Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Headache Pain ; 25(1): 126, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085771

RESUMEN

BACKGROUND: Pituitary adenylate cyclase-activating peptide (PACAP) is a neuropeptide pivotal in migraine pathophysiology and is considered a promising new migraine drug target. Although intravenous PACAP triggers migraine attacks and a recent phase II trial with a PACAP-inhibiting antibody showed efficacy in migraine prevention, targeting the PACAP receptor PAC1 alone has been unsuccessful. The present study investigated the role of three PACAP receptors (PAC1, VPAC1 and VPAC2) in inducing migraine-relevant hypersensitivity in mice. METHODS: Hindpaw hypersensitivity was induced by repeated PACAP38 injections. Tactile sensitivity responses were quantified using von Frey filaments in three knockout (KO) mouse strains, each lacking one of the PACAP-receptors (Ntotal = 160). Additionally, ex vivo wire myography was used to assess vasoactivity of the carotid artery, and gene expression of PACAP receptors was examined by qPCR. RESULTS: PACAP38 induced hypersensitivity in WT controls (p < 0.01) that was diminished in VPAC1 and VPAC2 KO mice (p < 0.05). In contrast, PAC1 KO mice showed similar responses to WT controls (p > 0.05). Myograph experiments supported these findings showing diminished vasoactivity in VPAC1 and VPAC2 KO mice. We found no upregulation of the non-modified PACAP receptors in KO mice. CONCLUSIONS: This study assessed all three PACAP receptors in a migraine mouse model and suggests a significant role of VPAC receptors in migraine pathophysiology. The lack of hypersensitivity reduction in PAC1 KO mice suggests the involvement of other PACAP receptors or compensatory mechanisms. The results indicate that targeting only individual PACAP receptors may not be an effective migraine treatment.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Noqueados , Trastornos Migrañosos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Receptores de Tipo II del Péptido Intestinal Vasoactivo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo , Animales , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/fisiopatología , Trastornos Migrañosos/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/genética , Ratones , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/fisiopatología , Hiperalgesia/fisiopatología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Masculino , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Ratones Endogámicos C57BL , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Miembro Posterior/fisiopatología
2.
J Biol Chem ; 294(25): 9679-9688, 2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31053641

RESUMEN

The unfolded protein response (UPR) is commonly associated with a range of neurodegenerative diseases, and targeting UPR components has been suggested as a therapeutic strategy. The UPR surveys protein folding within the endoplasmic reticulum. However, many of the misfolded proteins that accumulate in neurodegeneration are localized so that they do not directly cause endoplasmic reticulum triggers that activate this pathway. Here, using a transgenic mouse model and primary cell cultures along with quantitative PCR, immunoblotting, and immunohistochemistry, we tested whether the UPR is induced in in vivo and in vitro murine models of tauopathy that are based on expression of mutant tauP301L We found no evidence for the UPR in the rTg4510 mouse model, in which mutant tau is transgenically expressed under the control of tetracycline-controlled transactivator protein. This observation was supported by results from acute experiments in which neuronal cultures expressed mutant tau and accumulated misfolded cytoplasmic tau aggregates but exhibited no UPR activation. These results suggest that the UPR is not induced as a response to tau misfolding and aggregation despite clear evidence for progressive cellular dysfunction and degeneration. We propose that caution is needed when evaluating the implied significance of the UPR as a critical determinant across major neurodegenerative diseases.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Tauopatías/patología , Respuesta de Proteína Desplegada/fisiología , Proteínas tau/metabolismo , Animales , Humanos , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/metabolismo , Neuronas/metabolismo , Fosforilación , Tauopatías/metabolismo , Proteínas tau/genética
3.
Rapid Commun Mass Spectrom ; 33(22): 1711-1721, 2019 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-31307118

RESUMEN

RATIONALE: The molecular complexity of tissue features several signal-suppression effects which reduce the ionization of analytes significantly and thereby weakens the quality of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS) imaging (MALDI imaging). We report a novel approach in MALDI imaging by reducing signal-suppression effects for the analysis of beta-amyloid (Aß) plaques, one pathological hallmark of Alzheimer's disease (AD). METHODS: We analyzed Aß proteoforms from postmortem AD brains and brains from transgenic mice (APPPS1-21) overexpressing familial AD mutations by combining two techniques: (1) laser capture microdissection (LCM) to accumulate Aß plaques and (2) phosphoric acid (PA) as additive to the super-2,5-dihydroxybenzoic acid matrix. RESULTS: LCM and MALDI-MS enabled tandem mass spectrometric fragmentation of stained Aß plaques. PA improved the signal-to-noise (S/N) ratio, especially of the Aß1-42 peptide, by three-fold compared with the standard matrix additive trifluoroacetic acid. The beneficial effect of the PA matrix additive in MALDI imaging was particularly important for AD brain tissue. We identified several significant differences in Aß plaque composition from AD compared with APPPS1-21, underlining the value of reducing signal-suppressing effects in MALDI imaging. CONCLUSIONS: We present a novel strategy for overcoming signal-suppression effects in MALDI imaging of Aß proteoforms.

4.
J Biol Chem ; 289(7): 4532-45, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24366862

RESUMEN

Prion diseases are characterized by accumulation of misfolded protein, gliosis, synaptic dysfunction, and ultimately neuronal loss. This sequence, mirroring key features of Alzheimer disease, is modeled well in ME7 prion disease. We used iTRAQ(TM)/mass spectrometry to compare the hippocampal proteome in control and late-stage ME7 animals. The observed changes associated with reactive glia highlighted some specific proteins that dominate the proteome in late-stage disease. Four of the up-regulated proteins (GFAP, high affinity glutamate transporter (EAAT-2), apo-J (Clusterin), and peroxiredoxin-6) are selectively expressed in astrocytes, but astrocyte proliferation does not contribute to their up-regulation. The known functional role of these proteins suggests this response acts against protein misfolding, excitotoxicity, and neurotoxic reactive oxygen species. A recent convergence of genome-wide association studies and the peripheral measurement of circulating levels of acute phase proteins have focused attention on Clusterin as a modifier of late-stage Alzheimer disease and a biomarker for advanced neurodegeneration. Since ME7 animals allow independent measurement of acute phase proteins in the brain and circulation, we extended our investigation to address whether changes in the brain proteome are detectable in blood. We found no difference in the circulating levels of Clusterin in late-stage prion disease when animals will show behavioral decline, accumulation of misfolded protein, and dramatic synaptic and neuronal loss. This does not preclude an important role of Clusterin in late-stage disease, but it cautions against the assumption that brain levels provide a surrogate peripheral measure for the progression of brain degeneration.


Asunto(s)
Astrocitos/metabolismo , Clusterina/biosíntesis , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Enfermedades por Prión/metabolismo , Proteoma/biosíntesis , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Astrocitos/patología , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Humanos , Ratones , Enfermedades por Prión/inducido químicamente , Enfermedades por Prión/patología
5.
Neuropathol Appl Neurobiol ; 41(4): 445-57, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25486988

RESUMEN

AIMS: Active amyloid-ß (Aß) immunotherapy in Alzheimer's disease (AD) induces removal of Aß and phosphorylated tau (ptau). Glycogen synthase kinase (GSK)-3ß is a kinase, responsible for phosphorylation of tau, activation of which can be induced by phosphorylated double-stranded RNA-dependent protein kinase (pPKR). Using a post-mortem cohort of immunized AD cases, we investigated the effect of Aß immunization on GSK-3ß expression and pPKR. METHODS: We immunostained 11 immunized AD cases and 28 unimmunized AD cases for active, inactive and total GSK-3ß, and for pPKR. Quantification of protein load was performed in the hippocampal region including CA1, subiculum and entorhinal cortex. RESULTS: All three areas showed a significant decrease in the three forms of GSK-3ß (P < 0.05) and a nonsignificant trend towards lower pPKR load in the immunized AD cases compared with the unimmunized AD cases. CONCLUSION: The lower GSK-3ß expression generated by Aß immunotherapy shows evidence of a modification of the signalling pathway induced by GSK-3ß leading to the overall reduction of tau, supporting the contention that in humans, GSK-3ß unifies Aß and tau-related neuropathology.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología , Vacunas contra el Alzheimer/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Neuronas/metabolismo , Neuronas/patología , Fosforilación
6.
Ann Neurol ; 75(5): 684-99, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24687915

RESUMEN

OBJECTIVE: Proteolytic cleavage of the amyloid precursor protein (APP) generates ß-amyloid (Aß) peptides. Prolonged accumulation of Aß in the brain underlies the pathogenesis of Alzheimer disease (AD) and is regarded as a principal target for development of disease-modifying therapeutics. METHODS: Using Chinese hamster ovary (CHO) APP751SW cells, we identified and characterized effects of 2-([pyridine-2-ylmethyl]-amino)-phenol (2-PMAP) on APP steady-state level and Aß production. Outcomes of 2-PMAP treatment on Aß accumulation and associated memory deficit were studied in APPSW /PS1dE9 AD transgenic model mice. RESULTS: In CHO APP751SW cells, 2-PMAP lowered the steady-state APP level and inhibited Aßx-40 and Aßx-42 production in a dose-response manner with a minimum effective concentration ≤ 0.5µM. The inhibitory effect of 2-PMAP on translational efficiency of APP mRNA into protein was directly confirmed using a 35S-methionine/cysteine metabolic labeling technique, whereas APP mRNA level remained unaltered. Administration of 2-PMAP to APPSW /PS1dE9 mice reduced brain levels of full-length APP and its C-terminal fragments and lowered levels of soluble Aßx-40 and Aßx-42 . Four-month chronic treatment of APPSW /PS1dE9 mice revealed no observable toxicity and improved animals' memory performance. 2-PMAP treatment also caused significant reduction in brain Aß deposition determined by both unbiased quantification of Aß plaque load and biochemical analysis of formic acid-extracted Aßx-40 and Aßx-42 levels and the level of oligomeric Aß. INTERPRETATION: We demonstrate the potential of modulating APP steady-state expression level as a safe and effective approach for reducing Aß deposition in AD transgenic model mice.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Regulación de la Expresión Génica , Placa Amiloide/prevención & control , Precursor de Proteína beta-Amiloide/fisiología , Animales , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/metabolismo , Placa Amiloide/patología , Presenilina-1/genética
7.
Am J Pathol ; 182(5): 1750-68, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23499462

RESUMEN

Accumulation of ß-amyloid (Aß) in the brain is a key event in Alzheimer disease pathogenesis. Apolipoprotein (Apo) E is a lipid carrier protein secreted by astrocytes, which shows inherent affinity for Aß and has been implicated in the receptor-mediated Aß uptake by neurons. To characterize ApoE involvement in the intraneuronal Aß accumulation and to investigate whether blocking the ApoE/Aß interaction could reduce intraneuronal Aß buildup, we used a noncontact neuronal-astrocytic co-culture system, where synthetic Aß peptides were added into the media without or with cotreatment with Aß12-28P, which is a nontoxic peptide antagonist of ApoE/Aß binding. Compared with neurons cultured alone, intraneuronal Aß content was significantly increased in neurons co-cultured with wild-type but not with ApoE knockout (KO) astrocytes. Neurons co-cultured with astrocytes also showed impaired intraneuronal degradation of Aß, increased level of intraneuronal Aß oligomers, and marked down-regulation of several synaptic proteins. Aß12-28P treatment significantly reduced intraneuronal Aß accumulation, including Aß oligomer level, and inhibited loss of synaptic proteins. Furthermore, we showed significantly reduced intraneuronal Aß accumulation in APPSW/PS1dE9/ApoE KO mice compared with APPSW/PS1dE9/ApoE targeted replacement mice that expressed various human ApoE isoforms. Data from our co-culture and in vivo experiments indicate an essential role of ApoE in the mechanism of intraneuronal Aß accumulation and provide evidence that ApoE/Aß binding antagonists can effectively prevent this process.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Neuronas/patología , Sinapsis/patología , Animales , Astrocitos/patología , Células Cultivadas , Técnicas de Cocultivo , Endocitosis , Espacio Extracelular/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Hipocampo/patología , Humanos , Ratones , Ratones Noqueados , Degeneración Nerviosa/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Fracciones Subcelulares/metabolismo , Sinapsis/metabolismo
8.
Mol Imaging Biol ; 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39093482

RESUMEN

PURPOSE: In the emerging field of antibody treatments for neurodegenerative diseases, reliable tools are needed to evaluate new therapeutics, diagnose and select patients, monitor disease progression, and assess therapy response. Immuno-PET combines the high affinity and exceptional specificity of monoclonal antibodies with the non-invasive imaging technique positron emission tomography (PET). Its application in neurodegenerative disease brain imaging has been limited due to the marginal uptake across the blood-brain barrier (BBB). The emergence of BBB-shuttle antibodies with enhanced uptake across the BBB extended immuno-PET to brain imaging. We recently reported about specific brain uptake of a bispecific aducanumab mTfR antibody in APP/PS1 TG mice using 89Zr-immuno-PET. However, a sufficient target-to-background ratio was reached at a relatively late scanning time point of 7 days post-injection. To investigate if a better target-to-background ratio could be achieved earlier, an aducanumab BBB-shuttle with a mutated Fc region for reduced FcRn affinity was evaluated. PROCEDURES: AduH310A-8D3 and Adu-8D3 were modified with DFO*-NCS and subsequently radiolabeled with 89Zr. The potential influence of the H310A mutation, modification with DFO*-NCS, and subsequent radiolabeling on the in vitro binding to amyloid-beta and mTfR1 was investigated via amyloid-beta peptide ELISA and FACS analysis using mTfR1 transfected CHO-S cells. Blood kinetics, brain uptake, in vivo PET imaging and target engagement of radiolabeled AduH310A-8D3 were evaluated and compared to non-mutated Adu-8D3 in APP/PS1 TG mice and wild-type animals as controls. RESULTS: Radiolabeling was performed with sufficient radiochemical yields and radiochemical purity. In vitro binding to amyloid-beta and mTfR1 showed no impairment. [89Zr]Zr-AduH310A-8D3 showed faster blood clearance and earlier differentiation of amyloid-beta-related brain uptake compared to [89Zr]Zr-Adu-8D3. However, only half of the brain uptake was observed for [89Zr]Zr-AduH310A-8D3. CONCLUSIONS: Although a faster blood clearance of AduH310A-8D3 was observed, it was concluded that no beneficial effects for 89Zr-immuno-PET imaging of brain uptake were obtained.

9.
J Alzheimers Dis ; 87(1): 259-272, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35275551

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disorder and the most common cause of dementia worldwide. Despite decades of investigation, the etiology of AD is not fully understood, although emerging evidence suggest that chronic environmental and psychological stress plays a role in the mechanisms and contributes to the risk of developing AD. Thus, dissecting the impact of stress on the brain could improve our understanding of the pathological mechanisms. OBJECTIVE: We aimed to study the effect of chronic stress on the hippocampal proteome in male APPPS1 transgenic mice and wildtype (WT) littermates. METHODS: APPPS1 and WT mice were subjected to 4 weeks of chronic stress followed by 3 weeks of continued diurnal disruption. Hippocampal tissue was used for proteomics analysis using label-free quantitative DIA based LC-MS/MS analysis. RESULTS: We identified significantly up- and downregulated proteins in both APPPS1 and WT mice exposed to chronic stress compared to the control groups. Via interaction network mapping, significant proteins could be annotated to specific pathways of mitochondrial function (oxidative phosphorylation and TCA cycle), metabolic pathways, AD pathway and synaptic functions (long term potentiation). In WT mice, chronic stress showed the highest impact on complex I of the oxidative phosphorylation pathway, while in APPPS1 mice this pathway was compromised broadly by chronic stress. CONCLUSION: Our data shows that chronic stress and amyloidosis additively contribute to mitochondrial damage in hippocampus. Although these results do not explain all effects of chronic stress in AD, they add to the scientific knowledge on the topic.


Asunto(s)
Enfermedad de Alzheimer , Espectrometría de Masas en Tándem , Enfermedad de Alzheimer/patología , Animales , Cromatografía Liquida , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Transgénicos
10.
Theranostics ; 12(16): 7067-7079, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36276653

RESUMEN

The accelerated approval of the monoclonal antibody (mAb) aducanumab as a treatment option for Alzheimer's Disease and the continued discussions about its efficacy have shown that a better understanding of immunotherapy for the treatment of neurodegenerative diseases is needed. 89Zr-immuno-PET could be a suitable tool to open new avenues for the diagnosis of CNS disorders, monitoring disease progression, and assessment of novel therapeutics. Herein, three different 89Zr-labeling strategies and direct radioiodination with 125I of a bispecific anti-amyloid-beta aducanumab derivate, consisting of aducanumab with a C-terminal fused anti-transferrin receptor binding single chain Fab fragment derived from 8D3 (Adu-8D3), were compared ex vivo and in vivo with regard to brain uptake and target engagement in an APP/PS1 Alzheimer's disease mouse model and wild type animals. Methods: Adu-8D3 and a negative control antibody, based on the HIV specific B12 antibody also carrying C-terminal fused 8D3 scFab (B12-8D3), were each conjugated with NCS-DFO, NCS-DFO*, or TFP-N-suc-DFO-Fe-ester, followed by radiolabeling with 89Zr. 125I was used as a substitute for 124I for labeling of both antibodies. 30 µg of radiolabeled mAb, corresponding to approximately 6 MBq 89Zr or 2.5 MBq 125I, were injected per mouse. PET imaging was performed 1, 3 and 7 days post injection (p.i.). All mice were sacrificed on day 7 p.i. and subjected to ex vivo biodistribution and brain autoradiography. Immunostaining on brain tissue was performed after autoradiography for further validation. Results: Ex vivo biodistribution revealed that the brain uptake of [89Zr]Zr-DFO*-NCS-Adu-8D3 (2.19 ±0.12 %ID/g) was as high as for its 125I-analog (2.21 ±0.15 %ID/g). [89Zr]Zr-DFO-NCS-Adu-8D3 and [89Zr]Zr-DFO-N-suc-Adu-8D3 showed significantly lower uptake (< 0.65 %ID/g), being in the same range as for the 89Zr-labeled controls (B12-8D3). Autoradiography of [89Zr]Zr-DFO*-NCS-Adu-8D3 and [125I]I-Adu-8D3 showed an amyloid-beta related granular uptake pattern of radioactivity. In contrast, the [89Zr]Zr-DFO-conjugates and the control antibody groups did not show any amyloid-beta related uptake pattern, indicating that DFO is inferior for 89Zr-immuno-PET imaging of the brain in comparison to DFO* for Adu-8D3. This was confirmed by day 7 PET images showing only amyloid-beta related brain uptake for [89Zr]Zr-DFO*-NCS-Adu-8D3. In wild type animals, such an uptake was not observed. Immunostaining showed a co-localization of all administered Adu-8D3 conjugates with amyloid-beta plaques. Conclusion: We successfully demonstrated that 89Zr-immuno-PET is suitable for imaging and quantifying amyloid-beta specific brain uptake using a bispecific aducanumab brain shuttling antibody, Adu-8D3, but only when using the novel chelator DFO*, and not DFO, for labeling with 89Zr.


Asunto(s)
Enfermedad de Alzheimer , Anticuerpos Biespecíficos , Animales , Ratones , Radioisótopos de Yodo , Quelantes , Deferoxamina , Circonio , Distribución Tisular , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/tratamiento farmacológico , Línea Celular Tumoral , Tomografía de Emisión de Positrones/métodos , Anticuerpos Monoclonales/uso terapéutico , Péptidos beta-Amiloides , Fragmentos Fab de Inmunoglobulinas , Ésteres
11.
Genes Brain Behav ; 20(8): e12766, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34382343

RESUMEN

Neuropsychiatric disturbances (NPDs) are considered hallmarks of Alzheimer's disease (AD). Nevertheless, treatment of these symptoms has proven difficult and development of safe and effective treatment options is hampered by the limited understanding of the underlying pathophysiology. Thus, robust preclinical models are needed to increase knowledge of NPDs in AD and develop testable hypotheses and novel treatment options. Abnormal activity of the hypothalamic-pituitary-adrenal (HPA) axis is implicated in many psychiatric symptoms and might contribute to both AD and NPDs development and progression. We aimed to establish a mechanistic preclinical model of NPD-like behavior in the APPPS1 mouse model of AD and wildtype (WT) littermates. In APPPS1 and WT mice, we found that chronic stress increased anxiety-like behavior and altered diurnal locomotor activity suggestive of sleep disturbances. Also, chronic stress activated the HPA axis, which, in WT mice, remained heightened for additional 3 weeks. Chronic stress caused irregular expression of circadian regulatory clock genes (BMAL1, PER2, CRY1 and CRY2) in both APPPS1 and WT mice. Interestingly, APPPS1 and WT mice responded differently to chronic stress in terms of expression of serotonergic markers (5-HT1A receptor and MAOA) and inflammatory genes (IL-6, STAT3 and ADMA17). These findings indicate that, although the behavioral response to chronic stress might be similar, the neurobiochemical response was different in APPPS1 mice, which is an important insight in the efforts to develop safe and effective treatments options for NPDs in AD patients. Further work is needed to substantiate these findings.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Estrés Psicológico/genética , Transcriptoma , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Ratones , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Receptor de Serotonina 5-HT1A/genética , Receptor de Serotonina 5-HT1A/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología
12.
J Alzheimers Dis ; 79(1): 249-265, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33252074

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is characterized by accumulation of amyloid-ß (Aß) species and deposition of senile plaques (SPs). Clinical trials with the anti-Aß antibody aducanumab have been completed recently. OBJECTIVE: To characterize the proteomic profile of SPs and surrounding tissue in a mouse model of AD in 10-month-old tgAPPPS1-21 mice after chronic treatment with aducanumab for four months with weekly dosing (10 mg/kg). METHODS: After observing significant reduction of SP numbers in hippocampi of aducanumab-treated mice, we applied a localized proteomic analysis by combining laser microdissection and liquid chromatography-tandem mass spectrometry (LC-MS/MS) of the remaining SPs in hippocampi. We microdissected three subregions, containing SPs, SP penumbra level 1, and an additional penumbra level 2 to follow the proteomic profile as gradient. RESULTS: In the aducanumab-treated mice, we identified 17 significantly regulated proteins that were associated with 1) mitochondria and metabolism (ACAT2, ATP5J, ETFA, EXOG, HK1, NDUFA4, NDUFS7, PLCB1, PPP2R4), 2) cytoskeleton and axons (ADD1, CAPZB, DPYSL3, MAG), 3) stress response (HIST1H1C/HIST1H1D, HSPA12A), and 4) AßPP trafficking/processing (CD81, GDI2). These pathways and some of the identified proteins are implicated in AD pathogenesis. Proteins associated with mitochondria and metabolism were mainly upregulated while proteins associated with AßPP trafficking/processing and stress response pathways were mainly downregulated, suggesting that aducanumab could lead to a beneficial proteomic profile around SPs in tgAPPPS1-21 mice. CONCLUSION: We identified novel proteomic patterns of SPs and surrounding tissue indicating that chronic treatment with aducanumab could inhibit Aß toxicity and increase phagocytosis and cell viability.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/efectos de los fármacos , Precursor de Proteína beta-Amiloide/metabolismo , Anticuerpos Monoclonales Humanizados/farmacología , Encéfalo/efectos de los fármacos , Placa Amiloide/metabolismo , Proteoma/efectos de los fármacos , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Cromatografía Liquida , Proteínas del Citoesqueleto/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Presenilina-1/genética , Transporte de Proteínas/efectos de los fármacos , Proteómica , Estrés Fisiológico/efectos de los fármacos , Espectrometría de Masas en Tándem
13.
Biochem Soc Trans ; 38(2): 545-51, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20298219

RESUMEN

Hyperphosphorylation of the microtubule-associated protein tau is a significant determinant in AD (Alzheimer's disease), where it is associated with disrupted axonal transport and probably causes synaptic dysfunction. Although less well studied, hyperphosphorylation has been observed in prion disease. We have investigated the expression of hyperphosphorylated tau in the hippocampus of mice infected with the ME7 prion agent. In ME7-infected animals, there is a selective loss of CA1 synapse, first discernable at 13 weeks of disease. There is a potential that dysfunctional axonal transport contributes to this synaptopathy. Thus investigating hyperphosphorylated tau that is dysfunctional in AD could illuminate whether and how they are significant in prion disease. We observed no differences in the levels of phosphorylated tau (using MC1, PHF-1 and CP13 antibodies) in detergent-soluble and detergent-insoluble fractions extracted from ME7- and NBH- (normal brain homogenate) treated animals across disease. In contrast, we observed an increase in phospho-tau staining for several epitopes using immunohistochemistry in ME7-infected hippocampal sections. Although the changes were not of the magnitude seen in AD tissue, clear differences for several phospho-tau species were seen in the CA1 and CA3 of ME7-treated animals (pSer(199-202)>pSer(214)>PHF-1 antibody). Temporally, these changes were restricted to animals at 20 weeks and none of the disease-related staining was associated with the axons or dendrites that hold CA1 synapses. These findings suggest that phosphorylation of tau at the epitopes examined does not underpin the early synaptic dysfunction. These data suggest that the changes in tau phosphorylation recorded here and observed by others relate to end-stage prion pathology when early dysfunctions have progressed to overt neuronal loss.


Asunto(s)
Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Ratones , Degeneración Nerviosa/metabolismo , Enfermedades por Prión/metabolismo , Proteínas tau/metabolismo , Animales , Humanos , Fosforilación , Enfermedades por Prión/etiología , Enfermedades por Prión/patología , Priones/efectos adversos , Proteínas tau/química , Proteínas tau/fisiología
15.
Front Aging Neurosci ; 12: 56, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32210790

RESUMEN

Alzheimer's disease (AD) is the most common form of dementia worldwide. It is mostly known for its devastating effect on memory and learning but behavioral alterations commonly known as neuropsychiatric disturbances (NPDs) are also characteristics of the disease. These include apathy, depression-like behavior, and sleep disturbances, and they all contribute to an increased caregiver burden and earlier institutionalization. The interaction between NPDs and AD pathology is not well understood, but the consensus is that they contribute to disease progression and faster decline. Consequently, recognizing and treating NPDs might improve AD pathology and increase the quality of life for both patients and caregivers. In this review article, we examine previous and current literature on apathy, depressive symptoms, and sleep disturbances in AD patients and preclinical AD mechanistic models. We hypothesize that tau accumulation, beta-amyloid (Aß) aggregation, neuroinflammation, mitochondrial damage, and loss of the locus coeruleus (LC)-norepinephrine (NE) system all collectively impact the development of NPDs and contribute synergistically to AD pathology. Targeting more than one of these processes might provide the most optimal strategy for treating NPDs and AD. The development of such clinical approaches would be preceded by preclinical studies, for which robust and reliable mechanistic models of NPD-like behavior are needed. Thus, developing effective preclinical research models represents an important step towards a better understanding of NPDs in AD.

16.
J Alzheimers Dis ; 73(1): 393-411, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31771055

RESUMEN

Amyloid plaques are one of the hallmarks of Alzheimer's disease (AD). The main constituent of amyloid plaques is amyloid-ß peptides, but a complex interplay of other infiltrating proteins also co-localizes. We hypothesized that proteomic analysis could reveal differences between amyloid plaques and adjacent control tissue in the transgenic mouse model of AD (APPPS1-21) and in similar regions from non-transgenic littermates. Our microproteomic strategy included isolation of regions of interest by laser capture microdissection and analysis by liquid chromatography mass spectrometry-based label-free relative quantification. We consistently identified 183, 224, and 307 proteins from amyloid plaques, adjacent control and non-tg samples, respectively. Pathway analysis revealed 27 proteins that were significantly regulated when comparing amyloid plaques and corresponding adjacent control regions. We further elucidated that co-localized proteins were subjected to post-translational modifications and are the first to report 193 and 117 unique modifications associated to amyloid plaques and adjacent control extracts, respectively. The three most common modifications detected in proteins from the amyloid plaques were oxidation, deamidation, and pyroglutamylation. Together, our data provide novel information about the biological processes occurring within and around amyloid plaques in the APPPS1-21 mouse model of AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Placa Amiloide/genética , Procesamiento Proteico-Postraduccional/genética , Proteómica , Enfermedad de Alzheimer/patología , Amidas/metabolismo , Péptidos beta-Amiloides/genética , Animales , Encéfalo/patología , Cromatografía Líquida de Alta Presión , Femenino , Glutamatos/metabolismo , Humanos , Espectrometría de Masas , Ratones , Ratones Transgénicos , Oxidación-Reducción , Placa Amiloide/patología
17.
J Neurosci ; 27(34): 9115-29, 2007 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-17715348

RESUMEN

Immunotherapies for various neurodegenerative diseases have recently emerged as a promising approach for clearing pathological protein conformers in these disorders. This type of treatment has not been assessed in models that develop neuronal tau aggregates as observed in frontotemporal dementia and Alzheimer's disease. Here, we present that active immunization with a phosphorylated tau epitope, in P301L tangle model mice, reduces aggregated tau in the brain and slows progression of the tangle-related behavioral phenotype. Females had more tau pathology than males but were also more receptive to the immunotherapy. The tau antibodies generated in these animals recognized pathological tau on brain sections. Performance on behavioral assays that require extensive motor coordination correlated with tau pathology in corresponding brain areas, and antibody levels against the immunogen correlated inversely with tau pathology. Interestingly, age-dependent autoantibodies that recognized recombinant tau protein but not the immunogen were detected in the P301L mice. To confirm that anti-tau antibodies could enter the brain and bind to pathological tau, FITC-tagged antibodies purified from a P301L mouse, with a high antibody titer against the immunogen, were injected into the carotid artery of P301L mice. These antibodies were subsequently detected within the brain and colocalized with PHF1 and MC1 antibodies that recognize pathological tau. Currently, no treatment is available for clearing tau aggregates. Our present findings may lead to a novel therapy targeting one of the major hallmarks of Alzheimer's disease and frontotemporal dementia.


Asunto(s)
Anticuerpos/uso terapéutico , Encéfalo/patología , Demencia/terapia , Inmunoterapia/métodos , Proteínas tau/inmunología , Proteínas tau/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Conducta Animal , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular Tumoral , Demencia/genética , Demencia/patología , Modelos Animales de Enfermedad , Humanos , Leucina/genética , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuroblastoma , Prolina/genética , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Factores Sexuales , Proteínas tau/genética
18.
Brain Res ; 1233: 1-7, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18706894

RESUMEN

The ME7 model of prion disease is a chronic slowly evolving model of neurodegeneration in which cell death is preceded by synaptic dysfunction. Previous studies in cell culture show that accumulation of misfolded prion inhibits the formation of the SNARE complexes involving synaptobrevin, syntaxin and SNAP-25 that play an essential role in neurotransmitter release. Such observations suggest that similar phenomenon may contribute to synaptic dysfunction observed in vivo. We have thus used detergent extraction of hippocampal tissue to investigate the status of SNARE complexes in the ME7 model. In the presence of increasing PrP(Sc) deposition we failed to see a change in the amount of SNARE complexes directly extracted into SDS and resolved by SDS-PAGE. Conversely pre-extraction in Triton X-100, a treatment that promotes SNARE complexes ex vivo, demonstrated a modest reduction in hippocampal SNARE complexes when homogenates were made from tissue at late stage disease. This suggests that accumulated PrP(Sc), or perhaps fibrillar complexes formed of prion only inhibit SNARE complexes that are formed ex vivo following biochemical extraction. Thus the accumulation of PrP(Sc) although deleterious to synaptic function in vivo, does not exert its synaptic effects by disrupting the formation of SNARE complexes that are core to transmitter release.


Asunto(s)
Hipocampo/metabolismo , Degeneración Nerviosa/metabolismo , Proteínas PrPSc/metabolismo , Enfermedades por Prión/metabolismo , Pliegue de Proteína , Proteínas SNARE/metabolismo , Animales , Electroforesis en Gel de Poliacrilamida , Hipocampo/patología , Estudios Longitudinales , Ratones , Ratones Endogámicos C57BL , Octoxinol , Proteínas PrPSc/aislamiento & purificación , Proteínas Qa-SNARE/aislamiento & purificación , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/aislamiento & purificación , Proteínas R-SNARE/metabolismo , Proteínas SNARE/aislamiento & purificación , Transmisión Sináptica/fisiología , Proteína 25 Asociada a Sinaptosomas/aislamiento & purificación , Proteína 25 Asociada a Sinaptosomas/metabolismo , Extractos de Tejidos
19.
Neurosci Lett ; 444(1): 27-30, 2008 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-18706475

RESUMEN

The distribution of metabotropic glutamate (mGlu) receptors within the synapse is an important determinant of function. mGlu have been grouped together into three main sub-classes: Group I mGlu (1 and 5) are predominantly situated on the post-synaptic membrane, whereas Group III (4, 6, 7 and 8) are largely pre-synaptic. Group II mGlu (2 and 3) are distributed peripheral to the active zone, on both sides of the synaptic cleft. Methods based on a distinct pH-dependent extractability of the pre- and post-synaptic marker proteins can provide insight into the molecular organization of synaptic junctions [G.R. Phillips, J.K. Huang, Y. Wang, H. Tanaka, L. Shapiro, W. Zhang, W. Shan, K. Arndt, M. Frank, R.E. Gordon, M.A. Gawinowicz, Y. Zhao and D.R. Colman, The presynaptic particle web: ultrastructure, composition, dissolution and reconstitution, Neuron 32 (2001) 63-77]. We have applied such procedures to rat brain cortical synaptosomes to explore the biochemical evidence for the accepted localisations of metabotropic glutamate receptors. As shown previously a number of post-synaptic marker proteins remained detergent-insoluble at both pH 6 and pH 8. There was an increased extraction of a number of pre-synaptic plasma membrane and cytomatrix proteins consistent with dissolution of the pre-synaptic aspect of synaptic junctions at elevated pH. We similarly observed modest extraction of Group I mGlu at either pH consistent with their post-synaptic organization. However, we observed increased extractability of Group II mGlu at pH 8. The extractability of Group III mGlu was slightly increased at pH 8 but these receptors were largely refractory to extraction. We have also applied the approach to scaffolding proteins implicated in mGlu localisation to define the biochemical correlates of mGlu scaffolding.


Asunto(s)
Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Animales , Fraccionamiento Celular/métodos , Corteza Cerebral/ultraestructura , Femenino , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/ultraestructura , Ratas , Ratas Wistar , Receptores de Glutamato Metabotrópico/clasificación , Sinaptosomas/metabolismo
20.
Front Neurosci ; 12: 254, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29740272

RESUMEN

There are nearly 50 million people with Alzheimer's disease (AD) worldwide and currently no disease modifying treatment is available. AD is characterized by deposits of Amyloid-ß (Aß), neurofibrillary tangles, and neuroinflammation, and several drug discovery programmes studies have focussed on Aß as therapeutic target. Active immunization and passive immunization against Aß leads to the clearance of deposits in humans and transgenic mice expressing human Aß but have failed to improve memory loss. This review will discuss the possible explanations for the lack of efficacy of Aß immunotherapy, including the role of a pro-inflammatory response and subsequent vascular side effects, the binding site of therapeutic antibodies and the timing of the treatment. We further discuss how antibodies can be engineered for improved efficacy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA