Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33737393

RESUMEN

Neurons are postmitotic cells. Reactivation of the cell cycle by neurons has been reported in Alzheimer's disease (AD) brains and models. This gave rise to the hypothesis that reentering the cell cycle renders neurons vulnerable and thus contributes to AD pathogenesis. Here, we use the fluorescent ubiquitination-based cell cycle indicator (FUCCI) technology to monitor the cell cycle in live neurons. We found transient, self-limited cell cycle reentry activity in naive neurons, suggesting that their postmitotic state is a dynamic process. Furthermore, we observed a diverse response to oligomeric amyloid-ß (oAß) challenge; neurons without cell cycle reentry activity would undergo cell death without activating the FUCCI reporter, while neurons undergoing cell cycle reentry activity at the time of the oAß challenge could maintain and increase FUCCI reporter signal and evade cell death. Accordingly, we observed marked neuronal FUCCI positivity in the brains of human mutant Aß precursor protein transgenic (APP23) mice together with increased neuronal expression of the endogenous cell cycle control protein geminin in the brains of 3-mo-old APP23 mice and human AD brains. Taken together, our data challenge the current view on cell cycle in neurons and AD, suggesting that pathways active during early cell cycle reentry in neurons protect from Aß toxicity.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ciclo Celular/fisiología , Neuronas/fisiología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Péptidos beta-Amiloides/toxicidad , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Biomarcadores , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Ratones , Ratones Transgénicos
2.
Pharmacol Res ; 187: 106631, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36586644

RESUMEN

According to the ß-amyloid (Aß) hypothesis of Alzheimer's disease (AD), brain Aß accumulation is the primary cascade event leading to cognitive deficit and dementia. Numerous anti-Aß drugs either inhibiting production or aggregation of Aß or stimulating its clearance have failed to show clinical benefit in large scale AD trials, with ß- and γ-secretase inhibitors consistently worsening cognitive and clinical decline. In June 2021, the FDA approved aducanumab, an anti-Aß monoclonal antibody for early AD based on its ability to reduce brain amyloid plaques, while two other amyloid-clearing antibodies (lecanemab and donanemab) have recently produced encouraging cognitive and clinical results. We reviewed AD trials using PubMed, meeting abstracts and ClinicalTrials.gov and evaluated the effects of such drugs on cerebrospinal fluid (CSF) Aß levels, correlating them with cognitive effects. We found that ß-secretase and γ-secretase inhibitors produce detrimental cognitive effects by significantly reducing CSF Aß levels. We speculate that monoclonal antibodies targeting Aß protofibrils, fibrils or plaques may improve cognitive performance in early AD by increasing soluble Aß levels through Aß aggregate disassembly and/or stabilization of existing Aß monomers.These findings suggest that the real culprit in AD may be decreased levels of soluble monomeric Aß due to sequestration into brain Aß aggregates and plaques.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Péptidos beta-Amiloides , Cognición
3.
Biochem J ; 478(7): 1471-1484, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33769438

RESUMEN

Tau pathology initiates in defined brain regions and is known to spread along neuronal connections as symptoms progress in Alzheimer's disease (AD) and other tauopathies. This spread requires the release of tau from donor cells, but the underlying molecular mechanisms remained unknown. Here, we established the interactome of the C-terminal tail region of tau and identified syntaxin 8 (STX8) as a mediator of tau release from cells. Similarly, we showed the syntaxin 6 (STX6), part of the same SNARE family as STX8 also facilitated tau release. STX6 was previously genetically linked to progressive supranuclear palsy (PSP), a tauopathy. Finally, we demonstrated that the transmembrane domain of STX6 is required and sufficient to mediate tau secretion. The differential role of STX6 and STX8 in alternative secretory pathways suggests the association of tau with different secretory processes. Taken together, both syntaxins, STX6 and STX8, may contribute to AD and PSP pathogenesis by mediating release of tau from cells and facilitating pathology spreading.


Asunto(s)
Enfermedad de Alzheimer/patología , Dominios y Motivos de Interacción de Proteínas , Proteínas Qa-SNARE/metabolismo , Vías Secretoras , Tauopatías/patología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Unión Proteica , Proteínas Qa-SNARE/genética , Tauopatías/genética , Tauopatías/metabolismo , Proteínas tau/genética
4.
Alzheimers Dement ; 18(5): 1008-1037, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34533272

RESUMEN

INTRODUCTION: Primary tauopathies are neurological disorders in which tau protein deposition is the predominant pathological feature. Alzheimer's disease is a secondary tauopathy with tau forming hyperphosphorylated insoluble aggregates. Tau pathology can propagate from region to region in the brain, while alterations in tau processing may impair tau physiological functions. METHODS: We reviewed literature on tau biology and anti-tau drugs using PubMed, meeting abstracts, and ClnicalTrials.gov. RESULTS: The past 15 years have seen >30 drugs interfering with tau aggregation, processing, and accumulation reaching the clinic. Initial results with tau aggregation inhibitors and anti-tau monoclonal antibodies have not shown clinical efficacy. DISCUSSION: The reasons for these clinical failures are unclear but could be linked to the clearing of physiological forms of tau by non-specific drugs. Research is now concentrating efforts on developing reliable translational animal models and selective compounds targeting specific tau epitopes, neurotoxic tau aggregates, and post-translational tau modifications.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Humanos , Tauopatías/patología , Proteínas tau/metabolismo
5.
Am J Physiol Cell Physiol ; 316(2): C235-C245, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30485136

RESUMEN

Carnosine (CAR) is an endogenous dipeptide physiologically present in excitable tissues, such as central nervous system (CNS) and muscle. CAR is acknowledged as a substrate involved in many homeostatic pathways and mechanisms and, due to its biochemical properties, as a molecule intertwined with the homeostasis of heavy metals such as copper (Cu). In CNS, Cu excess and dysregulation imply oxidative stress, free-radical production, and functional impairment leading to neurodegeneration. Here, we report that CAR intercepts the regulatory routes of Cu homeostasis in nervous cells and tissues. Specifically, in a murine neuron-derived cell model, i.e., the B104 neuroblastoma cells, extracellular CAR exposure up to 24 h influenced intracellular Cu entry and affected (downregulated) the key Cu-sensing system, consisting of the gene coding for the Slc31a1 transmembrane Cu importer (alias Ctr1), and the gene coding for the Cu-responsive transcription factor Sp1 ( Sp1). Also, CAR exposure upregulated CAR biosynthesis ( Carns1), extracellular degradation ( Cndp1), and transport ( Slc15a4, alias Pht1) genes and elicited CAR intracellular accumulation, contributing to the outline of functional association between CAR and Cu within the cell. Interestingly, the same gene modulation scheme acting in vitro operates in vivo in brains of mice undergoing dietary administration of CAR in drinking water for 2 wk. Overall, our findings describe for the first time a regulatory interaction between CAR and Cu pathways in CNS and indicate CAR as a novel active molecule within the network of ligands and chaperones that physiologically regulate Cu homeostasis.


Asunto(s)
Encéfalo/metabolismo , Carnosina/farmacología , Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Factor de Transcripción Sp1/metabolismo , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Células Cultivadas , Transportador de Cobre 1 , Relación Dosis-Respuesta a Droga , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C
6.
Am J Pathol ; 188(6): 1447-1456, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29577934

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressing and fatal disease characterized by muscular atrophy because of loss of upper and lower motor neurons. Histopathologically, most patients with ALS have abnormal cytoplasmic accumulation and aggregation of the nuclear RNA-regulating protein TAR DNA-binding protein 43 (TDP-43). Pathogenic mutations in the TARDBP gene that encode TDP-43 have been identified in familial ALS. We have previously reported transgenic mice with neuronal expression of human TDP-43 carrying the pathogenic A315T mutation (iTDP-43A315T mice), presenting with early-onset motor deficits in adolescent animals. Here, we analyzed aged iTDP-43A315T mice, focusing on the spatiotemporal profile and progression of neurodegeneration in upper and lower motor neurons. Magnetic resonance imaging and histologic analysis revealed a differential loss of upper motor neurons in a hierarchical order as iTDP-43A315T mice aged. Furthermore, we report progressive gait problems, profound motor deficits, and muscle atrophy in aged iTDP-43A315T mice. Despite these deficits and TDP-43 pathologic disorders in lower motor neurons, stereological analysis did not show cell loss in spinal cords. Taken together, neuronal populations in aging iTDP-43A315T mice show differential susceptibility to the expression of human TDP-43A315T.


Asunto(s)
Sistema Nervioso Central/patología , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Trastornos Motores/patología , Atrofia Muscular/patología , Enfermedades Neurodegenerativas/patología , Envejecimiento , Animales , Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Motores/genética , Trastornos Motores/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Análisis Espacio-Temporal
7.
Int J Mol Sci ; 19(6)2018 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-29890740

RESUMEN

In excitable tissues, the endogenous dipeptide carnosine (CAR, ß-Ala-l-His) sustains homeostatic responses to various challenges. By eliciting hypoglycemic effects via actions on the autonomic nervous system and protection of pancreatic beta-cells, CAR is also relevant in diabetes. We investigated the expression of genes involved in CAR biosynthesis, degradation, and membrane transport pathways, in the pancreas and brains of mice treated with streptozotocin (STZ) and then exposed to dietary CAR. We induced hyperglycemia by STZ intraperitoneal injections; then, STZ-treated mice received drinking water with or without CAR for two weeks. We report that CAR administration elicits beneficial effects on blood glucose levels and weight loss in STZ-treated mice and, remarkably, on the insulin gene products in the pancreas, preserving gene expression from STZ challenge. Also, we describe mRNA downregulation of the Slc15a2/Pept2 (dipeptide transporter) and Cndp2 (intracellular dipeptidase) genes in the pancreas of hyperglycemic mice, and dysregulation of Carns1 (CAR synthase), Pept2 and Cndp2 in brains; interestingly, dietary CAR elicits counteracting effects. These expression patterns associate with variations of CAR content in tissues of mice. Overall, our report suggests a direct role of CAR in the diabetes-affected pancreas and in the diabetes-targeted CNS, proposing (dys)regulation of CAR's homeostasis as a marker condition.


Asunto(s)
Encéfalo/metabolismo , Carnosina/genética , Dieta , Homeostasis/genética , Páncreas/metabolismo , Administración Oral , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Carnosina/administración & dosificación , Hiperglucemia/sangre , Hiperglucemia/patología , Insulina/genética , Masculino , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Estreptozocina , Extractos de Tejidos
9.
Acta Neuropathol ; 130(5): 661-78, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26437864

RESUMEN

The nuclear transactive response DNA-binding protein 43 (TDP-43) undergoes relocalization to the cytoplasm with formation of cytoplasmic deposits in neurons in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Pathogenic mutations in the TDP-43-encoding TARDBP gene in familial ALS as well as non-mutant human TDP-43 have been utilized to model FTD/ALS in cell culture and animals, including mice. Here, we report novel A315T mutant TDP-43 transgenic mice, iTDP-43(A315T), with controlled neuronal over-expression. Constitutive expression of human TDP-43(A315T) resulted in pronounced early-onset and progressive neurodegeneration, which was associated with compromised motor performance, spatial memory and disinhibition. Muscle atrophy resulted in reduced grip strength. Cortical degeneration presented with pronounced astrocyte activation. Using differential protein extraction from iTDP-43(A315T) brains, we found cytoplasmic localization, fragmentation, phosphorylation and ubiquitination and insolubility of TDP-43. Surprisingly, suppression of human TDP-43(A315T) expression in mice with overt neurodegeneration for only 1 week was sufficient to significantly improve motor and behavioral deficits, and reduce astrogliosis. Our data suggest that functional deficits in iTDP-43(A315T) mice are at least in part a direct and transient effect of the presence of TDP-43(A315T). Furthermore, it illustrates the compensatory capacity of compromised neurons once transgenic TDP-43 is removed, with implications for future treatments.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Degeneración Lobar Frontotemporal/fisiopatología , Mutación , Recuperación de la Función/fisiología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/patología , Astrocitos/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Doxiciclina , Femenino , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/patología , Gliosis/patología , Gliosis/fisiopatología , Fuerza de la Mano/fisiología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Neuronas/metabolismo , Neuronas/patología , Memoria Espacial/fisiología
10.
Neuron ; 112(8): 1249-1264.e8, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38366598

RESUMEN

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are characterized by cytoplasmic deposition of the nuclear TAR-binding protein 43 (TDP-43). Although cytoplasmic re-localization of TDP-43 is a key event in the pathogenesis of ALS/FTD, the underlying mechanisms remain unknown. Here, we identified a non-canonical interaction between 14-3-3θ and TDP-43, which regulates nuclear-cytoplasmic shuttling. Neuronal 14-3-3θ levels were increased in sporadic ALS and FTD with TDP-43 pathology. Pathogenic TDP-43 showed increased interaction with 14-3-3θ, resulting in cytoplasmic accumulation, insolubility, phosphorylation, and fragmentation of TDP-43, resembling pathological changes in disease. Harnessing this increased affinity of 14-3-3θ for pathogenic TDP-43, we devised a gene therapy vector targeting TDP-43 pathology, which mitigated functional deficits and neurodegeneration in different ALS/FTD mouse models expressing mutant or non-mutant TDP-43, including when already symptomatic at the time of treatment. Our study identified 14-3-3θ as a mediator of cytoplasmic TDP-43 localization with implications for ALS/FTD pathogenesis and therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Animales , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/metabolismo , Neuronas/metabolismo
11.
Expert Opin Drug Discov ; 16(7): 727-735, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33653187

RESUMEN

Introduction: As the global burden of Alzheimer's disease (AD) grows, an effective disease-modifying therapy remains a distant prospect following the repeated failure of multiple therapeutics targeting ß-amyloid and (it seems) tau over many years of costly effort. The repeated failure of single-target therapies to meaningfully modify disease progression raises major questions about the validity of many aspects of drug development in this area, especially target selection.Area covered: The authors explore the critical questions raised by a review of the collective experience to date, relating to why findings with non-clinical models and clinical biomarkers so frequently fail to translate to positive outcomes in clinical trials, which alternatives should be considered, and how we can design and conduct clinical trials that can successfully identify and quantify meaningful benefits in the future.Expert opinion: It is our opinion that we must recognize and accept the need to consider less specific, more multimodal approaches to addressing neurodegeneration in AD if we are to make progress - and we must avoid repeating the well intentioned, but ultimately erroneous, assumptions of the past.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides , Progresión de la Enfermedad , Desarrollo de Medicamentos , Descubrimiento de Drogas , Humanos
12.
Cancers (Basel) ; 13(5)2021 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-33673630

RESUMEN

Several trials have tried for decades to improve the outcome of extensive disease small cell lung cancer (ED-SCLC) through attempts to modify the standard treatments. Nevertheless, platinum/etoposide combination and topotecan have remained respectively the first and the second line standard treatments for the last 40 years. With the advent of immunotherapy, this scenario has finally changed. Our review aims to provide an overview of the primary studies on the actual therapeutic strategies available for ED-SCLC patients, and to highlight emerging evidence supporting the use of immunotherapy in SCLC patients.

13.
Sci Rep ; 11(1): 9616, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33953334

RESUMEN

Long pentraxin PTX3, a pattern recognition molecule involved in innate immune responses, is upregulated by pro-inflammatory stimuli, contributors to secondary damage in traumatic brain injury (TBI). We analyzed PTX3 involvement in mice subjected to controlled cortical impact, a clinically relevant TBI mouse model. We measured PTX3 mRNA and protein in the brain and its circulating levels at different time point post-injury, and assessed behavioral deficits and brain damage progression in PTX3 KO mice. PTX3 circulating levels significantly increased 1-3 weeks after injury. In the brain, PTX3 mRNA was upregulated in different brain areas starting from 24 h and up to 5 weeks post-injury. PTX3 protein significantly increased in the brain cortex up to 3 weeks post-injury. Immunohistochemical analysis showed that, 48 h after TBI, PTX3 was localized in proximity of neutrophils, likely on neutrophils extracellular traps (NETs), while 1- and 2- weeks post-injury PTX3 co-localized with fibrin deposits. Genetic depletion of PTX3 did not affect sensorimotor deficits up to 5 weeks post-injury. At this time-point lesion volume and neuronal count, axonal damage, collagen deposition, astrogliosis, microglia activation and phagocytosis were not different in KO compared to WT mice. Members of the long pentraxin family, neuronal pentraxin 1 (nPTX1) and pentraxin 4 (PTX4) were also over-expressed in the traumatized brain, but not neuronal pentraxin 2 (nPTX2) or short pentraxins C-reactive protein (CRP) and serum amyloid P-component (SAP). The long-lasting pattern of activation of PTX3 in brain and blood supports its specific involvement in TBI. The lack of a clear-cut phenotype in PTX3 KO mice may depend on the different roles of this protein, possibly involved in inflammation early after injury and in repair processes later on, suggesting distinct functions in acute phases versus sub-acute or chronic phases. Brain long pentraxins, such as PTX4-shown here to be overexpressed in the brain after TBI-may compensate for PTX3 absence.


Asunto(s)
Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Proteína C-Reactiva/metabolismo , Neuronas/metabolismo , Componente Amiloide P Sérico/metabolismo , Regulación hacia Arriba , Animales , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Proteína C-Reactiva/genética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Neuronas/patología , Neutrófilos/metabolismo , Componente Amiloide P Sérico/genética
14.
Cancers (Basel) ; 13(9)2021 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-33922215

RESUMEN

Tailored therapies based on the identification of molecular targets currently represent a well-established therapeutic scenario in the treatment of non-small cell lung cancer (NSCLC) patients. However, while aiming to improve patients' response to therapy, development of resistance is frequently observed in daily clinical practice. Intratumoral heterogeneity is a frequent event in NSCLC, responsible for several critical issues in patients' diagnosis and treatment. Advances in single-cell sequencing technologies have allowed in-depth profiling of tumors and attributed intratumoral heterogeneity to genetic, epigenetic, and protein modification driven diversities within cancer cell populations. This review highlights current research on the biological role of tumor heterogeneity and its impact on the development of acquired resistance in NSCLC patients.

15.
J Med Chem ; 64(12): 8333-8353, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34097384

RESUMEN

Acid-sensitive ion channels (ASICs) are sodium channels partially permeable to Ca2+ ions, listed among putative targets in central nervous system (CNS) diseases in which a pH modification occurs. We targeted novel compounds able to modulate ASIC1 and to reduce the progression of ischemic brain injury. We rationally designed and synthesized several diminazene-inspired diaryl mono- and bis-guanyl hydrazones. A correlation between their predicted docking affinities for the acidic pocket (AcP site) in chicken ASIC1 and their inhibition of homo- and heteromeric hASIC1 channels in HEK-293 cells was found. Their activity on murine ASIC1a currents and their selectivity vs mASIC2a were assessed in engineered CHO-K1 cells, highlighting a limited isoform selectivity. Neuroprotective effects were confirmed in vitro, on primary rat cortical neurons exposed to oxygen-glucose deprivation followed by reoxygenation, and in vivo, in ischemic mice. Early lead 3b, showing a good selectivity for hASIC1 in human neurons, was neuroprotective against focal ischemia induced in mice.


Asunto(s)
Bloqueadores del Canal Iónico Sensible al Ácido/uso terapéutico , Canales Iónicos Sensibles al Ácido/metabolismo , Guanidinas/uso terapéutico , Hidrazonas/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Bloqueadores del Canal Iónico Sensible al Ácido/síntesis química , Bloqueadores del Canal Iónico Sensible al Ácido/metabolismo , Canales Iónicos Sensibles al Ácido/química , Animales , Sitios de Unión , Células CHO , Pollos , Cricetulus , Diseño de Fármacos , Guanidinas/síntesis química , Guanidinas/metabolismo , Células HEK293 , Humanos , Hidrazonas/síntesis química , Hidrazonas/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Estructura Molecular , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/metabolismo , Unión Proteica , Ratas , Relación Estructura-Actividad
16.
Expert Opin Drug Discov ; 15(11): 1241-1251, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32686970

RESUMEN

INTRODUCTION: Alzheimer's Disease (AD) represents a large and growing challenge to patients, carers and healthcare systems, yet extensive efforts to develop therapeutics to modify its course have been met with repeated failure in recent decades. Although the evident presence of accumulated ß-amyloid (Aß) in AD brains has singled it out as an obvious therapeutic target, the effective reduction of plaque load or soluble Aß by numerous drug candidates has not produced commensurate clinical benefits - calling into question the Aß cascade hypothesis of AD. A similar path is now unfolding in the pursuit of therapeutics targeting hyperphosphorylated tau-comprised neurofibrillary tangles. AREAS COVERED: This perspective reviews the basis of the Aß cascade hypothesis of AD and how clinical trials of anti-Aß drugs have failed to support it, and reflects upon the early findings suggesting that a similar path is being followed with therapeutics targeting tau. Other potential approaches to identifying therapeutics for AD are explored herein. EXPERT OPINION: The relevance of the Aß cascade hypothesis to the development of therapeutics for AD appears disproven. Drugs targeting tau appear to be suffering the same fate but may yet produce better results. Alternative approaches are being pursued, some of them with initial small-scale, but promising, results.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Desarrollo de Medicamentos , Descubrimiento de Drogas , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Terapia Molecular Dirigida , Ovillos Neurofibrilares/metabolismo , Proteínas tau/metabolismo
17.
Alzheimers Dement (N Y) ; 6(1): e12004, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32211508

RESUMEN

Therapeutic plasma exchange, consisting of removing blood plasma and exchanging it with donated blood products, has been proposed for treating Alzheimer's disease (AD) to remove senescent or toxic factors. In preclinical studies, administration of plasma from young healthy mice to AD transgenic mice improved cognitive deficits without affecting brain amyloid plaques. Initial encouraging results have been collected in a double-blind, placebo-controlled study in nine AD patients receiving young plasma. In a 14-month double-blind, placebo-controlled study in 322 AD patients, multiple infusions with plasma enriched with albumin with or without immunoglobulins slowed cognitive, functional, and clinical decline, especially in moderately affected patients. Clinical trials of plasma fractions containing hypothetically beneficial proteins are also under way. These initial positive clinical results need to be confirmed in larger and more rigorous controlled studies in which the possible benefits of plasma exchange approaches can be weighed against the intrinsic side effects of repetitive infusion procedures.

18.
Diagnostics (Basel) ; 10(12)2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33333743

RESUMEN

Lung cancer remains the first cause of cancer-related deaths worldwide. Thanks to the improvement in the knowledge of the biology of non-small cell lung cancer (NSCLC), patients' survival has significantly improved. A growing number of targetable molecular alterations have been identified. Next-generation sequencing (NGS) has become one of the methodologies entered in clinical practice and was recently recommended by the European society for medical oncology (ESMO) to perform a comprehensive molecular characterization in patients with cancer. The current review provides an overview of the clinical trials that have explored the impact of NGS in patients with cancer, its limits, and advantages.

20.
PLoS One ; 11(10): e0163236, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27736877

RESUMEN

Several mouse lines with knockout of the tau-encoding MAPT gene have been reported in the past; they received recent attention due to reports that tau reduction prevented Aß-induced deficits in mouse models of Alzheimer's disease. However, the effects of long-term depletion of tau in vivo remained controversial. Here, we used the tau-deficient GFP knockin line Mapttm1(EGFP)kit on a pure C57Bl/6 background and subjected a large cohort of males and females to a range of motor, memory and behavior tests and imaging analysis, at the advanced age of over 16 months. Neither heterozygous nor homozygous Mapttm1(EGFP)kit mice presented with deficits or abnormalities compared to wild-type littermates. Differences to reports using other tau knockout models may be due to different genetic backgrounds, respective gene targeting strategies or other confounding factors, such as nutrition. To this end, we report no functional or morphological deficits upon tau reduction or depletion in aged mice.


Asunto(s)
Enfermedad de Alzheimer/genética , Técnicas de Inactivación de Genes , Proteínas tau/genética , Enfermedad de Alzheimer/fisiopatología , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Locomoción , Masculino , Aprendizaje por Laberinto , Memoria , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA