (GlcCer)-positive tau inclusions. In neuronal culture, GCase inhibition enhances tau aggregation induced by AD-tau. Furthermore, purified GlcCer directly promotes tau aggregation in vitro. Neurofibrillary tangles in human tauopathies are also GlcCer-immunoreactive. Thus, in addition to TDP-43, PGRN regulates tau- and synucleinopathies via GCase and GlcCer. A lysosomal PGRN-GCase pathway may be a common therapeutic target for age-related comorbid proteinopathies.


Asunto(s)
Enfermedad de Alzheimer , Deficiencias en la Proteostasis , Tauopatías , Masculino , Humanos , Ratones , Animales , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Progranulinas , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo
18.
J Pediatr Endocrinol Metab ; 37(3): 280-288, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38253347

RESUMEN

OBJECTIVES: Neuronal ceroid lipofuscinosis type 11 (NCL11) is a rare disease that presents with progressive cognitive decline, epilepsy, visual impairment, retinal atrophy, cerebellar ataxia and cerebellar atrophy. We present herein a case of NCL11 in a patient diagnosed with neuromotor developmental delay, epilepsy, bronchiolitis obliterans and hypothyroidism. CASE PRESENTATION: A 4-year-old male patient was admitted to our clinic with global developmental delay and a medical history that included recurrent hospitalizations for pneumonia at the age of 17 days, and in months 4, 5 and 7. Family history revealed a brother with similar clinical findings (recurrent pneumonia, hypothyroidism, hypotonicity, swallowing dysfunction and neuromotor delay) who died from pneumonia at the age of 22 months. Computed tomography of the thorax was consistent with bronchiolitis obliterans, while epileptic discharges were identified on electroencephalogram with a high incidence of bilateral fronto-centro-temporal and generalized spike-wave activity but no photoparoxysmal response. Cranial MRI revealed T2 hyperintense areas in the occipital periventricular white matter and volume loss in the white matter, a thin corpus callosum and vermis atrophy. A whole-exome sequencing molecular analysis revealed compound heterozygous c.430G>A (p.Asp144Asn) and c.415T>C (p.Cys139Arg) variants in the GRN gene. CONCLUSIONS: The presented case indicates that NCL11 should be taken into account in patients with epilepsy and neurodegenerative diseases.


Asunto(s)
Bronquiolitis Obliterante , Epilepsia , Hipotiroidismo , Lipofuscinosis Ceroideas Neuronales , Neumonía , Masculino , Humanos , Recién Nacido , Lactante , Preescolar , Lipofuscinosis Ceroideas Neuronales/complicaciones , Lipofuscinosis Ceroideas Neuronales/diagnóstico , Lipofuscinosis Ceroideas Neuronales/genética , Epilepsia/genética , Atrofia , Progranulinas/genética
19.
J Med Genet ; 61(6): 543-548, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38228392

RESUMEN

BACKGROUND: METHODS: The GRN mutations, especially of the loss of function type, are causative of frontotemporal dementia (FTD). However, several GRN variants can be found in other neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease. So far, there have been over 300 GRN mutations reported globally. However, the genetic spectrum and phenotypic characteristics have not been fully elucidated in Chinese population.The participants were from the dementia cohort of Peking Union Medical College Hospital (n=1945). They received history inquiry, cognitive evaluation, brain imaging and exome sequencing. The dementia subjects carrying the rare variants of the GRN were included in this study. Those with the pathogenic or likely pathogenic variants of other dementia-related genes were excluded. RESULTS: 14 subjects carried the rare variants of GRN. They were clinically diagnosed with behavioural variant of FTD (n=2), non-fluent/agrammatic variant primary progressive aphasia (PPA, n=3), semantic variant PPA (n=1), AD (n=6) and mixed dementia (n=2). 13 rare variants of GRN were found, including 6 novel variants (W49X, S226G, M152I, A91E, G79E and A303S). The most prevalent symptom was amnesia (85.7%, 12/14), followed by psychiatric and behavioural disorder (78.6%, 11/14). In terms of lobar atrophy, temporal atrophy/hypometabolism was the most common (85.7%, 12/14), followed by parietal atrophy/hypometabolism (78.6%, 11/14). CONCLUSION: The novel GRN variants identified in this study contribute to enrich the GRN mutation repertoire. There is phenotypic similarity and diversity among Chinese patients with the GRN mutations.


Asunto(s)
Demencia Frontotemporal , Estudios de Asociación Genética , Mutación , Progranulinas , Humanos , Progranulinas/genética , Masculino , Femenino , Anciano , Persona de Mediana Edad , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Estudios de Cohortes , Demencia/genética , Demencia/patología , Demencia/epidemiología , Pueblo Asiatico/genética , Secuenciación del Exoma , Fenotipo , China/epidemiología , Predisposición Genética a la Enfermedad , Pueblos del Este de Asia
20.
BMC Med Genomics ; 17(1): 43, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38291418

RESUMEN

OBJECTIVE: To report the clinical manifestation and genetic characteristics of a patient having frontotemporal dementia (FTD) with abnormal behavior and unstable walking. METHODS: The clinical and imaging features of a patient who was eventually diagnosed with FTD were analyzed. The patient's neuropsychological, PET-CT, electromyography, and genetic data were collected. Furthermore, the patient's blood samples were examined for FTD-related genes. RESULTS: The patient was a 52-year-old man with hidden onset. The symptoms progressed gradually, presenting with abnormal behaviors, including repeated shopping, taking away other people's things, constantly eating snacks, and frequently calling friends at night. The patient also exhibited executive dysfunction, such as the inability to cook and multiple driving problems, e.g., constantly deviates from his lane while driving. In addition, the patient showed personality changes such as irritability, indifference, and withdrawal, as well as motor symptoms, including unstable walking and frequent falls when walking. Brain magnetic resonance imaging revealed hippocampal sclerosis along with widening and deepening of the bilateral temporal lobe sulcus. Brain metabolic imaging via PET-CT demonstrated decreased metabolism in the bilateral prefrontal lobe, with the abnormal energy metabolism indicating FTD. Lastly, blood sample analysis detected mutations in the amyotrophic lateral sclerosis (ALS)-related GRN gene c.1352C > T (p.P451L) and ErbB4 gene c.256 T > C (p.Y86H). CONCLUSION: This is the first case of heterozygous mutations in the GRN and ErbB4 genes in FTD alone. The GRN and ErbB4 genes are likely to be important in the pathogenesis of FTD, expanding the common genetic profile of ALS and FTD.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Masculino , Humanos , Persona de Mediana Edad , Demencia Frontotemporal/genética , Esclerosis Amiotrófica Lateral/genética , Tomografía Computarizada por Tomografía de Emisión de Positrones , Progranulinas/genética , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA