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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 430, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715084

RESUMEN

Passive immunotherapy with specific antibodies targeting Amyloid ß (Aß) peptide or tubulin-associated unit (tau) protein has emerged as a promising therapeutic approach in Alzheimer's disease (AD). However, in a recent phase III clinical study, Sperling et al. (N Engl J Med 10.1056/NEJMoa2305032, 2023) reported that solanezumab, a monoclonal antibody targeting Aß peptide, failed to slow cognitive decline in AD patients. Previously, three other anti-Aß antibodies, bapineuzumab, crenezumab, and gantenerumab, have also failed to show similar beneficial effects. In addition, three humanized antibodies targeting tau protein failed in their phase II trials. However, other anti-Aß antibodies, such as lecanemab (a humanized mAb binds to soluble Aß protofibrils), donanemab (a humanized mAb binds to insoluble, N-terminal truncated form of Aß peptides) and aducanumab (a human mAb binds to the aggregated form of Aß), have been shown to slow the decline of cognitive functions in early stage AD patients. The specific targets used in passive immunotherapy in these clinical trials may explain the divergent clinical outcomes. There are several challenges and limitations of passive immunotherapy using anti-Aß antibodies and long term longitudinal studies are needed to assess their efficacy, side effects and cost effectiveness in a wider spectrum of subjects, from pre-dementia to more advanced dementia. A combination therapeutic approach using both anti-Aß antibodies and other pharmaceutical agents should also be explored.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales/uso terapéutico , Inmunización Pasiva
2.
Can J Neurol Sci ; : 1-16, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38267254

RESUMEN

BACKGROUND: Genetic association studies have not produced consistent results in restless legs syndrome (RLS). OBJECTIVES: To conduct a systematic review on genetic association studies in RLS to highlight the common gene variants and ethnic differences. METHODOLOGY: We conducted Pubmed, Embase, and Cochrane search using terms "Genetic association studies" and "restless legs syndrome" for candidate gene-based studies. Out of the initial 43 studies, 18 case control studies (from 2012 to 2022) were included. Thirteen studies including 10794 Caucasian subjects (4984 RLS cases and 5810 controls) and five studies involving 2009 Asian subjects (796 RLS cases and 1213 controls) were tabulated and analyzed. In addition, three Genome-Wide Association Studies (GWAS) in Asians and Europeans/Caucasians were included for comparisons. RESULTS: In the Asian population, gene variants in BST1, SNCA Rep1, IL1B, BTBD9, and MAP2K5/SKOR1 increased the risk of RLS (odds ratio range 1.2-2.8). In Caucasian populations, examples of variants that were associated with an increased risk of RLS (odds ratio range 1.1-1.9) include those in GABRR3 TOX3, ADH1B, HMOX1, GLO1, DCDC2C, BTBD9, SKOR1, and SETBP1. Based on the meta-analysis of GWAS studies, the rs9390170 variant in UTRN gene was identified to be a novel genetic marker for RLS in Asian cohorts, whereas rs113851554 in MEIS1 gene was a strong genetic factor among the >20 identified gene variants for RLS in Caucasian populations. CONCLUSION: Our systemic review demonstrates that multiple genetic variants modulate risk of RLS in Caucasians (such as MEIS1 BTBD9, MAP2K5) and in Asians (such as BTBD9, MAP2K5, and UTRN).

3.
Int J Mol Sci ; 25(9)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38731862

RESUMEN

There are currently no disease-modifying therapies for Parkinson's disease (PD), a progressive neurodegenerative disorder associated with dopaminergic neuronal loss. There is increasing evidence that endogenous dopamine (DA) can be a pathological factor in neurodegeneration in PD. Tyrosine hydroxylase (TH) is the key rate-limiting enzyme for DA generation. Drugs that inhibit TH, such as alpha-methyltyrosine (α-MT), have recently been shown to protect against neurodegeneration in various PD models. DA receptor agonists can activate post-synaptic DA receptors to alleviate DA-deficiency-induced PD symptoms. However, DA receptor agonists have no therapeutic effects against neurodegeneration. Thus, a combination therapy with DA receptor agonists plus TH inhibitors may be an attractive therapeutic approach. TH inhibitors can protect and promote the survival of remaining dopaminergic neurons in PD patients' brains, whereas DA receptor agonists activate post-synaptic DA receptors to alleviate PD symptoms. Additionally, other PD drugs, such as N-acetylcysteine (NAC) and anticholinergic drugs, may be used as adjunctive medications to improve therapeutic effects. This multi-drug cocktail may represent a novel strategy to protect against progressive dopaminergic neurodegeneration and alleviate PD disease progression.


Asunto(s)
Agonistas de Dopamina , Enfermedad de Parkinson , Tirosina 3-Monooxigenasa , Animales , Humanos , Dopamina/metabolismo , Agonistas de Dopamina/uso terapéutico , Agonistas de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Quimioterapia Combinada , Inhibidores Enzimáticos/uso terapéutico , Inhibidores Enzimáticos/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Tirosina 3-Monooxigenasa/metabolismo
4.
Cell Mol Life Sci ; 79(12): 599, 2022 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-36409355

RESUMEN

BACKGROUND: Parkinson's disease (PD) is characterized by selective and progressive dopamine (DA) neuron loss in the substantia nigra and other brain regions, with the presence of Lewy body formation. Most PD cases are sporadic, whereas monogenic forms of PD have been linked to multiple genes, including Leucine kinase repeat 2 (LRRK2) and PTEN-induced kinase 1 (PINK1), two protein kinase genes involved in multiple signaling pathways. There is increasing evidence to suggest that endogenous DA and DA-dependent neurodegeneration have a pathophysiologic role in sporadic and familial PD. METHODS: We generated patient-derived dopaminergic neurons and human midbrain-like organoids (hMLOs), transgenic (TG) mouse and Drosophila models, expressing both mutant and wild-type (WT) LRRK2 and PINK1. Using these models, we examined the effect of LRRK2 and PINK1 on tyrosine hydroxylase (TH)-DA pathway. RESULTS: We demonstrated that PD-linked LRRK2 mutations were able to modulate TH-DA pathway, resulting in up-regulation of DA early in the disease which subsequently led to neurodegeneration. The LRRK2-induced DA toxicity and degeneration were abrogated by wild-type (WT) PINK1 (but not PINK1 mutations), and early treatment with a clinical-grade drug, α-methyl-L-tyrosine (α-MT), a TH inhibitor, was able to reverse the pathologies in human neurons and TG Drosophila models. We also identified opposing effects between LRRK2 and PINK1 on TH expression, suggesting that functional balance between these two genes may regulate the TH-DA pathway. CONCLUSIONS: Our findings highlight the vital role of the TH-DA pathway in PD pathogenesis. LRRK2 and PINK1 have opposing effects on the TH-DA pathway, and its balance affects DA neuron survival. LRRK2 or PINK1 mutations can disrupt this balance, promoting DA neuron demise. Our findings provide support for potential clinical trials using TH-DA pathway inhibitors in early or prodromic PD.


Asunto(s)
Proteínas de Drosophila , Enfermedad de Parkinson , Ratones , Animales , Humanos , Dopamina/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Enfermedad de Parkinson/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Ratones Transgénicos , Drosophila/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
5.
Int J Mol Sci ; 24(18)2023 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-37762040

RESUMEN

Neurodegenerative diseases are characterized by the progressive degeneration or death of neurons in the central or peripheral nervous system [...].


Asunto(s)
Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/terapia , Neuronas , Sistema Nervioso Periférico
6.
Hum Mol Genet ; 26(22): 4494-4505, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28973420

RESUMEN

Pathogenic leucine-rich repeat kinase 2 (LRRK2) mutations are recognized as the most common cause of familial Parkinson's disease in certain populations. Recently, LRRK2 mutations were shown to be associated with a higher risk of hormone-related cancers. However, how LRRK2 itself contributes to cancer risk remains unknown. DNA damage causes cancer, and DNA damage responses are among the most important pathways in cancer biology. To understand the role of LRRK2 in DNA damage response pathway, we induced DNA damage by applying genotoxic stress to the cells with Adriamycin. We found that DNA damage enhances LRRK2 phosphorylation at Serine 910, Serine 935 and Serine 1292. We further showed that LRRK2 phosphorylation is abolished in the absence of ATM, suggesting that LRRK2 phosphorylation requires ATM. It should also be noted that LRRK2 interacts with ATM. In contrast, overexpression or knockdown of LRRK2 does not affect ATM phosphorylation, indicating that LRRK2 is the downstream target of ATM in response to DNA damage. Moreover, we demonstrated that LRRK2 increases the expression of p53 and p21 by increasing the Mdm2 phosphorylation in response to DNA damage. Loss-of-function in LRRK2 has the opposite effect to that of LRRK2. In addition, FACS analysis revealed that LRRK2 enhances cell cycle progression into S phase in response to DNA damage, a finding that was confirmed by 5-bromo-2'-deoxyuridine immunostaining. Taken together, our findings demonstrate that LRRK2 plays an important role in the ATM-Mdm2-p53 pathway that regulates cell proliferation in response to DNA damage.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN/genética , Daño del ADN/fisiología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de Ciclo Celular/genética , División Celular/genética , Proliferación Celular/fisiología , Proteínas de Unión al ADN/genética , Doxorrubicina/farmacología , Células HEK293 , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células 3T3 NIH , Fosforilación , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
7.
Biochim Biophys Acta Mol Cell Res ; 1864(7): 1308-1317, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28433685

RESUMEN

Parkin/PINK1-mediated mitophagy is implicated in the pathogenesis of Parkinson's disease (PD). Prior to elimination of damaged mitochondria, Parkin translocates to mitochondria and induces mitochondrial clustering. While the mechanism of PINK1-dependent Parkin redistribution to mitochondria is now becoming clear, the role of mitochondrial clustering has been less well understood. In our study, we found that loss of p62 disrupted mitochondrial aggregation and specifically sensitized Parkin-expressing cells to apoptosis induced by mitochondrial depolarization. Notably, altering mitochondrial aggregation through regulating p62 or other methods was sufficient to affect such apoptosis. Moreover, disruption of mitochondrial aggregation promoted proteasome-dependent degradation of outer mitochondrial membrane (OMM) proteins. The accelerated degradation in turn facilitated cytochrome c release from mitochondria, leading to apoptosis. Together, our study demonstrates a protective role of mitochondrial clustering in mitophagy and helps in understanding how aggregation defends cells against stress.


Asunto(s)
Apoptosis , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Proteína Sequestosoma-1/metabolismo , Citocromos c/metabolismo , Células HEK293 , Células HeLa , Humanos , Membranas Mitocondriales/metabolismo , Mitofagia , Proteolisis , Proteína Sequestosoma-1/genética , Ubiquitina-Proteína Ligasas/metabolismo
8.
Hum Mol Genet ; 24(22): 6314-30, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26310625

RESUMEN

The mutations of F-box protein 7 (FBXO7) gene (T22M, R378G and R498X) are associated with a severe form of autosomal recessive juvenile-onset Parkinson's disease (PD) (PARK 15). Here we demonstrated that wild-type (WT) FBXO7 is a stress response protein and it can play both cytoprotective and neurotoxic roles. The WT FBXO7 protein is vital to cell mitophagy and can facilitate mitophagy to protect cells, whereas mutant FBXO7 inhibits mitophagy. Upon stress, the endogenous WT FBXO7 gets up-regulated, concentrates into mitochondria and forms FBXO7 aggregates in mitochondria. However, FBXO7 mutations aggravate deleterious FBXO7 aggregation in mitochondria. The FBXO7 aggregation and toxicity can be alleviated by Proline, glutathione (GSH) and coenzyme Q10, whereas deleterious FBXO7 aggregation in mitochondria can be aggravated by prohibitin 1 (PHB1), a mitochondrial protease inhibitor. The overexpression of WT FBXO7 could lead to FBXO7 protein aggregation and dopamine neuron degeneration in transgenic Drosophila heads. The elevated FBXO7 expression and aggregation were identified in human fibroblast cells from PD patients. FBXO7 can also form aggregates in brains of PD and Alzheimer's disease. Our study provides novel pathophysiologic insights and suggests that FBXO7 may be a potential therapeutic target in FBXO7-linked neuron degeneration in PD.


Asunto(s)
Proteínas F-Box/genética , Mutación , Trastornos Parkinsonianos/genética , Animales , Células Cultivadas , Drosophila , Proteínas F-Box/metabolismo , Femenino , Humanos , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mitofagia/genética , Trastornos Parkinsonianos/metabolismo , Embarazo , Prohibitinas , Agregado de Proteínas/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
9.
Hum Mol Genet ; 23(12): 3157-65, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24459295

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) are common causes of familial Parkinson's disease (PD). LRRK2 has been shown to bind peroxiredoxin-3 (PRDX3), the most important scavenger of hydrogen peroxide in the mitochondria, in vitro. Here, we examined the interactions of LRRK2 and PRDX3 in Drosophila models by crossing transgenic LRRK2 and PRDX3 flies. As proof of principle experiments, we subsequently challenged LRRK2 and LRRK2/PRDX3 flies with a peroxidase mimic, Ebselen. We demonstrated that co-expression of PRDX3 with the LRRK2 kinase mutant G2019S in bigenic Drosophila ameliorated the G2019S mutant-induced reduction in peroxidase capacity, loss of dopaminergic neurons, shortened lifespan and mitochondrial defects of flight muscles in monogenic flies expressing the G2019S alone. Challenges with Ebselen recapitulated similar rescue of these phenotypic features in mutant-expressing Drosophila. The peroxidase mimic preserved neuronal and mitochondrial and neuronal integrity and improved mobility and survival in mutant-expressing Drosophila. Taken together, our study provides the first in vivo evidence to suggest that phosphoinhibition of endogenous peroxidases could be a mechanism in LRRK2-induced oxidant-mediated neurotoxicity. Our therapeutic experiments also highlight the potential of thiol peroxidases as neuroprotective agents in PD patients carrying LRRK2 mutations.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Mitocondrias/metabolismo , Peroxiredoxina III/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Animales Modificados Genéticamente , Azoles/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Drosophila melanogaster/citología , Femenino , Humanos , Isoindoles , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mitocondrias/efectos de los fármacos , Mutación , Fármacos Neuroprotectores/farmacología , Compuestos de Organoselenio/farmacología , Peroxiredoxina III/genética , Fosforilación
10.
Ageing Res Rev ; 98: 102339, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38754634

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder that affects the cerebral cortex and hippocampus, and is characterised by progressive cognitive decline and memory loss. A recent report of a patient carrying a novel gain-of-function variant of RELN (H3447R, termed RELN-COLBOS) who developed resilience against presenilin-linked autosomal-dominant AD (ADAD) has generated enormous interest. The RELN-COLBOS variant enhances interactions with the apolipoprotein E receptor 2 (ApoER2) and very-low-density lipoprotein receptor (VLDLR), which are associated with delayed AD onset and progression. These findings were validated in a transgenic mouse model. Reelin is involved in neurodevelopment, neurogenesis, and neuronal plasticity. The evidence accumulated thus far has demonstrated that the Reelin pathway links apolipoprotein E4 (ApoE4), amyloid-ß (Aß), and tubulin-associated unit (Tau), which are key proteins that have been implicated in AD pathogenesis. Reelin and key components of the Reelin pathway have been highlighted as potential therapeutic targets and biomarkers for AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Apolipoproteína E4 , Moléculas de Adhesión Celular Neuronal , Proteínas de la Matriz Extracelular , Proteínas del Tejido Nervioso , Proteína Reelina , Serina Endopeptidasas , Proteínas tau , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Humanos , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Péptidos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Proteínas tau/genética , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Ratones
11.
Ageing Res Rev ; 99: 102381, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38914264

RESUMEN

BACKGROUND: Caffeine is one of the most consumed psychoactive substances globally. Caffeine-gene interactions in Parkinson's disease (PD) has not been systematically examined. OBJECTIVES: To conduct a systematic review on the interaction between caffeine consumption and genetic susceptibility to PD. METHODOLOGY: We conducted PubMed and Embase search using terms "Genetic association studies", "Caffeine", "polymorphism" and "Parkinson's disease", from inception till 2023. Of the initial 2391 studies, 21 case-control studies were included. The demographic, genetic and clinical data were extracted and analyzed. RESULTS: We identified 21 studies which involved a total of 607,074 study subjects and 17 gene loci (SNCA, MAPT, HLA-DRA, NOS1, NOS3, GBA, ApoE, BST1, ESR2, NAT2, SLC2A13, LRRK2, NOS2A, GRIN2A, CYP1A2, ESR1, ADORA2A) have been investigated for the effect of gene-caffeine interaction and PD risk. The genes were identified through PD GWAS or involved in caffeine or related metabolism pathways. Based on the genetic association and interaction studies, only MAPT, SLC2A13, LRRK2, ApoE, NOS2A, GRIN2A, CYP1A2, and ADORA2A have been shown by at least one study to have a positive caffeine-gene interaction influencing the risk of PD. CONCLUSION: Studies have shown an interaction between caffeine with genetic variants of MAPT, SLC2A13, LRRK2, ApoE, NOS2A, GRIN2A, CYP1A2, and ADORA2A in modulating the risk of PD. Due to the potential limitations of these discovery/pilot studies, further independent replication studies are needed. Better designed genetic association studies in multi-ancestry and admixed cohorts to identify potential shared or unique multivariate gene-environmental interactions, as well as functional studies of gene-caffeine interactions will be useful.


Asunto(s)
Cafeína , Predisposición Genética a la Enfermedad , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/genética , Interacción Gen-Ambiente
12.
Zhongguo Zhong Yao Za Zhi ; 38(7): 1056-60, 2013 Apr.
Artículo en Zh | MEDLINE | ID: mdl-23847957

RESUMEN

Achyranthes bidentata polysaccharides (ABPS) was extracted from the root of A. bidentata. Dendritic cells (DC), which were stimulated with ABPS and/or tumor antigen SW480, were co-cultured with cytokine induced killer cells (CIK) to test the cytotoxic effect on colon cancer cell line SW480. Peripheral blood mononuclear cells (PBMNCs) which were separated from human peripheral blood were cultured to DC and CIK separately. (1) DC were divided into four groups: pure DC served as control group; ABPS (50 mg x L(-1)) stimulated DC served as experimental group; SW480 tumor antigen stimulated DC served as the second experimental group; ABPS (50 mg x L(-1)) and SW480 tumor antigen co-stimulated DC served as the third experimental group. Flow cytometry was used to detect the difference of the positive rate of molecules in the cell surface of DC, include CD80, CD86, CD1c, CD40, HLA-DR (6 samples for each group). (2) The four DC groups were mixed with CIK at the ratio 1:5 and acted as effect cells (DC + CIK groups), and the colon cancer cell line SW480 acted as target cells. The effect cells and the target cells were mixed together at the ratio 30: 1, 20:1 and 10:1 separately, and the CCK-8 kit was used to test the cytotoxic effect on colon cancer cell line SW480. (3) At the mixing ratio 30:1 of effect cells and target cells, ELISA was used to test the level of cytokines secretion, including IL-2, IL-12p70, IL-17 and TNF-alpha, in the liquid supernatant of every test group (3 duplication per sample). The results showed as following: (1) The positive rates of CD80, CD11c, HLA-DR, in the cell surface of DC which was co-stimulated by ABPS (50 mg x L(-1)) and SW480 tumor antigen, were obviously higher than the other DC groups (P < 0.05), and the positive rates of CD86, CD40 were obviously higher than the pure DC group (P < 0.05), and there was no remarkable difference with the other two DC groups. (2) At the mixing ratio 30:1, 20:1 and 10:1 of the effect cells and the target cells, the cytotoxic effect of ABPS stimulated DC + CIK group and SW480 tumor antigen stimulated DC + CIK group was obviously higher than DC + CIK group (P < 0.05), the cytotoxic effect of ABPS and SW480 tumor antigen co-stimulated DC + CIK group was obviously higher than all the other groups. (3) At the mixing ratio 30:1 of the effect cells and the target cells, the secretion levels of IL-12p70 and TNF-alpha in the liquid supernatant of the ABPS and SW480 tumor antigen co-stimulated DC + CIK group were obviously higher than all the other groups (P < 0.05), the secretion levels of IL-2 and IL-17 in the liquid supernatant of every test group have no remarkable difference. The cytotoxic effect of ABPS stimulated DC + CIK on SW480 was obviously increased. The cytotoxic effect of ABPS and SW480 tumor antigen co-stimulated DC + CIK group was obviously higher than all the other.


Asunto(s)
Achyranthes/química , Neoplasias del Colon/inmunología , Células Asesinas Inducidas por Citocinas/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Polisacáridos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/fisiopatología , Células Asesinas Inducidas por Citocinas/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Interferón gamma/inmunología , Interleucina-2/inmunología
13.
Cell Rep Med ; 4(6): 101075, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37343522

RESUMEN

Neel et al. identified pathophysiologic clues linking gasdermin-E (GSDME) with frontotemporal dementia and amyotrophic lateral sclerosis.1 Therapeutic studies targeting GSDME may provide a viable approach for neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Enfermedades Neurodegenerativas , Humanos , Gasderminas
14.
Transl Neurodegener ; 12(1): 44, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37718439

RESUMEN

A pathological feature of Parkinson's disease (PD) is the progressive loss of dopaminergic neurons and decreased dopamine (DA) content in the substantia nigra pars compacta in PD brains. DA is the neurotransmitter of dopaminergic neurons. Accumulating evidence suggests that DA interacts with environmental and genetic factors to contribute to PD pathophysiology. Disturbances of DA synthesis, storage, transportation and metabolism have been shown to promote neurodegeneration of dopaminergic neurons in various PD models. DA is unstable and can undergo oxidation and metabolism to produce multiple reactive and toxic by-products, including reactive oxygen species, DA quinones, and 3,4-dihydroxyphenylacetaldehyde. Here we summarize and highlight recent discoveries on DA-linked pathophysiologic pathways, and discuss the potential protective and therapeutic strategies to mitigate the complications associated with DA.


Asunto(s)
Dopamina , Enfermedad de Parkinson , Humanos , Encéfalo , Neuronas Dopaminérgicas
15.
Nat Rev Neurol ; 18(3): 145-157, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35022573

RESUMEN

Non-coding CGG repeat expansions cause multiple neurodegenerative disorders, including fragile X-associated tremor/ataxia syndrome, neuronal intranuclear inclusion disease, oculopharyngeal myopathy with leukodystrophy, and oculopharyngodistal myopathy. The underlying genetic causes of several of these diseases have been identified only in the past 2-3 years. These expansion disorders have substantial overlapping clinical, neuroimaging and histopathological features. The shared features suggest common mechanisms that could have implications for the development of therapies for this group of diseases - similar therapeutic strategies or drugs may be effective for various neurodegenerative disorders induced by non-coding CGG expansions. In this Review, we provide an overview of clinical and pathological features of these CGG repeat expansion diseases and consider the likely pathological mechanisms, including RNA toxicity, CGG repeat-associated non-AUG-initiated translation, protein aggregation and mitochondrial impairment. We then discuss future research needed to improve the identification and diagnosis of CGG repeat expansion diseases, to improve modelling of these diseases and to understand their pathogenesis. We also consider possible therapeutic strategies. Finally, we propose that CGG repeat expansion diseases may represent manifestations of a single underlying neuromyodegenerative syndrome in which different organs are affected to different extents depending on the gene location of the repeat expansion.


Asunto(s)
Síndrome del Cromosoma X Frágil , Enfermedades Neurodegenerativas , Ataxia/genética , Ataxia/patología , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Temblor/genética , Temblor/patología , Expansión de Repetición de Trinucleótido/genética
16.
Sci Signal ; 15(748): eabk3411, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35998231

RESUMEN

Gain-of-function mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are common in familial forms of Parkinson's disease (PD), which is characterized by progressive neurodegeneration that impairs motor and cognitive function. We previously demonstrated that LRRK2-mediated phosphorylation of ß-amyloid precursor protein (APP) triggers the production and nuclear translocation of the APP intracellular domain (AICD). Here, we connected LRRK2 to AICD in a feed-forward cycle that enhanced LRRK2-mediated neurotoxicity. In cooperation with the transcription factor FOXO3a, AICD promoted LRRK2 expression, thus increasing the abundance of LRRK2 that promotes AICD activation. APP deficiency in LRRK2G2019S mice suppressed LRRK2 expression, LRRK2-mediated mitochondrial dysfunction, α-synuclein accumulation, and tyrosine hydroxylase (TH) loss in the brain, phenotypes associated with toxicity and loss of dopaminergic neurons in PD. Conversely, AICD overexpression increased LRRK2 expression and LRRK2-mediated neurotoxicity in LRRK2G2019S mice. In LRRK2G2019S mice or cultured dopaminergic neurons from LRRK2G2019S patients, treatment with itanapraced reduced LRRK2 expression and was neuroprotective. Itanapraced showed similar effects in a neurotoxin-induced PD mouse model, suggesting that inhibiting the AICD may also have therapeutic benefits in idiopathic PD. Our findings reveal a therapeutically targetable, feed-forward mechanism through which AICD promotes LRRK2-mediated neurotoxicity in PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Enfermedad de Parkinson , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Neuronas Dopaminérgicas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones , Mutación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo
17.
IUBMB Life ; 62(8): 618-23, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20681026

RESUMEN

Notch signaling pathway, originally discovered in Drosophila, impinges on a wide array of cellular processes including cell proliferation, cell differentiation, and cell apoptosis. Recent accumulating evidences implicated the important roles of Notch signal pathway in different aspects of stem cell biology of neural stem cell (NSC). In vivo and in vitro studies illustrated that Notch signal pathway could promote gliogenesis, inhibit premature neurogenesis, and be involved in self-renew of NSC. This short review summarizes the roles of the Notch signaling pathway on gliogenesis, neurogenesis, and self-renew of NSC and their underlying molecular mechanisms.


Asunto(s)
Neuronas/citología , Receptores Notch , Células Madre/fisiología , Animales , Humanos
18.
Neurochem Res ; 35(11): 1805-18, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20721623

RESUMEN

In this study we demonstrate for the first time that short-lived intermediate glutathione (GSH) conjugates (5-S-GSH-DA-o-quinone and 2-S-GSH-DA-o-quinone) must have first formed when GSH reacted with dopamine (DA)-derived DA-o-quinones without enzymatic catalysis in solutions. These intermediate GSH-conjugates are unstable and would finally transform into reactive or non-reactive GSH-conjugates dependent on ambient reductive forces. We demonstrated that, under sufficient reductive force, the intermediate GSH-conjugates could be reduced and transform into non-reactive 5-S-GSH-DA and 2-S-GSH-DA. However, under insufficient reductive forces, the intermediate GSH-conjugates could cyclize spontaneously to form reactive 7-S-GSH-aminochrome (7-S-GSH-AM). The 7-S-GSH-AM is so reactive and toxic that it could further conjugate with another GSH to form non-reactive 4,7-bi-GSH-5,6-dihydroindole in solutions. Furthermore 7-S-GSH-AM could abrogate tyrosinase activity rapidly and even inhibit proteasome activity in solutions. However, 7-S-GSH-AM could undergo automatically internal rearrangement and transform into non-reactive 7-S-GSH-5,6-dihydroindole if it had not conjugated with GSH. Therefore, insufficient ambient reductive force, such as decreased GSH concentration, could lead to decreased GSH detoxification efficiency for toxic DA quinones. Based on findings in this study, we propose two potential detrimental positive feedback loops involving accelerated DA oxidation, increased GSH consumption and impaired GSH detoxification efficiency, as the potential underlying chemical explanation for dopaminergic neuron degeneration in Parkinson's disease.


Asunto(s)
Dopamina/análogos & derivados , Glutatión/análogos & derivados , Glutatión/química , Indoles/síntesis química , Dopamina/química , Retroalimentación , Glutatión/síntesis química , Glutatión/metabolismo , Indoles/química , Monofenol Monooxigenasa/antagonistas & inhibidores , Monofenol Monooxigenasa/metabolismo , Oxidación-Reducción
19.
Ageing Res Rev ; 62: 101107, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32535274

RESUMEN

Mitochondrial impairment is associated with progressive dopamine (DA) neuron degeneration in Parkinson's disease (PD). Recent findings highlight that Sirtuin-3 (SIRT3), a mitochondrial protein, is an oxidized nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase and a key modulator in maintaining integrity and functions of mitochondria. SIRT3 plays vital roles in regulation of mitochondrial functions, including mitochondrial ATP generation and energy metabolism, anti-oxidant defense, and cell death and proliferation. SIRT3 can deacetylate the transcriptional factors and crosstalk with different signaling pathways to cooperatively modulate mitochondrial functions and regulate defensive mitochondrial quality control (QC) systems. Down-regulated NAD+ level and decreased SIRT3 activity are related to aging process and has been pathologically linked to PD pathogenesis. Further, SIRT3 can bind and deacetylate PTEN-induced kinase 1 (PINK1) and PD protein 2 E3 ubiquitin protein ligase (Parkin) to facilitate mitophagy. Leucine Rich Repeat Kinase 2 (LRRK2)-G2019S mutation in PD is linked to SIRT3 impairment. Furthermore, SIRT3 is inversely associated with α-synuclein aggregation and DA neuron degeneration in PD. SIRT3 chemical activators and NAD+ precursors can up-regulate SIRT3 activity to protect against DA neuron degeneration in PD models. Taken together, SIRT3 is a promising PD therapeutic target and studies of SIRT3 functional modulators with neuroprotective capability will be of clinical interest.


Asunto(s)
Enfermedad de Parkinson , Humanos , Mitocondrias , NAD , Enfermedad de Parkinson/tratamiento farmacológico , Sirtuina 3/genética , alfa-Sinucleína
20.
Mol Brain ; 13(1): 164, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33261640

RESUMEN

Vacuolar protein sorting 35 (VPS35) is a major component of the retromer complex that mediates the retrograde transport of cargo proteins from endosomes to the trans-Golgi network. Mutations such as D620N in the VPS35 gene have been identified in patients with autosomal dominant Parkinson's disease (PD). However, it remains poorly understood whether and how VPS35 deficiency or mutation contributes to PD pathogenesis; specifically, the studies that have examined VPS35 thus far have differed in results and methodologies. We generated a VPS35 D620N mouse model using a Rosa26-based transgene expression platform to allow expression in a spatial manner, so as to better address these discrepancies. Here, aged (20-months-old) mice were first subjected to behavioral tests. Subsequently, DAB staining analysis of substantia nigra (SN) dopaminergic neurons with the marker for tyrosine hydroxylase (TH) was performed. Next, HPLC was used to determine dopamine levels, along with levels of its two metabolites, 3,4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), in the striatum. Western blotting was also performed to study the levels of key proteins associated with PD. Lastly, autoradiography (ARG) evaluation of [3H]FE-PE2I binding to the striatal dopamine transporter DAT was carried out. We found that VPS35 D620N Tg mice displayed a significantly higher dopamine level than NTg counterparts. All results were then compared with that of current VPS35 studies to shed light on the disease pathogenesis. Our model allows future studies to explicitly control spatial expression of the transgene which would generate a more reliable PD phenotype.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteínas de Transporte Vesicular/genética , Envejecimiento/patología , Animales , Autorradiografía , Conducta Animal , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Femenino , Metaboloma , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA