Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Clin Invest ; 131(14)2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34263740

RESUMEN

Patients with congenital lymphedema suffer from tissue swelling in part due to mutations in genes regulating lymphatic valve development. Lymphatic valve leaflets grow and are maintained throughout life in response to oscillatory shear stress (OSS), which regulates gene transcription in lymphatic endothelial cells (LECs). Here, we identified the first transcription factor, Foxo1, that repressed lymphatic valve formation by inhibiting the expression of valve-forming genes. We showed that both embryonic and postnatal ablation of Foxo1 in LECs induced additional valve formation in postnatal and adult mice in multiple tissues. Our quantitative analyses revealed that after deletion, the total number of valves in the mesentery was significantly (P < 0.01) increased in the Foxo1LEC-KO mice compared with Foxo1fl/fl controls. In addition, our quantitative real-time PCR (RT-PCR) data from cultured LECs showed that many valve-forming genes were significantly (P < 0.01) upregulated upon knockdown of FOXO1. To confirm our findings in vivo, rescue experiments showed that Foxc2+/- mice, a model of lymphedema-distichiasis, had 50% fewer lymphatic valves and that the remaining valves exhibited backleak. Both valve number and function were completely restored to control levels upon Foxo1 deletion. These findings established FOXO1 as a clinically relevant target to stimulate de novo lymphatic valve formation and rescue defective valves in congenital lymphedema.


Asunto(s)
Proteína Forkhead Box O1/deficiencia , Eliminación de Gen , Linfangiogénesis , Vasos Linfáticos/metabolismo , Animales , Proteína Forkhead Box O1/metabolismo , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Noqueados
3.
Am J Transl Res ; 11(8): 5076-5085, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31497223

RESUMEN

Apolipoprotein E (apoE) and apoE-mimetic peptides exert prominent anti-inflammatory effects. We determined the anti-inflammatory effects of novel apoE receptor mimetics, composed of the LDL receptor-binding domain of apoE (aa 133-152, ApoEp) or ApoEp with 6 lysines (6KApoEp) or 6 aspartates added at the N-terminus (6DApoEp). BV2 microglia and human THP-1 monocytes were treated with lipopolysaccharide (LPS) in the absence or presence of ApoEp, 6KApoEp or 6DApoEp, followed by determination of pro-inflammatory tumor necrosis factor α (TNFα) and interleukin-6 (IL-6) release by ELISA. As signaling intermediates of inflammation, Signal Transducer and Activator of Transcription 3 (STAT3), Janus-Activated Kinase2 (JAK2) and p38 and p44/42 MAPK phosphorylation levels were determined by Western blot analysis. In addition, we isolated splenocytes from female htau mice treated with 6KApoEp or 6K for 28 weeks, followed by determination of concanavalinA (conA)-mediated interferon gamma (IFNγ) release. 6KApoEp starting at 2.5 µM significantly reduced LPS-mediated TNFα and IL-6 secretion in BV2 and THP-1 cells in a dose-dependent manner. In BV2 cells, 6KApoEp reduced TNFα secretion more effectively than 6DApoEp and ApoEp, which was blocked by PCSK9 treatment, suggesting a role for LDL receptors. 6KApoEp also inhibited LPS-induced p44/42 MAPK, JAK2 and STAT3 phosphorylation, while enhancing p38 MAPK phosphorylation. In addition, conA induced significantly less IFNγ release in splenocytes derived from htau mice treated with 6KApoEp compared with those treated with 6K. Thus, 6KApoEp most effectively reduces LPS-mediated neuroinflammation by interacting with LDL receptors, thus representing a novel anti-inflammatory agent for treatment of neurodegenerative disease.

4.
Biol Psychiatry ; 86(3): 208-220, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31208706

RESUMEN

BACKGROUND: The ɛ4 isoform of apolipoprotein E (apoE4) is a major genetic risk factor for the development of sporadic Alzheimer's disease (AD), and its modification has been an intense focus for treatment of AD during recent years. METHODS: We investigated the binding of apoE, a peptide corresponding to its low-density lipoprotein receptor binding domain (amino acids 133-152; ApoEp), and modified ApoEp to amyloid precursor protein (APP) and their effects on amyloid-ß (Aß) production in cultured cells. Having discovered a peptide (6KApoEp) that blocks the interaction of apoE with N-terminal APP, we investigated the effects of this peptide and ApoEp on AD-like pathology and behavioral impairment in 3XTg-AD and 5XFAD transgenic mice. RESULTS: ApoE and ApoEp, but not truncated apoE lacking the low-density lipoprotein receptor binding domain, physically interacted with N-terminal APP and thereby mediated Aß production. Interestingly, the addition of 6 lysine residues to the N-terminus of ApoEp (6KApoEp) directly inhibited apoE binding to N-terminal APP and markedly limited apoE- and ApoEp-mediated Aß generation, presumably through decreasing APP cellular membrane trafficking and p44/42 mitogen-activated protein kinase phosphorylation. Moreover, while promoting apoE interaction with APP by ApoEp exacerbated Aß and tau brain pathologies in 3XTg-AD mice, disrupting this interaction by 6KApoEp ameliorated cerebral Aß and tau pathologies, neuronal apoptosis, synaptic loss, and hippocampal-dependent learning and memory impairment in 5XFAD mice without altering cholesterol, low-density lipoprotein receptor, and apoE expression levels. CONCLUSIONS: These data suggest that disrupting apoE interaction with N-terminal APP may be a novel disease-modifying therapeutic strategy for AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteínas E/antagonistas & inhibidores , Apolipoproteínas E/metabolismo , Encéfalo/patología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Apolipoproteínas E/genética , Células CHO , Cognición/efectos de los fármacos , Cricetulus , Modelos Animales de Enfermedad , Femenino , Humanos , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Transgénicos
5.
J Neurosci Res ; 97(9): 1066-1080, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31102295

RESUMEN

Alzheimer's disease (AD) is characterized by progressive decline of cognition and associated neuropsychiatric signs including weight loss, anxiety, depression, agitation, and aggression, which is particularly pronounced in the female gender. Previously, we have shown that a novel ionic co-crystal of lithium salicylate proline (LISPRO) is an improved lithium formulation compared to the carbonate or salicylate form of lithium in terms of safety and efficacy in reducing AD pathology in Alzheimer's mice. The current study is designed to compare the prophylactic effects of LISPRO, lithium carbonate (LC), and lithium salicylate (LS) on cognitive and noncognitive impairments in female transgenic APPswe/PS1dE9 AD mice. Female APPswe/PS1dE9 mice at 4 months of age were orally treated with low-dose LISPRO, LS, or LC for 9 months at 2.25 mmol lithium/kg/day followed by determination of body weight, growth of internal organs, and cognitive and noncognitive behavior. No significant differences in body or internal organ weight, anxiety or locomotor activity were found between lithium treated and untreated APPswe/PS1dE9 cohorts. LISPRO, LC, and LS prevented spatial cognitive decline, as determined by Morris water maze and depression as determined by tail suspension test. In addition, LISPRO treatment was superior in preventing associative memory decline determined by contextual fear conditioning and reducing irritability determined by touch escape test in comparison with LC and LS. In conclusion, low-dose LISPRO, LC, and LS treatment prevent spatial cognitive decline and depression-like behavior, while LISPRO prevented hippocampal-dependent associative memory decline and irritability in APPswe/PS1dE9 mice.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Conducta/efectos de los fármacos , Carbonato de Litio/farmacología , Compuestos de Litio/farmacología , Memoria/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Suspensión Trasera , Locomoción/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Prolina , Salicilatos , Memoria Espacial/efectos de los fármacos
6.
Cell Transplant ; 27(4): 666-676, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29871524

RESUMEN

Alzheimer's Disease (AD) is the leading cause of dementia in the elderly. In healthy individuals, amyloid precursor protein (APP) is cleaved by α-secretase, generating soluble α-amyloid precursor protein (sAPPα), which contributes neuroprotective functions in the neuronal environment. In contrast, in the neurodegenerative environment of AD patients, amyloid-ß-peptide (Aß) of either 40 or 42 residues are generated by increased activity of ß- and γ-secretase. These proteins amalgamate in specific regions of the brain, which disrupts neuronal functions and leads to cognitive impairment. Human umbilical cord blood cells (HUCBC) have proven useful as potential immunomodulatory therapies in various models of neurodegenerative diseases, including AD. Our most recent work studied the impact of umbilical cord blood serum (CBS) on modulation of sAPPα production. Heat-sensitive CBS significantly promoted sAPPα production, indicating that heat-sensitive factor(s) play(s) a role in this process. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis was used to determine the molecular source of α-secretase in purified CBS and aged blood serum (AgBS) fraction. Of the proteins identified, the subunits of C1 complex (C1q, C1r, and C1s) and alpha-2-macroglobulin showed significantly greater levels in purified α-CBS fraction (α-CBSF) compared with the AgBS fraction (AgBSF). Specifically, C1 markedly increased sAPPα and alpha-carboxyl-terminal fragment (α-CTF) production in a dose-dependent fashion, whereas C1q alone only minimally increased and C3 did not increase sAPPα production in the absence of sera. Furthermore, C1q markedly increased sAPPα and α-CTF, while decreasing Aß, in CHO/APPwt cells cultured in the presence of whole sera. These results confirm our initial assumption that APP α-secretase activity in human blood serum is mediated by complement C1, opening a potential therapeutic modality for the future of AD.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Complemento C1/metabolismo , Sangre Fetal/enzimología , Anciano , Envejecimiento/sangre , Péptidos beta-Amiloides/metabolismo , Animales , Células CHO , Complemento C1/aislamiento & purificación , Complemento C3b/metabolismo , Cricetinae , Cricetulus , Calor , Humanos , Ratones , Proteómica
7.
Cell Transplant ; 27(3): 438-455, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29560732

RESUMEN

Alzheimer's disease (AD) is an age-related disorder that affects cognition. Our previous studies showed that the neuroprotective fragment of amyloid procurer protein (APP) metabolite, soluble APPα (sAPPα), interferes with ß-site APP-cleaving enzyme 1 (BACE1, ß-secretase) cleavage and reduces amyloid-ß (Aß) generation. In an attempt to identify approaches to restore sAPPα levels, we found that human cord blood serum (CBS) significantly promotes sAPPα production compared with adult blood serum (ABS) and aged blood serum (AgBS) in Chinese hamster ovary cells stably expressing wild-type human APP. Interestingly, CBS selectively mediated the α-secretase cleavage of human neuron-specific recombinant APP695 in a cell-free system independent of tumor necrosis factor-α converting enzyme (TACE; a disintegrin and metalloproteinase domain-containing protein 17 [ADAM17]) and ADAM. Subsequently, using 3-step chromatographic separation techniques (i.e., diethylaminoethanol, size-exclusion, and ion-exchange chromatography), we purified and ultimately identified a CBS-specific fraction with enhanced α-secretase catalytic activity (termed αCBSF) and found that αCBSF has more than 3,000-fold increased α-secretase catalytic activity compared with the original pooled CBS. Furthermore, intracerebroventricular injection of αCBSF markedly increased cerebral sAPPα levels together with significant decreases in cerebral Aß production and abnormal tau (Thr231) phosphorylation compared with the AgBS fraction with enhanced α-secretase activity (AgBSF) treatment in triple transgenic Alzheimer's disease (3xTg-AD) mice. Moreover, AgBSF administered intraperitoneally to transgenic mice with five familial Alzheimer's disease mutations (5XFAD) via an osmotic mini pump for 6 weeks (wk) ameliorated ß-amyloid plaques and reversed cognitive impairment measures. Together, our results propose the necessity for further study aimed at identification and characterization of α-secretase in CBS for novel and effective AD therapy.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Células CHO , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Ratones , Fosforilación , Proteínas tau/metabolismo
8.
Cell Death Dis ; 8(6): e2880, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28617434

RESUMEN

Lithium has been marketed in the United States of America since the 1970s as a treatment for bipolar disorder. More recently, studies have shown that lithium can improve cognitive decline associated with Alzheimer's disease (AD). However, the current United States Food and Drug Administration-approved lithium pharmaceutics (carbonate and citrate chemical forms) have a narrow therapeutic window and unstable pharmacokinetics that, without careful monitoring, can cause serious adverse effects. Here, we investigated the safety profile, pharmacokinetics, and therapeutic efficacy of LISPRO (ionic co-crystal of lithium salicylate and l-proline), lithium salicylate, and lithium carbonate (Li2CO3). We found that LISPRO (8-week oral treatment) reduces ß-amyloid plaques and phosphorylation of tau by reducing neuroinflammation and inactivating glycogen synthase kinase 3ß in transgenic Tg2576 mice. Specifically, cytokine profiles from the brain, plasma, and splenocytes suggested that 8-week oral treatment with LISPRO downregulates pro-inflammatory cytokines, upregulates anti-inflammatory cytokines, and suppresses renal cyclooxygenase 2 expression in transgenic Tg2576 mice. Pharmacokinetic studies indicated that LISPRO provides significantly higher brain lithium levels and more steady plasma lithium levels in both B6129SF2/J (2-week oral treatment) and transgenic Tg2576 (8-week oral treatment) mice compared with Li2CO3. Oral administration of LISPRO for 28 weeks significantly reduced ß-amyloid plaques and tau-phosphorylation. In addition, LISPRO significantly elevated pre-synaptic (synaptophysin) and post-synaptic protein (post synaptic density protein 95) expression in brains from transgenic 3XTg-AD mice. Taken together, our data suggest that LISPRO may be a superior form of lithium with improved safety and efficacy as a potential new disease modifying drug for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Compuestos de Litio/administración & dosificación , Prolina/administración & dosificación , Administración Oral , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Autofagia , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células HeLa , Humanos , Inflamación , Carbonato de Litio/administración & dosificación , Carbonato de Litio/sangre , Compuestos de Litio/sangre , Compuestos de Litio/química , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Fagocitosis , Fosforilación , Prolina/sangre , Prolina/química , Resultado del Tratamiento
9.
Neuromolecular Med ; 19(2-3): 300-308, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28612181

RESUMEN

Previous studies have demonstrated that the low-density lipoprotein receptor-related protein-1 (LRP1) plays conflicting roles in Alzheimer's disease (AD) pathogenesis, clearing ß-amyloid (Aß) from the brain while also enhancing APP endocytosis and resultant amyloidogenic processing. We have recently discovered that co-expression of mutant LRP1 C-terminal domain (LRP1-CT C4408R) with Swedish mutant amyloid precursor protein (APPswe) in Chinese hamster ovary (CHO) cells decreases Aß production, while also increasing sAPPα and APP α-C-terminal fragment (α-CTF), compared with CHO cells expressing APPswe alone. Surprisingly, the location of this mutation on LRP1 corresponded with the α-secretase cleavage site of APP. Further experimentation confirmed that in CHO cells expressing APPswe or wild-type APP (APPwt), co-expression of LRP1-CT C4408R decreases Aß and increases sAPPα and α-CTF compared with co-expression of wild-type LRP1-CT. In addition, LRP1-CT C4408R enhanced the unglycosylated form of LRP1-CT and reduced APP endocytosis as determined by flow cytometry. This finding identifies a point mutation in LRP1 which slows LRP1-CT-mediated APP endocytosis and amyloidogenic processing, while enhancing APP α-secretase cleavage, thus demonstrating a potential novel target for slowing AD pathogenesis.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Mutación Missense , Mutación Puntual , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Endocitosis , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Dominios Proteicos , Proteínas Recombinantes/metabolismo
10.
J Neuroimmunol ; 299: 98-106, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27725131

RESUMEN

Naturally-occurring bioactive flavonoids such as diosmin significantly reduces amyloid beta (Aß) associated pathology in Alzheimer's disease (AD) mouse models. In the present study, oral administration of diosmin reduced cerebral Aß oligomer levels, tau-hyperphosphorylation and cognitive impairment in the 3xTg-AD mouse model through glycogen synthase kinase-3 (GSK-3) and transient receptor potential canonical 6-related mechanisms. Diosmetin, one major bioactive metabolite of diosmin, increased inhibitory GSK-3ß phosphorylation, while selectively reducing γ-secretase activity, Aß generation, tau hyperphosphorylation and pro-inflammatory activation of microglia in vitro, without altering Notch processing. Therefore, both diosmin and diosmetin could be considered as potential candidates for novel anti-AD therapy.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Corteza Cerebral/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Diosmina/uso terapéutico , Proteínas tau/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Células CHO , Corteza Cerebral/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Cricetinae , Cricetulus , Diosmina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Células HeLa , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas tau/antagonistas & inhibidores
11.
J Neurochem ; 135(3): 630-7, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342176

RESUMEN

We recently found that sAPPα decreases amyloid-beta generation by directly associating with ß-site amyloid precursor protein (APP)-converting enzyme 1 (BACE1), thereby modulating APP processing. Because inhibition of BACE1 decreases glycogen synthase kinase 3 beta (GSK3ß)-mediated Alzheimer's disease (AD)-like tau phosphorylation in AD patient-derived neurons, we determined whether sAPPα also reduces GSK3ß-mediated tau phosphorylation. We initially found increased levels of inhibitory phosphorylation of GSK3ß (Ser9) in primary neurons from sAPPα over-expressing mice. Further, recombinant human sAPPα evoked the same phenomenon in SH-SY5Y cells. Further, in SH-SY5Y cells over-expressing BACE1, and HeLa cells over-expressing human tau, sAPPα reduced GSK3ß activity and tau phosphorylation. Importantly, the reductions in GSK3ß activity and tau phosphorylation elicited by sAPPα were prevented by BACE1 but not γ-secretase inhibition. In accord, AD mice over-expressing human sAPPα had less GSK3ß activity and tau phosphorylation compared with controls. These results implicate a direct relationship between APP ß-processing and GSK3ß-mediated tau phosphorylation and further define the central role of sAPPα in APP autoregulation and AD pathogenesis.


Asunto(s)
Precursor de Proteína beta-Amiloide/farmacología , Glucógeno Sintasa Quinasa 3/fisiología , Transducción de Señal/fisiología , Proteínas tau/antagonistas & inhibidores , Proteínas tau/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Relación Dosis-Respuesta a Droga , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transducción de Señal/efectos de los fármacos
12.
Cell Transplant ; 24(11): 2237-50, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26230612

RESUMEN

Alzheimer's disease (AD) is the fourth major cause of mortality in the elderly in the US and the leading cause of dementia worldwide. While pharmacological targets have been discovered, there are no true disease-modifying therapies. We have recently discovered that multiple low-dose infusions of human umbilical cord blood cells (HUCBCs) ameliorate cognitive impairments and reduce Aß-associated neuropathology in PSAPP transgenic mice. However, the mechanism for these effects of HUCBCs remains unclear. In the present study, we examined whether monocytes, as important components of HUCBCs, would have beneficial outcomes on the reduction of AD-like pathology and associated cognitive impairments in PSAPP transgenic AD model mice. PSAPP mice and their wild-type littermates were treated monthly with an infusion of peripheral human umbilical cord blood cell (HUCBC)-derived monocytes over a period of 2 and 4 months, followed by behavioral evaluations, biochemical, and histological analyses. The principal findings of the present study confirmed that monocytes derived from HUCBCs (CB-M) play a central role in HUCBC-mediated cognition-enhancing and Aß pathology-ameliorating activities. Most importantly, we found that compared with CB-M, aged monocytes showed an ineffective phagocytosis of Aß, while exogenous soluble amyloid precursor protein α (sAPPα) could reverse this deficiency. Pretreating monocytes with sAPPα upregulates Aß internalization. Our further studies suggested that sAPPα could form a heterodimer with Aßs, with the APP672-688 (Aß1-16) region being responsible for this effect. This in turn promoted binding of these heterodimers to monocyte scavenger receptors and thus promoted enhanced Aß clearance. In summary, our findings suggest an interesting hypothesis that peripheral monocytes contribute to Aß clearance through heterodimerization of sAPPα with Aß. Further, declined or impaired sAPPα production, or reduced heterodimerization with Aß, would cause a deficiency in Aß clearance and thus accelerate the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Monocitos/trasplante , Cordón Umbilical/citología , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Humanos , Ratones , Monocitos/metabolismo , Dominios y Motivos de Interacción de Proteínas
13.
Sci Rep ; 5: 11322, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-26091071

RESUMEN

BACE1 initiates amyloid-ß (Aß) generation and the resultant cerebral amyloidosis, as a characteristic of Alzheimer's disease (AD). Thus, inhibition of BACE1 has been the focus of a large body of research. The most recent clinical trials highlight the difficulty involved in this type of anti-AD therapy as evidenced by side effects likely due to the ubiquitous nature of BACE1, which cleaves multiple substrates. The human Swedish mutant form of amyloid protein precursor (APPswe) has been shown to possess a higher affinity for BACE1 compared to wild-type APP (APPwt). We pursued a new approach wherein harnessing this greater affinity to modulate BACE1 APP processing activity. We found that one peptide derived from APPswe, containing the ß-cleavage site, strongly inhibits BACE1 activity and thereby reduces Aß production. This peptide, termed APPswe BACE1 binding site peptide (APPsweBBP), was further conjugated to the fusion domain of the HIV-1 Tat protein (TAT) at the C-terminus to facilitate its biomembrane-penetrating activity. APPwt and APPswe over-expressing CHO cells treated with this TAT-conjugated peptide resulted in a marked reduction of Aß and a significant increase of soluble APPα. Intraperitoneal administration of this peptide to 5XFAD mice markedly reduced ß-amyloid deposits as well as improved hippocampal-dependent learning and memory.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Sitios de Unión , Mutación , Péptidos/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/química , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Conducta Animal , Sitios de Unión/genética , Barrera Hematoencefálica/metabolismo , Células CHO , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cricetulus , Modelos Animales de Enfermedad , Expresión Génica , Hipocampo/metabolismo , Memoria , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Unión Proteica , Conformación Proteica , Proteolisis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
14.
J Biol Chem ; 289(44): 30303-30317, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25157105

RESUMEN

Amyloid precursor protein (APP) proteolysis is required for production of amyloid-ß (Aß) peptides that comprise ß-amyloid plaques in the brains of patients with Alzheimer disease (AD). Here, we tested whether the experimental agent methylene blue (MB), used for treatment of methemoglobinemia, might improve AD-like pathology and behavioral deficits. We orally administered MB to the aged transgenic PSAPP mouse model of cerebral amyloidosis and evaluated cognitive function and cerebral amyloid pathology. Beginning at 15 months of age, animals were gavaged with MB (3 mg/kg) or vehicle once daily for 3 months. MB treatment significantly prevented transgene-associated behavioral impairment, including hyperactivity, decreased object recognition, and defective spatial working and reference memory, but it did not alter nontransgenic mouse behavior. Moreover, brain parenchymal and cerebral vascular ß-amyloid deposits as well as levels of various Aß species, including oligomers, were mitigated in MB-treated PSAPP mice. These effects occurred with inhibition of amyloidogenic APP proteolysis. Specifically, ß-carboxyl-terminal APP fragment and ß-site APP cleaving enzyme 1 protein expression and activity were attenuated. Additionally, treatment of Chinese hamster ovary cells overexpressing human wild-type APP with MB significantly decreased Aß production and amyloidogenic APP proteolysis. These results underscore the potential for oral MB treatment against AD-related cerebral amyloidosis by modulating the amyloidogenic pathway.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Amiloidosis/tratamiento farmacológico , Encefalopatías/tratamiento farmacológico , Cognición/efectos de los fármacos , Azul de Metileno/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidosis/patología , Amiloidosis/psicología , Animales , Encefalopatías/patología , Encefalopatías/psicología , Células CHO , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteolisis
15.
Can J Microbiol ; 60(6): 355-61, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24848979

RESUMEN

To investigate the effect of the luxS gene on the expression of virulence factors in Shiga-like toxin producing and verotoxin-producing Escherichia coli, the luxS gene from E. coli 107/86 (wild type, O139:H1:F18ab, Stx2e) was deleted. The successful deletion of luxS was confirmed by bioluminescence assays. The luxS deletion mutant exhibited changed flagella-related phenotypes, like impaired expression of flagella, decreased flagella motility, reduced biofilm formation, and reduced ability to induce pro-immunity response in host cells, which were restored after complementation with the intact luxS gene. The mutant strain also displayed attenuated production of Stx2e. This study provides new information to the crucial function of luxS in regulating Shiga-like toxin producing E. coli virulence.


Asunto(s)
Proteínas Bacterianas/genética , Liasas de Carbono-Azufre/genética , Flagelos/fisiología , Percepción de Quorum/genética , Toxinas Shiga/biosíntesis , Escherichia coli Shiga-Toxigénica/genética , Animales , Proteínas Bacterianas/fisiología , Liasas de Carbono-Azufre/fisiología , Chlorocebus aethiops , Flagelos/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Mediciones Luminiscentes , Toxinas Shiga/genética , Escherichia coli Shiga-Toxigénica/metabolismo , Escherichia coli Shiga-Toxigénica/patogenicidad , Células Vero , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
16.
Neuroendocrinology ; 99(3-4): 139-55, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24718029

RESUMEN

All reproductively competent adults have gone through puberty. While key genes and signaling pathways that lead to the onset of sexual maturation are known, the molecular mechanisms that determine when an individual enters puberty are only beginning to be understood. Both genetic and environmental factors determine the timing of puberty. New advances in understanding how environmentally sensitive, yet highly heritable developmental processes are regulated have come from the field of epigenetics. Of note, studies investigating the epigenetic control of the onset of puberty suggest that epigenetic repression of key inhibitory loci may play a fundamental role in the initiation of puberty. Current technologies that not only read out the DNA sequence, but also determine how the DNA is modified in response to the environment, promise new insight into how puberty is regulated, including the identification and understanding of gene regulatory networks that control the biological pathways affecting pubertal timing. Here we review the findings to date and discuss how epigenetic investigation can further our understanding of this fundamental aspect of human development.


Asunto(s)
Epigénesis Genética/fisiología , Epigenómica , Pubertad/fisiología , Maduración Sexual/fisiología , Animales , Humanos
17.
Br J Med Med Res ; 4(1): 416-432, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24466509

RESUMEN

AIMS: Implicated in autoimmune encephalitis, neuromyotonia and genetic forms of autism, here we report that contactin-associated protein-like 2 (CNTNAP2) contains a potential autoepitope within the extracellular region. METHODOLOGY: CNTNAP2 sequence-similar regions (CSSRs) from human pathogens were identified. Sera from autistic and control children were obtained and analyzed for the presence of antibodies able to bind CSSRs. One such candidate CSSR was evaluated for evidence of autoimmune responses to CNTNAP2 in a mouse model of acute infection. RESULTS: Autistic and control children sera contained antibodies able to discrete regions of CNTNAP2. In a murine model of acute infection, a CSSR derived from the N-terminal extracellular region of CNTNAP2 resulted in anti-CNTNAP2 antibody production, proinflammatory cytokine elevation, cerebellar and cortical white matter T-cell infiltration as well as motor dysfunction. CONCLUSION: Taken together, these data suggest that CNTNAP2 contains a potential autoepitope within the extracellular region.

18.
Am J Transl Res ; 5(6): 634-42, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24093060

RESUMEN

Accumulation of amyloid-ß (Aß) peptides (predominantly Aß40, 42) and their aggregation into plaques in the brain are thought to be the one of the major causes of Alzheimer's disease (AD). Originally discovered in our Chinese hamster ovary (CHO) cell line stably expressing human wild-type amyloid precursor protein (APP) (CHO/APPwt) cultures devoid of Aß production, we found that Mycoplasma selectively degrades soluble Aß in a time and dose (colony forming unit) dependent manner. Moreover, we fully characterized the Mycoplasma species as Mycoplasma hyorhinis (M. hyorhinis) by genetic and colony morphological analyses by light microscopy. Most interestingly, we attenuated the pathogenicity of M. hyorhinis by γ irradiation (3.5 Gy), and found that its ability to degrade Aß was retained. On the other hand, heated and sonicated M. hyorhinis failed to retain this ability to degrade Aß, suggesting that this degradation requires viable cells and likely a biologically active signaling pathway. In addition, we found that M. hyorhinis can degrade Aß produced in AD model mice (PSAPP mice) ex vivo. Finally, we found that irradiated (non-pathogenic) M. hyorhinis also can degrade Aß produced in PSAPP mice in vivo. These studies suggest that irradiated (non-pathogenic) M. hyorhinis can be a novel and alternative biological strategy for AD treatment.

19.
Microb Pathog ; 64: 18-22, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23933167

RESUMEN

The role of SEF14 fimbriae in virulence remains to be elucidated and in this study, we showed that sefA mutant constructed in the wild-type (WT) Salmonella enteritidis strain 50336 displayed increased invasion to IPEC-J2 cell lines and survival in mouse peritoneal macrophages, and the lethal dose 50% (LD50) in 6-week-old Balb/c mice intra-peritoneally injected with WT S. enteritidis strain decreased significantly upon deletion of sefA indicating their role in virulence. Overall, these results demonstrated that expression of sefA of SEF14 fimbriae enhances S. enteritidis adhesion to epithelial cells and survival in macrophages and contributes to S. enteritidis virulence in mice.


Asunto(s)
Proteínas Fimbrias/metabolismo , Salmonella enteritidis/patogenicidad , Factores de Virulencia/metabolismo , Animales , Células Cultivadas , Células Epiteliales/microbiología , Dosificación Letal Mediana , Macrófagos Peritoneales/microbiología , Ratones , Ratones Endogámicos BALB C , Salmonelosis Animal , Porcinos , Virulencia
20.
PLoS One ; 8(8): e71913, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977176

RESUMEN

Our previous studies showed that the green tea-derived polyphenolic compound (-)-epigallocatechin-3 gallate (EGCG) reduces amyloid-ß (Aß) production in both neuronal and mouse Alzheimer's disease (AD) models in concert with activation of estrogen receptor-α/phosphatidylinositide 3-kinase/protein kinase B (ERα/PI3K/Akt) signaling and anti-amyloidogenic amyloid precursor protein (APP) α-secretase (a disintegrin and metallopeptidase domain-10, ADAM10) processing. Since the gallate moiety in EGCG may correspond to the 7α position of estrogen, thereby facilitating ER binding, we extensively screened the effect of other gallate containing phenolic compounds on APP anti-amyloidogenic processing. Octyl gallate (OG; 10 µM), drastically decreased Aß generation, in concert with increased APP α-proteolysis, in murine neuron-like cells transfected with human wild-type APP or "Swedish" mutant APP. OG markedly increased production of the neuroprotective amino-terminal APP cleavage product, soluble APP-α (sAPPα). In accord with our previous study, these cleavage events were associated with increased ADAM10 maturation and reduced by blockade of ERα/PI3k/Akt signaling. To validate these findings in vivo, we treated Aß-overproducing Tg2576 mice with OG daily for one week by intracerebroventricular injection and found decreased Aß levels associated with increased sAPPα. These data indicate that OG increases anti-amyloidogenic APP α-secretase processing by activation of ERα/PI3k/Akt signaling and ADAM10, suggesting that this compound may be an effective treatment for AD.


Asunto(s)
Proteínas ADAM/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Receptor alfa de Estrógeno/metabolismo , Ácido Gálico/análogos & derivados , Proteínas de la Membrana/metabolismo , Proteína ADAM10 , Enfermedad de Alzheimer/metabolismo , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Receptor alfa de Estrógeno/antagonistas & inhibidores , Femenino , Ácido Gálico/farmacología , Humanos , Masculino , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...