Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38804841

RESUMEN

Cisplatin is widely employed in tumor chemotherapy, but nephrotoxicity is an unavoidable side effect of cisplatin. Several studies have demonstrated that mesenchymal stromal cells (MSCs) ameliorate cisplatin-induced kidney injury, but the underlying mechanisms are unknown. In this study, the cisplatin-induced kidney injury mouse model was established by subjecting a single intraperitoneal injection with cisplatin. One hour before cisplatin injection, the mice received human bone marrow MSCs (hBM-MSCs) with or without siRNA-transfection, recombinant human tumor necrosis factor (TNF)-α-stimulated gene/protein 6 (rhTSG-6), or PBS through tail vein. In addition, cisplatin-stimulated HK-2 cells were treated with hBM-MSCs or rhTSG-6. hBM-MSCs treatment remarkably ameliorated cisplatin-induced acute and chronic kidney injury, as evidenced by significant reductions in serum creatinine (Scr), blood urea nitrogen (BUN), tubular injury, collagen deposition, α-smooth muscle actin accumulation, as well as inflammatory responses, and by remarkable increased anti-inflammatory factor expression and Treg cells infiltration in renal tissues. Furthermore, we found that only a few hBM-MSCs engrafted into damaged kidney and that the level of human TSG-6 in serum of mice increased significantly following hBM-MSCs administration. Moreover, hBM-MSCs significantly increased the viability of damaged HK-2 cells and decreased the levels of inflammatory cytokines in the culture supernatant. However, knockdown of TSG-6 gene in hBM-MSCs significantly attenuated their beneficial effects in vivo and in vitro. On the contrary, treated with rhTSG-6 achieved similar beneficial effects of hBM-MSCs. Our results indicate that systemic administration of hBM-MSCs alleviate cisplatin-induced acute and chronic kidney injury in part by paracrine TSG-6 secretion.

2.
Neuropathol Appl Neurobiol ; 48(3): e12791, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35067965

RESUMEN

AIMS: Our understanding of the pathological interactions between amyloidosis and tauopathy in Alzheimer's disease is incomplete. We sought to determine if the relative timing of the amyloidosis and tauopathy is critical for amyloid-enhanced tauopathy. METHODS: We crossed an inducible tauopathy model with two ß-amyloid models utilising the doxycycline-repressible transgenic system to modulate timing and duration of human tau expression in the context of amyloidosis and then assessed tauopathy, amyloidosis and gliosis. RESULTS: We combined inducible rTg4510 tau with APPswe/PS1dE9 [Line 85 (L85)] mice to examine the interactions between Aß and tauopathy at different stages of amyloidosis. When we initially suppressed mutant human tau expression for 14-15 months and subsequently induced tau expression for 6 months, severe amyloidosis with robust tauopathy resulted in rTg4510/L85 but not rTg4510 mice. When we suppressed mutant tau for 7 months before inducing expression for a subsequent 6 months in another cohort of rTg4510/L85 and rTg4510 mice, only rTg4510/L85 mice displayed robust tauopathy. Lastly, we crossed rTg4510 mice to tet-regulated APPswe/ind [Line 107 (L107)] mice, using doxycycline to initially suppress both transgenes for 1 month before inducing expression for 5 months to model early amyloidosis. In contrast to rTg4510, rTg4510/L107 mice rapidly developed amyloidosis, accompanied by robust tauopathy. CONCLUSIONS: These data suggest that tau misfolding is exacerbated by both newly forming Aß deposits in younger brain and mature deposits in older brains. Refined use and repurposing of these models provide new tools to explore the intersection of ageing, amyloid and tauopathy and to test interventions to disrupt the amyloid cascade.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Anciano , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Tauopatías/patología , Proteínas tau/genética , Proteínas tau/metabolismo
3.
Neuropathol Appl Neurobiol ; 48(2): e12779, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34825397

RESUMEN

AIMS: To illuminate the pathological synergy between Aß and tau leading to emergence of neurofibrillary tangles (NFT) in Alzheimer's disease (AD), here, we have performed a comparative neuropathological study utilising three distinctive variants of human tau (WT tau, P301L mutant tau and S320F mutant tau). Previously, in non-transgenic mice, we showed that WT tau or P301L tau does not form NFT while S320F tau can spontaneously aggregate into NFT, allowing us to test the selective vulnerability of these different tau conformations to the presence of Aß plaques. METHODS: We injected recombinant AAV-tau constructs into neonatal APP transgenic TgCRND8 mice or into 3-month-old TgCRND8 mice; both cohorts were aged 3 months post injection. This allowed us to test how different tau variants synergise with soluble forms of Aß (pre-deposit cohort) or with frank Aß deposits (post-deposit cohort). RESULTS: Expression of WT tau did not produce NFT or altered Aß in either cohort. In the pre-deposit cohort, S320F tau induced Aß plaque deposition, neuroinflammation and synaptic abnormalities, suggesting that early tau tangles affect the amyloid cascade. In the post-deposit cohort, contemporaneous expression of S320F tau did not exacerbate amyloid pathology, showing a dichotomy in Aß-tau synergy based on the nature of Aß. P301L tau produced NFT-type inclusions in the post-deposit cohort, but not in the pre-deposit cohort, indicating pathological synergy with pre-existing Aß deposits. CONCLUSIONS: Our data show that different tau mutations representing specific folding variants of tau synergise with Aß to different extents, depending on the presence of cerebral deposits.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Ovillos Neurofibrilares/patología , Placa Amiloide/patología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/metabolismo , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Neuronas/metabolismo , Neuronas/patología , Placa Amiloide/metabolismo
4.
FASEB J ; 34(8): 10590-10604, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32557852

RESUMEN

Psoriasis is a chronic relapsing inflammatory skin disease, affecting up to 3% of the global population. Accumulating evidence suggests that the complement system is involved in its pathogenesis. Our previous study revealed that the C5a/C5aR1 pathway is crucial for disease development. However, the underlying mechanisms remain largely unknown. To explore potential mechanisms, psoriatic skin lesions and histological changes were assessed following imiquimod (IMQ) cream treatment. Inflammatory cytokine expression was tested by real-time RT-PCR. Immunohistochemistry and flow cytometry were used to identify inflammatory cell infiltration and interleukin (IL-17A) IL-17A expression. A C5aR1 antagonist (C5aR1a) and PI3K inhibitor (wortmannin) were used for blocking experiments (both in vivo and in vitro) to explore the mechanism. C5a/C5aR1-pathway inhibition significantly attenuated psoriasis-like skin lesions with decreased epidermal hyperplasia, downregulated type 17-related inflammatory gene expression, and reduced IL-17A-producing γδ-T cell responses. Mechanistically, C5a/C5aR1 promoted the latter phenotype via PI3K-Akt signaling. Consistently, C5aR1 deficiency clearly ameliorated IMQ-induced chronic psoriasiform dermatitis, with a significant decrease in IL-17A expression. Finally, blocking C5aR1 signaling further decreased psoriasiform skin inflammation in IL-17-deficient mice. Results suggest that C5a/C5aR1 mediates experimental psoriasis and skin inflammation by upregulating IL-17A expression from γδ-T cells. Blocking C5a/C5aR1/IL-17A axis is expected to be a promising strategy for psoriasis treatment.


Asunto(s)
Inflamación/metabolismo , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Piel/metabolismo , Animales , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Imiquimod/farmacología , Inflamación/tratamiento farmacológico , Linfocitos Intraepiteliales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Psoriasis/tratamiento farmacológico , Psoriasis/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Piel/efectos de los fármacos
5.
J Cell Mol Med ; 24(20): 11936-11948, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32881263

RESUMEN

Sepsis-associated acute kidney injury (SA-AKI) is a common clinical critical care syndrome. It has received increasing attention due to its high morbidity and mortality; however, its pathophysiological mechanisms remain elusive. LIGHT, the 14th member of the tumour necrosis factor (TNF) superfamily and a bidirectional immunoregulatory molecule that regulates inflammation, plays a pivotal role in disease pathogenesis. In this study, mice with an intraperitoneal injection of LPS and HK-2 cells challenged with LPS were employed as a model of SA-AKI in vivo and in vitro, respectively. LIGHT deficiency notably attenuated kidney injury in pathological damage and renal function and markedly mitigated the inflammatory reaction by decreasing inflammatory mediator production and inflammatory cell infiltration in vivo. The TLR4-Myd88-NF-κB signalling pathway in the kidney of LIGHT knockout mice was dramatically down-regulated compared to the controls. Recombinant human LIGHT aggravated LPS-treated HK-2 cell injury by up-regulating the expression of the TLR4-Myd88-NF-κB signalling pathway and inflammation levels. TAK 242 (a selective TLR4 inhibitor) reduced this trend to some extent. In addition, blocking LIGHT with soluble receptor fusion proteins HVEM-Fc or LTßR-Fc in mice attenuated renal dysfunction and pathological damage in SA-AKI. Our findings indicate that LIGHT aggravates inflammation and promotes kidney damage in LPS-induced SA-AKI via the TLR4-Myd88-NF-κB signalling pathway, which provide potential strategies for the treatment of SA-AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Sepsis/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Lesión Renal Aguda/patología , Animales , Línea Celular , Regulación hacia Abajo , Humanos , Inflamación/patología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Ratones Noqueados , Modelos Biológicos , Análisis de Supervivencia , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia
6.
Plant Physiol ; 177(3): 1254-1266, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29784768

RESUMEN

Boron (B) alleviates aluminum (Al) toxicity in higher plants; however, the underlying mechanisms behind this phenomenon remain unknown. Here, we used bromocresol green pH indicator, noninvasive microtest, and microelectrode ion flux estimation techniques to demonstrate that B promotes root surface pH gradients in pea (Pisum sativum) roots, leading to alkalization in the root transition zone and acidification in the elongation zone, while Al inhibits these pH gradients. B significantly decreased Al accumulation in the transition zone (∼1.0-2.5 mm from the apex) of lateral roots, thereby alleviating Al-induced inhibition of root elongation. Net indole acetic acid (IAA) efflux detected by an IAA-sensitive platinum microelectrode showed that polar auxin transport, which peaked in the root transition zone, was inhibited by Al toxicity, while it was partially recovered by B. Electrophysiological experiments using the Arabidopsis (Arabidopsis thaliana) auxin transporter mutants (auxin resistant1-7; pin-formed2 [pin2]) and the specific polar auxin transporter inhibitor1-naphthylphthalamic acid showed that PIN2-based polar auxin transport is involved in root surface alkalization in the transition zone. Our results suggest that B promotes polar auxin transport driven by the auxin efflux transporter PIN2 and leads to the downstream regulation of the plasma membrane-H+-ATPase, resulting in elevated root surface pH, which is essential to decrease Al accumulation in this Al-targeted apical root zone. These findings provide a mechanistic explanation for the role of exogenous B in alleviation of Al accumulation and toxicity in plants.


Asunto(s)
Aluminio/toxicidad , Boro/farmacología , Ácidos Indolacéticos/metabolismo , Pisum sativum/efectos de los fármacos , Raíces de Plantas/efectos de los fármacos , Aluminio/farmacocinética , Arabidopsis/efectos de los fármacos , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Transporte Biológico/efectos de los fármacos , Membrana Celular/metabolismo , Concentración de Iones de Hidrógeno , Mutación , Pisum sativum/metabolismo , Ftalimidas/farmacología , Proteínas de Plantas/metabolismo , Raíces de Plantas/química , Raíces de Plantas/metabolismo , ATPasas de Translocación de Protón/metabolismo
7.
J Cell Sci ; 129(9): 1892-901, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27026526

RESUMEN

The capacity of the cell to produce, fold and degrade proteins relies on components of the proteostasis network. Multiple types of insults can impose a burden on this network, causing protein misfolding. Using thermal stress, a classic example of acute proteostatic stress, we demonstrate that ∼5-10% of the soluble cytosolic and nuclear proteome in human HEK293 cells is vulnerable to misfolding when proteostatic function is overwhelmed. Inhibiting new protein synthesis for 30 min prior to heat-shock dramatically reduced the amount of heat-stress induced polyubiquitylation, and reduced the misfolding of proteins identified as vulnerable to thermal stress. Following prior studies in C. elegans in which mutant huntingtin (Q103) expression was shown to cause the secondary misfolding of cytosolic proteins, we also demonstrate that mutant huntingtin causes similar 'secondary' misfolding in human cells. Similar to thermal stress, inhibiting new protein synthesis reduced the impact of mutant huntingtin on proteostatic function. These findings suggest that newly made proteins are vulnerable to misfolding when proteostasis is disrupted by insults such as thermal stress and mutant protein aggregation.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteína Huntingtina/metabolismo , Mutación Missense , Biosíntesis de Proteínas , Deficiencias en la Proteostasis/metabolismo , Sustitución de Aminoácidos , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Células HEK293 , Humanos , Proteína Huntingtina/genética , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/patología
8.
Acta Neuropathol ; 136(6): 919-938, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30140941

RESUMEN

The deposition of pathologic misfolded proteins in neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, frontotemporal dementia and amyotrophic lateral sclerosis is hypothesized to burden protein homeostatic (proteostatic) machinery, potentially leading to insufficient capacity to maintain the proteome. This hypothesis has been supported by previous work in our laboratory, as evidenced by the perturbation of cytosolic protein solubility in response to amyloid plaques in a mouse model of Alzheimer's amyloidosis. In the current study, we demonstrate changes in proteome solubility are a common pathology to mouse models of neurodegenerative disease. Pathological accumulations of misfolded tau, α-synuclein and mutant superoxide dismutase 1 in CNS tissues of transgenic mice were associated with changes in the solubility of hundreds of CNS proteins in each model. We observed that changes in proteome solubility were progressive and, using the rTg4510 model of inducible tau pathology, demonstrated that these changes were dependent upon sustained expression of the primary pathologic protein. In all of the models examined, changes in proteome solubility were robust, easily detected, and provided a sensitive indicator of proteostatic disruption. Interestingly, a subset of the proteins that display a shift towards insolubility were common between these different models, suggesting that a specific subset of the proteome is vulnerable to proteostatic disruption. Overall, our data suggest that neurodegenerative proteinopathies modeled in mice impose a burden on the proteostatic network that diminishes the ability of neural cells to prevent aberrant conformational changes that alter the solubility of hundreds of abundant cellular proteins.


Asunto(s)
Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Enfermedades Neurodegenerativas/patología , Ovillos Neurofibrilares/patología , Proteoma/metabolismo , Factores de Edad , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Enfermedades Neurodegenerativas/genética , Ovillos Neurofibrilares/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Pliegue de Proteína , Proteoma/genética , Solubilidad , Espectrometría de Masas en Tándem , alfa-Sinucleína/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
9.
Hum Mol Genet ; 24(4): 1019-35, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305079

RESUMEN

Co-expression of wild-type human superoxide dismutase 1 (WT-hSOD1) with ALS mutant hSOD1 accelerates disease onset relative to mice expressing only mutant protein. Here, we analyzed the effect of co-expressed WT-hSOD1 in two established mutant mouse models (L126Z and G37R), and a new model that expresses the first 102 amino acids of SOD1 with mutations at histidines 46, 48 and 63 to eliminate Cu binding (Cu-V103Z). A subset of Cu-V103Z mice developed paralysis between 500 and 730 days. Similar to mice expressing L126Z-SOD1, the spinal cords of this new model showed SOD1 immunoreactive fibrillar inclusions. Co-expression of WT-hSOD1 with Cu-V103Z SOD1 moderately accelerated the age to paralysis, similar in magnitude to WT/L126Z mice. In either combination of these bigenic mice, the severity of fibrillar inclusion pathology was diminished and unreactive to antibodies specific for the C terminus of WT protein. Co-expression of WT-hSOD1 fused to yellow fluorescent protein (WT-hSOD1:YFP) with G37R-hSOD1 produced earlier disease, and spinal cords of paralyzed bigenic mice showed YFP fluorescent inclusion-like structures. In bigenic L126Z/WT-hSOD1:YFP mice, disease was not accelerated and WT-hSOD1:YFP remained diffusely distributed. A combination of split luciferase complementation assays and affinity capture-binding assays demonstrated that soluble G37R-hSOD1 efficiently and tightly bound soluble WT-hSOD1, whereas soluble forms of the Cu-V103Z and L126Z variants demonstrated low affinity. These data indicate that WT-hSOD1 may indirectly augment the toxicity of mutant protein by competing for protective factors, but disease onset seems to be most accelerated when WT-hSOD1 interacts with mutant SOD1 and becomes misfolded.


Asunto(s)
Mutación , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/mortalidad , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Proteínas Mutantes/metabolismo , Unión Proteica , Multimerización de Proteína , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Superóxido Dismutasa/química , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
10.
J Immunol ; 195(1): 194-202, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26026056

RESUMEN

We previously showed that LIGHT and its receptor herpes virus entry mediator (HVEM) are important for development of optimal CD4(+) Th1 cell immunity and resistance to primary Leishmania major infection in mice. In this study, we further characterized the contributions of this molecule in dendritic cell (DC) maturation, initiation, and maintenance of primary immunity and secondary anti-Leishmania immunity. Flow-cytometric studies showed that CD8α(+) DC subset was mostly affected by HVEM-Ig and lymphotoxin ß receptor-Ig treatment. LIGHT signaling is required at both the priming and the maintenance stages of primary anti-Leishmania immunity but is completely dispensable during secondary immunity in wild type mice. However, LIGHT blockade led to impaired IL-12 and IFN-γ responses and loss of resistance in healed CD40-deficient mice after L. major challenge. The protective effect of LIGHT was mediated primarily via its interaction with lymphotoxin ß receptor on CD8α(+) DCs. Collectively, our results show that although LIGHT is critical for maintenance of primary Th1 response, it is dispensable during secondary anti-Leishmania immunity in the presence of functional CD40 signaling as seen in wild type mice.


Asunto(s)
Antígenos CD40/deficiencia , Interacciones Huésped-Patógeno/inmunología , Leishmaniasis Cutánea/inmunología , Células TH1/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/parasitología , Células de la Médula Ósea/patología , Antígenos CD40/genética , Antígenos CD40/inmunología , Antígenos CD8/genética , Antígenos CD8/inmunología , Técnicas de Cocultivo , Células Dendríticas/inmunología , Células Dendríticas/parasitología , Células Dendríticas/patología , Femenino , Regulación de la Expresión Génica , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Leishmania major/inmunología , Leishmaniasis Cutánea/genética , Leishmaniasis Cutánea/parasitología , Leishmaniasis Cutánea/patología , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/inmunología , Ratones , Transducción de Señal , Células TH1/parasitología , Células TH1/patología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
11.
J Cell Mol Med ; 20(10): 1861-71, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27241100

RESUMEN

LIGHT recruits and activates naive T cells in the islets at the onset of diabetes. IFN-γ secreted by activated T lymphocytes is involved in beta cell apoptosis. However, whether LIGHT sensitizes IFNγ-induced beta cells destruction remains unclear. In this study, we used the murine beta cell line MIN6 and primary islet cells as models for investigating the underlying cellular mechanisms involved in LIGHT/IFNγ - induced pancreatic beta cell destruction. LIGHT and IFN-γ synergistically reduced MIN6 and primary islet cells viability; decreased cell viability was due to apoptosis, as demonstrated by a significant increase in Annexin V(+) cell percentage, detected by flow cytometry. In addition to marked increases in cytochrome c release and NF-κB activation, the combination of LIGHT and IFN-γ caused an obvious decrease in expression of the anti-apoptotic proteins Bcl-2 and Bcl-xL, but an increase in expression of the pro-apoptotic proteins Bak and Bax in MIN6 cells. Accordingly, LIGHT deficiency led to a decrease in NF-κB activation and Bak expression, and peri-insulitis in non-obese diabetes mice. Inhibition of NF-κB activation with the specific NF-κB inhibitor, PDTC (pyrrolidine dithiocarbamate), reversed Bcl-xL down-regulation and Bax up-regulation, and led to a significant increase in LIGHT- and IFN-γ-treated cell viability. Moreover, cleaved caspase-9, -3, and PARP (poly (ADP-ribose) polymerase) were observed after LIGHT and IFN-γ treatment. Pretreatment with caspase inhibitors remarkably attenuated LIGHT- and IFNγ-induced cell apoptosis. Taken together, our results indicate that LIGHT signalling pathway combined with IFN-γ induces beta cells apoptosis via an NF-κB/Bcl2-dependent mitochondrial pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Interferón gamma/farmacología , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Células Secretoras de Insulina/metabolismo , Ratones Endogámicos NOD , Mitocondrias/efectos de los fármacos , Modelos Biológicos , Proteínas Recombinantes de Fusión/metabolismo , Estrés Fisiológico/efectos de los fármacos
12.
Microb Pathog ; 100: 250-256, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27725282

RESUMEN

LIGHT, a costimulatory member of the immunoglobulin superfamily (Ig SF), can greatly impact T cell activation. The role of the LIGHT signaling pathway in chlamydial infection was evaluated in mice following respiratory tract infection with Chlamydia psittaci. Compared with wild type (WT) mice, LIGHT knockout (KO) mice showed significant reduction of body weight, much lower survival rate, higher bacterial burden, prolonged infection time courses and more severe pathological changes in lung tissue. The mRNA levels of IFN-γ, TNF-α, IL-17 and IL-12 in the lung tissue of LIGHT KO mice were significantly lower than those in WT mice. While there was no obvious difference in the percentages of CD4+ and CD8+ T cells in the spleens of the two groups of mice, there was a markedly elevated percentage of CD4+ CD25+ FoxP3+ Treg cells in LIGHT KO mice. Together, these results demonstrate that the LIGHT signaling pathway is not only required for inflammatory cytokine production as part of the host response to chlamydial infection, but also influences the differentiation of CD4+ CD25+ FoxP3+ Treg cells, both of which may be essential for control of C. psittaci respiratory tract infection.


Asunto(s)
Chlamydophila psittaci/inmunología , Chlamydophila psittaci/patogenicidad , Psitacosis/patología , Transducción de Señal , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia , Animales , Carga Bacteriana , Peso Corporal , Citocinas/análisis , Citocinas/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Psitacosis/microbiología , ARN Mensajero/análisis , ARN Mensajero/genética , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Subgrupos de Linfocitos T/inmunología
13.
J Neurochem ; 133(3): 452-64, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25557022

RESUMEN

There has been great interest in enhancing endogenous protein maintenance pathways such as the heat-shock chaperone response, as it is postulated that enhancing clearance of misfolded proteins could have beneficial disease modifying effects in amyotrophic lateral sclerosis and other neurodegenerative disorders. In cultured cell models of mutant SOD1 aggregation, co-expression of αB-crystallin (αB-crys) has been shown to inhibit the formation of detergent-insoluble forms of mutant protein. Here, we describe the generation of a new line of transgenic mice that express αB-crys at > 6-fold the normal level in spinal cord, with robust increases in immunoreactivity throughout the spinal cord grey matter and, specifically, in spinal motor neurons. Surprisingly, spinal cords of mice expressing αB-crys alone contained 20% more motor neurons per section than littermate controls. Raising αB-crys by these levels in mice transgenic for either G93A or L126Z mutant SOD1 had no effect on the age at which paralysis developed. In the G93A mice, which showed the most robust degree of motor neuron loss, the number of these cells declined by the same proportion as in mice expressing the mutant SOD1 alone. In paralyzed bigenic mice, the levels of detergent-insoluble, misfolded, mutant SOD1 were similar to those of mice expressing mutant SOD1 alone. These findings indicate that raising the levels of αB-crys in spinal motor neurons by 6-fold does not produce the therapeutic effects predicted by cell culture models of mutant SOD1 aggregation. Enhancing the protein chaperone function may present a therapeutic approach to amyotrophic lateral sclerosis caused by mutations in SOD1, and other neurodegenerative disorders characterized by cytosolic protein aggregation. Previous studies in cell models suggested that the chaperone known as αB-crystallin (αB-crys) can prevent mutant SOD1 aggregation. We report that transgenic expression of αB-crys at > 6-fold the normal level in spinal cords of mice expressing mutant SOD1 produces no therapeutic benefit.


Asunto(s)
Neuronas Motoras/metabolismo , Proteínas Mutantes/biosíntesis , Parálisis/metabolismo , Agregación Patológica de Proteínas/metabolismo , Superóxido Dismutasa , Cadena B de alfa-Cristalina/biosíntesis , Animales , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Mutantes/genética , Parálisis/genética , Parálisis/prevención & control , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/prevención & control , Médula Espinal/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Cadena B de alfa-Cristalina/genética
14.
Hum Mol Genet ; 22(14): 2765-74, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23512986

RESUMEN

The extracellular accumulation of ß-amyloid peptide is a key trigger in the pathogenesis of Alzheimer's disease (AD). In humans, amyloid deposition precedes the appearance of intracellular inclusion pathology formed by cytosolic proteins such as Tau, α-synuclein and TDP-43. These secondary pathologies have not been observed in mice that model Alzheimer-type amyloidosis by expressing mutant amyloid precursor protein, with or without mutant presenilin 1. The lack of secondary pathology in these models has made it difficult to establish how amyloid deposition initiates the cascade of events that leads to secondary intracellular pathology that characterizes human AD. In transgenic mice that model Alzheimer-type amyloidosis, we sought to determine whether there is evidence of altered cytosolic protein folding by assessing whether amyloid deposition causes normally soluble proteins to misfold. Using a method that involved detergent extraction and sedimentation coupled with proteomic approaches, we identified numerous cytosolic proteins that show specific losses in solubility as amyloid accumulates. The proteins identified included glycolytic enzymes and members of the 14-3-3 chaperone family. A substantial accumulation of lysine 48-linked polyubiquitin was also detected. Overall, the data demonstrate that the accumulation of amyloid by some manner causes the loss of solubility intracellular cytosolic proteins.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Amiloidosis/metabolismo , Citosol/metabolismo , Enfermedad de Alzheimer/genética , Amiloide/química , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Amiloidosis/genética , Animales , Citosol/química , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Pliegue de Proteína
15.
Hepatology ; 60(1): 114-24, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24604562

RESUMEN

UNLABELLED: Viral fulminant hepatitis (FH) remains a serious clinical problem with very high mortality. Lacking understanding of FH pathogenesis has in essence hindered efficient clinical treatment. Inferring from a correlation observed between the genetic differences in the complement component 5 (C5) and the susceptibility of mouse strains to murine hepatitis virus strain-3 (MHV-3) infections, we propose that excessive complement activation plays a critical role in the development of FH. We show that MHV-3 infection causes massive complement activation, along with a rapid increase in serum C5a levels and quick development of FH in susceptible strains. Mice deficient in the C5a receptor (C5aR) or the susceptible strains treated with C5aR antagonists (C5aRa) exhibit significant attenuation of the disease, accompanied by a remarkable reduction of hepatic fibrinogen-like protein 2 (Fgl2), a hallmark protein that causes necrosis of infected livers. In accordance, biopsy of FH patients shows a dramatic increase of Fgl2 expression, which correlates with C5aR up-regulation in the liver. In vitro C5a administration accelerates MHV-3-induced Fgl2 secretion by macrophages. Furthermore, inhibiting ERK1/2 and p38 efficiently blocks C5a-mediated Fgl2 production during viral infections. CONCLUSION: These data provide evidence that mouse susceptibility to MHV-3-induced FH may rely on C5a/C5aR interactions, for which ERK1/2 and p38 pathways participate in up-regulating Fgl2 expression. Inhibition of C5a/C5aR interactions is expected to be beneficial in the clinical treatment of FH patients.


Asunto(s)
Complemento C5a/metabolismo , Fibrinógeno/metabolismo , Hepatitis Viral Animal/metabolismo , Virus de la Hepatitis Murina/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Enfermedad Aguda , Animales , Complemento C5a/inmunología , Femenino , Fibrinógeno/inmunología , Hepatitis Viral Animal/inmunología , Humanos , Fallo Hepático Agudo/inmunología , Fallo Hepático Agudo/metabolismo , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Hepatitis Murina/inmunología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Índice de Severidad de la Enfermedad , Regulación hacia Arriba/inmunología
16.
J Immunol ; 191(1): 178-86, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23709683

RESUMEN

The protective immunity induced by the whole-killed parasite vaccine against malarial blood-stage infection is dependent on the CD4(+) T cell response. However, the mechanism underlying this robust CD4(+) T cell response elicited by the whole-killed parasite vaccine is still largely unknown. In this study, we observe that immunization with Plasmodium yoelii-parasitized RBC lysate activates complement C5 and generates C5a. However, the protective efficacy against P. yoelii 17XL challenge is considerably reduced, and the malaria-specific CD4(+) T cell activation and memory T cell differentiation are largely suppressed in the C5aR-deficient (C5aR(-/-)) mice. An adoptive transfer assay demonstrates that the reduced protection of C5aR(-/-) mice is closely associated with the severely impaired CD4(+) T cell response. This is further confirmed by the fact that administration of C5aR antagonist significantly reduces the protective efficacy of the immunized B cell-deficient mice. Further study indicates that the defective CD4(+) T cell response in C5aR(-/-) mice is unlikely involved in the expansion of CD4(+)CD25(+)Foxp3(+) T cells, but strongly linked to a defect in dendritic cell (DC) maturation and the ability to allostimulate CD4(+) T cells. These results demonstrate that C5aR signaling is essential for the optimal induction of the malaria-specific CD4(+) T cell response by the whole-killed parasite vaccine through modulation of DCs function, which provides us with new clues to design an effective blood-stage subunit vaccine and helps us to understand the mechanism by which the T cell response is regulated by the complement system.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Malaria/patología , Plasmodium yoelii/inmunología , Receptor de Anafilatoxina C5a/fisiología , Transducción de Señal/inmunología , Animales , Linfocitos T CD4-Positivos/parasitología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Memoria Inmunológica/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Malaria/parasitología , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/sangre , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/patogenicidad , Receptor de Anafilatoxina C5a/sangre , Receptor de Anafilatoxina C5a/deficiencia , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/sangre
17.
Wei Sheng Wu Xue Bao ; 55(4): 492-500, 2015 Apr 04.
Artículo en Zh | MEDLINE | ID: mdl-26211324

RESUMEN

OBJECTIVE: To study the role of lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells (LIGHT) in the development of protective immunity and pathology during Chlamydia Muridarum urogenital infection in mice. METHODS: C57BL/6J wild type (wt) and mice deficient in LIGHT (LIGHT KO) were inoculated intravaginally with 1 x 10(4) IFUs of live C. muridarum organisms. Half mice of each group were reinfected on day 49 after primary infection. We took mice vaginal swabs every 3 or 4 days to monitor live organism shedding. On day 80 after the primary infection, mice were sacrificed, the vaginal tract was isolated for pathology analysis. The spleen cells were collected and IL-4, IL-5, IL-17 and IFN-y were detected by ELISA in the spleen cells culture supernatant after restimulated by UV-MoPn EB. The titers of different Ab isotypes were measured in mice serum by Indirect Immunofluorescence Assay. RESULTS: The chlamydia shedding time of LIGHT KO mice was similar to wild type mice, which cleared the organisms within 28 days after primary infection, and acquired protective immunity against C. muridarum reinfection. All mice regardless of genotypes developed severe upper genital tract pathology and showed no significant difference between LIGHT KO and wild type mice. All mice developed robust anti-C. muridarum organism IgG antibody responses and the ratios of IgG2a versus IgG1 showed no significant difference between LIGHT KO and wild type mice. Splenocytes from MoPn-infected LIGHT KO and wild type mice produced high levels of IFN-gamma and IL-17, but IL-4 and IL-5 couldn't be detected. CONCLUSIONS: LIGHT signal pathway may not correlated with protection against C. muridarum urogenital tract infection and urogenital tract pathology induced by C. muridarum.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia muridarum/fisiología , Transducción de Señal , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Vagina/inmunología , Animales , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/microbiología , Femenino , Humanos , Interferón gamma/inmunología , Interleucina-4/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Vagina/microbiología
18.
J Neurosci ; 33(9): 3765-79, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23447589

RESUMEN

Transgenic mice that express mutant amyloid precursor protein (APPsi) using tet-Off vector systems provide an alternative model for assessing short- and long-term effects of Aß-targeting therapies on phenotypes related to the deposition of Alzheimer-type amyloid. Here we use such a model, termed APPsi:tTA, to determine what phenotypes persist in mice with high amyloid burden after new production of APP/Aß has been suppressed. We find that 12- to 13-month-old APPsi:tTA mice are impaired in cognitive tasks that assess short- and long-term memories. Acutely suppressing new APPsi/Aß production produced highly significant improvements in performing short-term spatial memory tasks, which upon continued suppression translated to superior performance in more demanding tasks that assess long-term spatial memory and working memory. Deficits in episodic-like memory and cognitive flexibility, however, were more persistent. Arresting mutant APPsi production caused a rapid decline in the brain levels of soluble APP ectodomains, full-length APP, and APP C-terminal fragments. As expected, amyloid deposits persisted after new APP/Aß production was inhibited, whereas, unexpectedly, we detected persistent pools of solubilizable, relatively mobile, Aß42. Additionally, we observed persistent levels of Aß-immunoreactive entities that were of a size consistent with SDS-resistant oligomeric assemblies. Thus, in this model with significant amyloid pathology, a rapid amelioration of cognitive deficits was observed despite persistent levels of oligomeric Aß assemblies and low, but detectable solubilizable Aß42 peptides. These findings implicate complex relationships between accumulating Aß and activities of APP, soluble APP ectodomains, and/or APP C-terminal fragments in mediating cognitive deficits in this model of amyloidosis.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Amiloidosis/complicaciones , Amiloidosis/patología , Encéfalo/metabolismo , Trastornos del Conocimiento/etiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/dietoterapia , Amiloidosis/genética , Análisis de Varianza , Animales , Encéfalo/patología , Encéfalo/ultraestructura , Trastornos del Conocimiento/dietoterapia , Trastornos del Conocimiento/patología , Discriminación en Psicología/fisiología , Modelos Animales de Enfermedad , Doxiciclina/administración & dosificación , Ensayo de Inmunoadsorción Enzimática , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Fragmentos de Péptidos/metabolismo , Fenotipo , Placa Amiloide/dietoterapia , Placa Amiloide/metabolismo , Placa Amiloide/patología , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Reconocimiento en Psicología/efectos de los fármacos , Percepción Espacial , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/patología , Factores de Tiempo
19.
Acta Neuropathol ; 128(6): 791-803, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25262000

RESUMEN

By unknown mechanisms, the symptoms of amyotrophic lateral sclerosis (ALS) seem to spread along neuroanatomical pathways to engulf the motor nervous system. The rate at which symptoms spread is one of the primary drivers of disease progression. One mechanism by which ALS symptoms could spread is by a prion-like propagation of a toxic misfolded protein from cell to cell along neuroanatomic pathways. Proteins that can transmit toxic conformations between cells often can also experimentally transmit disease between individual organisms. To survey the ease with which motor neuron disease (MND) can be transmitted, we injected spinal cord homogenates prepared from paralyzed mice expressing mutant superoxide dismutase 1 (SOD1-G93A and G37R) into the spinal cords of genetically vulnerable SOD1 transgenic mice. From the various models we tested, one emerged as showing high vulnerability. Tissue homogenates from paralyzed G93A mice induced MND in 6 of 10 mice expressing low levels of G85R-SOD1 fused to yellow fluorescent protein (G85R-YFP mice) by 3-11 months, and produced widespread spinal inclusion pathology. Importantly, second passage of homogenates from G93A â†’ G85R-YFP mice back into newborn G85R-YFP mice induced disease in 4 of 4 mice by 3 months of age. Homogenates from paralyzed mice expressing the G37R variant were among those that transmitted poorly regardless of the strain of recipient transgenic animal injected, a finding suggestive of strain-like properties that manifest as differing abilities to transmit MND. Together, our data provide a working model for MND transmission to study the pathogenesis of ALS.


Asunto(s)
Enfermedad de la Neurona Motora/fisiopatología , Médula Espinal/fisiopatología , Superóxido Dismutasa/metabolismo , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Animales Recién Nacidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Estimación de Kaplan-Meier , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Enfermedad de la Neurona Motora/patología , Mutación , Parálisis/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
20.
Hum Mol Genet ; 20(14): 2770-82, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21515588

RESUMEN

Recent studies have implicated an N-terminal caspase-6 cleavage product of mutant huntingtin (htt) as an important mediator of toxicity in Huntington's disease (HD). To directly assess the consequences of such fragments on neurologic function, we produced transgenic mice that express a caspase-6 length N-terminal fragment of mutant htt (N586) with both normal (23Q) and disease (82Q) length glutamine repeats. In contrast to mice expressing N586-23Q, mice expressing N586-82Q accumulate large cytoplasmic inclusion bodies that can be visualized with antibodies to epitopes throughout the N586 protein. However, biochemical analyses of aggregated mutant huntingtin in these mice demonstrated that the inclusion bodies are composed largely of a much smaller htt fragment (terminating before residue 115), with lesser amounts of full-length N586-82Q fragments. Mice expressing the N586-82Q fragment show symptoms typical of previously generated mice expressing mutant huntingtin fragments, including failure to maintain weight, small brain weight and reductions in specific mRNAs in the striatum. Uniquely, these N586-82Q mice develop a progressive movement disorder that includes dramatic deficits in motor performance on the rotarod and ataxia. Our findings suggest that caspase-6-derived fragments of mutant htt are capable of inducing novel HD-related phenotypes, but these fragments are not terminal cleavage products as they are subject to further proteolysis. In this scenario, mutant htt fragments derived from caspase 6, or possibly other proteases, could mediate HD pathogenesis via a 'hit and run' type of mechanism in which caspase-6, or other larger N-terminal fragments, mediate a neurotoxic process before being cleaved to a smaller fragment that accumulates pathologically.


Asunto(s)
Cuerpo Estriado/metabolismo , Expresión Génica , Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión/metabolismo , Mutación Missense , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Sustitución de Aminoácidos , Animales , Ataxia/genética , Ataxia/metabolismo , Ataxia/patología , Caspasa 6 , Cuerpo Estriado/patología , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Cuerpos de Inclusión/patología , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Prueba de Desempeño de Rotación con Aceleración Constante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA