Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
EMBO Rep ; 25(3): 1326-1360, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38347225

RESUMO

ITM2B/BRI2 mutations cause Alzheimer's Disease (AD)-related dementias. We observe heightened ITM2B/BRI2 expression in microglia, a pivotal cell type in AD due to risk-increasing variants in the microglial gene TREM2. Single-cell RNA-sequencing demonstrates a Trem2/Bri2-dependent microglia cluster, underscoring their functional interaction. α-secretase cleaves TREM2 into TREM2-CTF and sTREM2. As BRI2 hinders α-secretase cleavage of the AD-related Aß-Precursor-Protein, we probed whether BRI2 influences TREM2 processing. Our findings indicate a BRI2-TREM2 interaction that inhibits TREM2 processing in heterologous cells. Recombinant BRI2 and TREM2 proteins demonstrate a direct, cell-free BRI2-TREM2 ectodomain interaction. Constitutive and microglial-specific Itm2b-Knock-out mice, and Itm2b-Knock-out primary microglia provide evidence that Bri2 reduces Trem2 processing, boosts Trem2 mRNA expression, and influences Trem2 protein levels through α-secretase-independent pathways, revealing a multifaceted BRI2-TREM2 functional interaction. Moreover, a mutant Itm2b dementia mouse model exhibits elevated Trem2-CTF and sTrem2, mirroring sTREM2 increases in AD patients. Lastly, Bri2 deletion reduces phagocytosis similarly to a pathogenic TREM2 variant that enhances processing. Given BRI2's role in regulating Aß-Precursor-Protein and TREM2 functions, it holds promise as a therapeutic target for AD and related dementias.


Assuntos
Doença de Alzheimer , Demência , Animais , Humanos , Camundongos , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Demência/genética , Modelos Animais de Doenças , Glicoproteínas de Membrana , Camundongos Knockout , Microglia/metabolismo , Receptores Imunológicos
2.
J Biol Chem ; 299(7): 104868, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37257821

RESUMO

About 2% of Alzheimer's disease (AD) cases have early onset (FAD) and are caused by mutations in either Presenilins (PSEN1/2) or amyloid-ß precursor protein (APP). PSEN1/2 catalyze production of Aß peptides of different length from APP. Aß peptides are the major components of amyloid plaques, a pathological lesion that characterizes AD. Analysis of mechanisms by which PSEN1/2 and APP mutations affect Aß peptide compositions lead to the implication of the absolute or relative increase in Aß42 in amyloid-ß plaques formation. Here, to elucidate the formation of pathogenic Aß cocktails leading to amyloid pathology, we utilized FAD rat knock-in models carrying the Swedish APP (Apps allele) and the PSEN1 L435F (Psen1LF allele) mutations. To accommodate the differences in the pathogenicity of rodent and human Aß, these rat models are genetically engineered to express human Aß species as both the Swedish mutant allele and the WT rat allele (called Apph) have been humanized in the Aß-coding region. Analysis of the eight possible FAD mutant permutations indicates that the CNS levels of Aß43, rather than absolute or relative increases in Aß42, determine the onset of pathological amyloid deposition in FAD knock-in rats. Notably, Aß43 was found in amyloid plaques in late onset AD and mild cognitive impairment cases, suggesting that the mechanisms initiating amyloid pathology in FAD knock-in rat reflect disease mechanisms driving amyloid pathology in late onset AD. This study helps clarifying the molecular determinants initiating amyloid pathology and supports therapeutic interventions targeting Aß43 in AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Ratos , Animais , Humanos , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Placa Amiloide/genética , Placa Amiloide/patologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Presenilina-1/genética , Mutação , Secretases da Proteína Precursora do Amiloide/metabolismo
3.
J Biol Chem ; 296: 100054, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33172889

RESUMO

Mutations in integral membrane protein 2B (ITM2b/BRI2) gene cause familial British and Danish dementia (FBD and FDD), autosomal dominant disorders characterized by progressive cognitive deterioration. Two pathogenic mechanisms, which may not be mutually exclusive, have been proposed for FDD and FBD: 1) loss of BRI2 function; 2) accumulation of amyloidogenic mutant BRI2-derived peptides, but the mechanistic details remain unclear. We have previously reported a physiological role of BRI2 in excitatory synaptic transmission at both presynaptic termini and postsynaptic termini. To test whether pathogenic ITM2b mutations affect these physiological BRI2 functions, we analyzed glutamatergic transmission in FDD and FBD knock-in mice, which carry pathogenic FDD and FBD mutations into the mouse endogenous Itm2b gene. We show that in both mutant lines, spontaneous glutamate release and AMPAR-mediated responses are decreased, while short-term synaptic facilitation is increased, effects similar to those observed in Itm2bKO mice. In vivo and in vitro studies show that both pathogenic mutations alter maturation of BRI2 resulting in reduced levels of functional mature BRI2 protein at synapses. Collectively, the data show that FDD and FBD mutations cause a reduction of BRI2 levels and function at synapses, which results in reduced glutamatergic transmission. Notably, other genes mutated in Familial dementia, such as APP, PSEN1/PSEN2, are implicated in glutamatergic synaptic transmission, a function that is altered by pathogenic mutations. Thus, defects in excitatory neurotransmitter release may represent a general and convergent mechanism leading to neurodegeneration. Targeting these dysfunction may offer a unique disease modifying method of therapeutic intervention in neurodegenerative disorders.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Demência/genética , Glutamatos/metabolismo , Mutação , Estabilidade Proteica , Transmissão Sináptica , Animais , Células Cultivadas , Demência/fisiopatologia , Dinamarca , Modelos Animais de Doenças , Feminino , Hipocampo/fisiologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Reino Unido
4.
J Biol Chem ; 297(3): 101089, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34416235

RESUMO

Familial British dementia and familial Danish dementia are neurodegenerative disorders caused by mutations in the gene integral membrane protein 2B (ITM2b) encoding BRI2, which tunes excitatory synaptic transmission at both presynaptic and postsynaptic termini. In addition, BRI2 interacts with and modulates proteolytic processing of amyloid-ß precursor protein (APP), whose mutations cause familial forms of Alzheimer's disease (AD) (familial AD). To study the pathogenic mechanisms triggered by the Danish mutation, we generated rats carrying the Danish mutation in the rat Itm2b gene (Itm2bD rats). Given the BRI2/APP interaction and the widely accepted relevance of human amyloid ß (Aß), a proteolytic product of APP, to AD, Itm2bD rats were engineered to express two humanized App alleles and produce human Aß. Here, we studied young Itm2bD rats to investigate early pathogenic changes in these diseases. We found that periadolescent Itm2bD rats not only present subtle changes in human Aß levels along with decreased spontaneous glutamate release and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor-mediated responses but also had increased short-term synaptic facilitation in the hippocampal Schaeffer-collateral pathway. These alterations in excitatory interneuronal communication can impair learning and memory processes and were akin to those observed in adult mice producing rodent Aß and carrying either the Danish or British mutations in the mouse Itm2b gene. Collectively, the data show that the pathogenic Danish mutation alters the physiological function of BRI2 at glutamatergic synapses across species and early in life. Future studies will determine whether this phenomenon represents an early pathogenic event in human dementia.


Assuntos
Catarata/fisiopatologia , Ataxia Cerebelar/fisiopatologia , Surdez/fisiopatologia , Demência/fisiopatologia , Proteínas de Membrana/genética , Transmissão Sináptica/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Catarata/metabolismo , Ataxia Cerebelar/metabolismo , Surdez/metabolismo , Demência/genética , Demência/metabolismo , Modelos Animais de Doenças , Fármacos Atuantes sobre Aminoácidos Excitatórios/metabolismo , Feminino , Masculino , Proteínas de Membrana/metabolismo , Memória , Terminações Pré-Sinápticas/metabolismo , Ratos , Receptores de Glutamato/metabolismo , Sinapses/metabolismo
5.
J Biol Chem ; 296: 100089, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33434745

RESUMO

Alzheimer's disease (AD) is a neurodegenerative dementia associated with deposition of amyloid plaques and neurofibrillary tangles, formed by amyloid ß (Aß) peptides and phosphor-tau, respectively, in the central nervous system. Approximately 2% of AD cases are due to familial AD (FAD); ∼98% of cases are sporadic AD (SAD). Animal models with FAD are commonly used to study SAD pathogenesis. Because mechanisms leading to FAD and SAD may be distinct, to study SAD pathogenesis, we generated Trem2R47H knock-in rats, which carry the SAD risk factor p.R47H variant of the microglia gene triggering receptor expressed on myeloid cells 2 (TREM2). Trem2R47H rats produce human-Aß from a humanized-App rat allele because human-Aß is more toxic than rodent-Aß and the pathogenic role of the p.R47H TREM2 variant has been linked to human-Aß-clearing deficits. Using periadolescent Trem2R47H rats, we previously demonstrated that supraphysiological tumor necrosis factor-α (TNF-α) boosts glutamatergic transmission, which is excitatory, and suppresses long-term potentiation, a surrogate of learning and memory. Here, we tested the effect of the p.R47H variant on the inhibitory neurotransmitter γ-aminobutyric acid. We report that GABAergic transmission is decreased in Trem2R47H/R47H rats. This decrease is due to acute and reversible action of TNF-α and is not associated with increased human-Aß levels and AD pathology. Thus, the p.R47H variant changes the excitatory/inhibitory balance, favoring excitation. This imbalance could potentiate glutamate excitotoxicity and contribute to neuronal dysfunction, enhanced neuronal death, and neurodegeneration. Future studies will determine whether this imbalance represents an early, Aß-independent pathway leading to dementia and may reveal the AD-modifying therapeutic potential of TNF-α inhibition in the central nervous system.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Neurônios GABAérgicos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Feminino , Masculino , Glicoproteínas de Membrana/metabolismo , Doenças Neurodegenerativas/metabolismo , Ratos , Receptores Imunológicos/metabolismo , Fatores de Risco , Ácido gama-Aminobutírico/metabolismo
6.
J Biol Chem ; 295(21): 7442-7451, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32265300

RESUMO

Familial forms of Alzheimer's disease (FAD) are caused by mutations in the gene encoding amyloid precursor protein, whose processing can result in formation of ß-amyloid (Aß). FAD can also result from mutations in the presenilin 1/2 (PSEN1/2) genes, whose protein products partially compose the γ-secretase complex that cleaves Aß from amyloid precursor protein fragments. Psen1 KO mice and knock-in (KI) mice with homozygous FAD-associated L435F mutations (Psen1LF/LF ) are embryonic and perinatally lethal, precluding a more rigorous examination of the effect of Alzheimer's disease-causing Psen1 mutations on neurodegeneration. Given that the rat is a more suitable model organism with regard to surgical interventions and behavioral testing, we generated a rat KI model of the Psen1LF mutation. In this study, we focused on young Psen1LF rats to determine potential early pathogenic changes caused by this mutation. We found that, unlike Psen1LF/LF mice, Psen1LF/LF rats survive into adulthood despite loss of γ-secretase activity. Consistent with loss of γ-secretase function, Psen1LF/LF rats exhibited low levels of Aß38, Aß40, and Aß42 peptides. In contrast, levels of Aß43, a longer and potentially more amyloidogenic Aß form, were significantly increased in Psen1LF/LF and Psen1LF/w rats. The longer survival of these KI rats affords the opportunity to examine the effect of homozygous Psen1 Alzheimer's disease-associated mutations on neurodegeneration in older animals.


Assuntos
Doença de Alzheimer , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides , Mutação de Sentido Incorreto , Fragmentos de Peptídeos , Presenilina-1 , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Substituição de Aminoácidos , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Técnicas de Introdução de Genes , Homozigoto , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Ratos , Ratos Transgênicos
7.
J Neurosci ; 39(35): 6992-7005, 2019 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-31235642

RESUMO

APP, whose mutations cause familial Alzheimer's disease (FAD), modulates neurotransmission via interaction of its cytoplasmic tail with the synaptic release machinery. Here we identified an intravesicular domain of APP, called intraluminal SV-APP interacting domain (ISVAID), which interacts with glutamatergic, but not GABAergic, synaptic vesicle proteins. ISVAID contains the ß- and α-secretase cleavage sites of APP: proteomic analysis of the interactome of ISVAID suggests that ß- and α-secretase cleavage of APP cuts inside the interaction domain of ISVAID and destabilizes protein-protein interactions. We have tested the functional significance of the ISVAID and of ß-/α-secretase-processing of APP using various ISVAID-derived peptides in competition experiments on both female and male mouse and rats hippocampal slices. A peptide encompassing the entire ISVAID facilitated glutamate, but not GABA, release acting as dominant negative inhibitor of the functions of this APP domain in acute hippocampal slices. In contrast, peptides representing the product of ß-/α-secretase-processing of ISVAID did not alter excitatory neurotransmitter release. These findings suggest that cleavage of APP by either ß- or α-secretase may inactivate the ISVAID, thereby enhancing glutamate release. Our present data support the notion that APP tunes glutamate release, likely through intravesicular and extravesicular interactions with synaptic vesicle proteins and the neurotransmitter release machinery, and that ß-/α cleavage of APP facilitates the release of excitatory neurotransmitter.SIGNIFICANCE STATEMENT Alzheimer's disease has been linked to mutations in APP. However, the biological function of APP is poorly understood. Here we show that an intravesicular APP domain interacts with the proteins that control the release of glutamate, but not GABA. Interfering with the function of this domain promotes glutamate release. This APP domain contains the sites cleaved by ß- and α-secretases: our data suggest that ß-/α cleavage of APP inactivates this functional APP domain promoting excitatory neurotransmitter release.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Glutâmico/metabolismo , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/metabolismo , Ácido gama-Aminobutírico/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Long-Evans
8.
J Neurosci ; 38(11): 2780-2795, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29459374

RESUMO

A homozygous nonsense mutation in the cereblon (CRBN) gene results in autosomal recessive, nonsyndromic intellectual disability that is devoid of other phenotypic features, suggesting a critical role of CRBN in mediating learning and memory. In this study, we demonstrate that adult male Crbn knock-out (CrbnKO) mice exhibit deficits in hippocampal-dependent learning and memory tasks that are recapitulated by focal knock-out of Crbn in the adult dorsal hippocampus, with no changes in social or repetitive behavior. Cellular studies identify deficits in long-term potentiation at Schaffer collateral CA1 synapses. We further show that Crbn is robustly expressed in the mouse hippocampus and CrbnKO mice exhibit hyperphosphorylated levels of AMPKα (Thr172). Examination of processes downstream of AMP-activated protein kinase (AMPK) finds that CrbnKO mice have a selective impairment in mediators of the mTORC1 translation initiation pathway in parallel with lower protein levels of postsynaptic density glutamatergic proteins and higher levels of excitatory presynaptic markers in the hippocampus with no change in markers of the unfolded protein response or autophagy pathways. Acute pharmacological inhibition of AMPK activity in adult CrbnKO mice rescues learning and memory deficits and normalizes hippocampal mTORC1 activity and postsynaptic glutamatergic proteins without altering excitatory presynaptic markers. Thus, this study identifies that loss of Crbn results in learning, memory, and synaptic defects as a consequence of exaggerated AMPK activity, inhibition of mTORC1 signaling, and decreased glutamatergic synaptic proteins. Thus, CrbnKO mice serve as an ideal model of intellectual disability to further explore molecular mechanisms of learning and memory.SIGNIFICANCE STATEMENT Intellectual disability (ID) is one of the most common neurodevelopmental disorders. The cereblon (CRBN) gene has been linked to autosomal recessive, nonsyndromic ID, characterized by an intelligence quotient between 50 and 70 but devoid of other phenotypic features, making cereblon an ideal protein for the study of the fundamental aspects of learning and memory. Here, using the cereblon knock-out mouse model, we show that cereblon deficiency disrupts learning, memory, and synaptic function via AMP-activated protein kinase hyperactivity, downregulation of mTORC1, and dysregulation of excitatory synapses, with no changes in social or repetitive behaviors, consistent with findings in the human population. This establishes the cereblon knock-out mouse as a model of pure ID without the confounding behavioral phenotypes associated with other current models of ID.


Assuntos
Deficiência Intelectual/genética , Deficiência Intelectual/fisiopatologia , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/fisiopatologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Transtornos da Memória/genética , Transtornos da Memória/fisiopatologia , Proteínas do Tecido Nervoso/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Região CA1 Hipocampal/fisiopatologia , Potenciais Pós-Sinápticos Excitadores/genética , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Deficiência Intelectual/tratamento farmacológico , Deficiências da Aprendizagem/tratamento farmacológico , Potenciação de Longa Duração/genética , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/biossíntese , Transtornos da Memória/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Inibidores de Proteínas Quinases/uso terapêutico , Comportamento Social
9.
J Biol Chem ; 291(33): 17209-27, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27325702

RESUMO

The amyloid precursor protein (APP), whose mutations cause Alzheimer disease, plays an important in vivo role and facilitates transmitter release. Because the APP cytosolic region (ACR) is essential for these functions, we have characterized its brain interactome. We found that the ACR interacts with proteins that regulate the ubiquitin-proteasome system, predominantly with the E3 ubiquitin-protein ligases Stub1, which binds the NH2 terminus of the ACR, and CRL4(CRBN), which is formed by Cul4a/b, Ddb1, and Crbn, and interacts with the COOH terminus of the ACR via Crbn. APP shares essential functions with APP-like protein-2 (APLP2) but not APP-like protein-1 (APLP1). Noteworthy, APLP2, but not APLP1, interacts with Stub1 and CRL4(CRBN), pointing to a functional pathway shared only by APP and APLP2. In vitro ubiquitination/ubiquitome analysis indicates that these E3 ligases are enzymatically active and ubiquitinate the ACR residues Lys(649/650/651/676/688) Deletion of Crbn reduces ubiquitination of Lys(676) suggesting that Lys(676) is physiologically ubiquitinated by CRL4(CRBN) The ACR facilitated in vitro ubiquitination of presynaptic proteins that regulate exocytosis, suggesting a mechanism by which APP tunes transmitter release. Other dementia-related proteins, namely Tau and apoE, interact with and are ubiquitinated via the ACR in vitro This, and the evidence that CRBN and CUL4B are linked to intellectual disability, prompts us to hypothesize a pathogenic mechanism, in which APP acts as a modulator of E3 ubiquitin-protein ligase(s), shared by distinct neuronal disorders. The well described accumulation of ubiquitinated protein inclusions in neurodegenerative diseases and the link between the ubiquitin-proteasome system and neurodegeneration make this concept plausible.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Complexos Multienzimáticos/metabolismo , Transmissão Sináptica , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Proteínas Adaptadoras de Transdução de Sinal , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Proteínas Culina/genética , Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Complexos Multienzimáticos/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteínas tau/genética , Proteínas tau/metabolismo
10.
EMBO J ; 30(12): 2501-9, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21587206

RESUMO

An autosomal dominant mutation in the BRI2/ITM2B gene causes familial Danish dementia (FDD). Analysis of FDD(KI) mice, a mouse model of FDD genetically congruous to the human disease since they carry one mutant and one wild-type Bri2/Itm2b allele, has shown that the Danish mutation causes loss of Bri2 protein, synaptic plasticity and memory impairments. BRI2 is a physiological interactor of Aß-precursor protein (APP), a gene associated with Alzheimer disease, which inhibits processing of APP. Here, we show that APP/Bri2 complexes are reduced in synaptic membranes of FDD(KI) mice. Consequently, APP metabolites derived from processing of APP by ß-, α- and γ-secretases are increased in Danish dementia mice. APP haplodeficiency prevents memory and synaptic dysfunctions, consistent with a role for APP metabolites in the pathogenesis of memory and synaptic deficits. This genetic suppression provides compelling evidence that APP and BRI2 functionally interact, and that the neurological effects of the Danish form of BRI2 only occur when sufficient levels of APP are supplied by two alleles. This evidence establishes a pathogenic sameness between familial Danish and Alzheimer's dementias.


Assuntos
Peptídeos beta-Amiloides/genética , Demência/genética , Triagem de Portadores Genéticos , Proteínas de Membrana/genética , Transtornos da Memória/genética , Fragmentos de Peptídeos/genética , Proteínas Adaptadoras de Transdução de Sinal , Doença de Alzheimer/genética , Animais , Dinamarca , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Masculino , Proteínas de Membrana/metabolismo , Transtornos da Memória/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Mutantes Quiméricas/genética , Mutação
11.
Neurobiol Learn Mem ; 126: 18-30, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26528887

RESUMO

Mutations in Amyloid ß Precursor Protein (APP) and in genes that regulate APP processing--such as PSEN1/2 and ITM2b/BRI2--cause familial dementia, such Familial Alzheimer disease (FAD), Familial Danish (FDD) and British (FBD) dementias. The ApoE gene is the major genetic risk factor for sporadic AD. Three major variants of ApoE exist in humans (ApoE2, ApoE3, and ApoE4), with the ApoE4 allele being strongly associated with AD. ITM2b/BRI2 is also a candidate regulatory node genes predicted to mediate the common patterns of gene expression shared by healthy ApoE4 carriers and late-onset AD patients not carrying ApoE4. This evidence provides a direct link between ITM2b/BRI2 and ApoE4. To test whether ApoE4 and pathogenic ITM2b/BRI2 interact to modulate learning and memory, we crossed a mouse carrying the ITM2b/BRI2 mutations that causes FDD knocked-in the endogenous mouse Itm2b/Bri2 gene (FDDKI mice) with human ApoE3 and ApoE4 targeted replacement mice. The resultant ApoE3, FDDKI/ApoE3, ApoE4, FDDKI/ApoE4 male mice were assessed longitudinally for learning and memory at 4, 6, 12, and 16-17 months of age. The results showed that ApoE4-carrying mice displayed spatial working/short-term memory deficits relative to ApoE3-carrying mice starting in early middle age, while long-term spatial memory of ApoE4 mice was not adversely affected even at 16-17 months, and that the FDD mutation impaired working/short-term spatial memory in ApoE3-carrying mice and produced impaired long-term spatial memory in ApoE4-carrying mice in middle age. The present results suggest that the FDD mutation may differentially affect learning and memory in ApoE4 carriers and non-carriers.


Assuntos
Doença de Alzheimer/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Catarata/genética , Ataxia Cerebelar/genética , Surdez/genética , Demência/genética , Aprendizagem/fisiologia , Proteínas de Membrana/genética , Mutação , Proteínas Adaptadoras de Transdução de Sinal , Animais , Catarata/psicologia , Ataxia Cerebelar/psicologia , Surdez/psicologia , Demência/psicologia , Humanos , Masculino , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas , Memória Espacial/fisiologia
12.
J Neurosci ; 32(16): 5481-5, 2012 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-22514310

RESUMO

Familial British Dementia (FBD) is caused by an autosomal dominant mutation in the BRI2/ITM2B gene (Vidal et al., 1999). FBD(KI) mice are a model of FBD that is genetically congruous to the human disease, because they carry one mutant and one wild-type Bri2/Itm2b allele. Analysis of these mice has shown that the British mutation causes memory impairments due to loss of Bri2 function (Tamayev et al., 2010b). BRI2 is a physiologic inhibitor of processing of the Aß-precursor protein (APP; Matsuda et al., 2008), a gene associated with Alzheimer's disease (Bertram et al., 2010). Here we show that APP haploinsufficiency prevents memory dysfunctions seen in FBD(KI) mice. This genetic suppression is consistent with a role for APP in the pathogenesis of memory deficits. Moreover, it provides compelling evidence that the memory dysfunctions caused by the British BRI2 mutant are dependent on endogenous APP and that BRI2 and APP functionally interact. This evidence establishes a mechanistic connection between Familial British and Alzheimer's dementias.


Assuntos
Peptídeos beta-Amiloides/deficiência , Demência/complicações , Glicoproteínas de Membrana/genética , Transtornos da Memória/etiologia , Transtornos da Memória/prevenção & controle , Proteínas Adaptadoras de Transdução de Sinal , Animais , Demência/genética , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Hipocampo/patologia , Humanos , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Transgênicos , Mutação/genética , Príons/metabolismo , Reconhecimento Psicológico/fisiologia , Percepção Espacial/fisiologia , Tubulina (Proteína)/metabolismo
13.
Proc Natl Acad Sci U S A ; 107(48): 20822-7, 2010 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21098268

RESUMO

According to the prevailing "amyloid cascade hypothesis," genetic dementias such as Alzheimer's disease and familial Danish dementia (FDD) are caused by amyloid deposits that trigger tauopathy, neurodegeneration, and behavioral/cognitive alterations. To efficiently reproduce amyloid lesions, murine models of human dementias invariably use transgenic expression systems. However, recent FDD transgenic models showed that Danish amyloidosis does not cause memory defects, suggesting that other mechanisms cause Danish dementia. We studied an animal knock-in model of FDD (FDD(KI/+)) genetically congruous with human cases. FDD(KI/+) mice present reduced Bri2 levels, impaired synaptic plasticity and severe hippocampal memory deficits. These animals show no cerebral lesions that are reputed characteristics of human dementia, such as tangles or amyloid plaques. Bri2(+/-) mice exhibit synaptic and memory deficits similar to FDD(KI/+) mice, and memory loss of FDD(KI/+) mice is prevented by expression of WT BRI2, indicating that Danish dementia is caused by loss of BRI2 function. Together, the data suggest that clinical dementia in Danish patients occurs via a loss of function mechanism and not as a result of amyloidosis and tauopathy.


Assuntos
Amiloide/metabolismo , Transtornos da Memória/complicações , Transtornos da Memória/fisiopatologia , Plasticidade Neuronal/fisiologia , Transdução de Sinais , Sinapses/patologia , Proteínas Adaptadoras de Transdução de Sinal , Amiloidose/complicações , Amiloidose/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Catarata/complicações , Catarata/patologia , Catarata/fisiopatologia , Ataxia Cerebelar/complicações , Ataxia Cerebelar/patologia , Ataxia Cerebelar/fisiopatologia , Surdez/complicações , Surdez/patologia , Surdez/fisiopatologia , Demência/complicações , Demência/patologia , Demência/fisiopatologia , Células HeLa , Humanos , Potenciação de Longa Duração/fisiologia , Glicoproteínas de Membrana , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Sinapses/metabolismo
14.
EMBO Mol Med ; 15(11): e18479, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37750482

RESUMO

Model organisms of human diseases are invaluable tools for unraveling pathogenic mechanisms, identifying potential targets for drug development, and evaluating the therapeutic efficacy of candidates in preclinical trials. The utility of model organisms hinges upon their ability to faithfully replicate the underlying pathogenic mechanisms of the human disease. For rodent models of Alzheimer's disease (AD) and AD-related dementias (ADRD), the limited translatability to human disease raises concerns about their overall utility. What factors contribute to this limitation? Is AD inherently too complex to be accurately modeled in nonhumans? Is the divergence between rodent brains and the human brain so pronounced that rodents are unsuitable as model organisms for AD? Or is it plausible that the commonly used rodent models don't capture the genuine pathogenic mechanisms underlying these diseases? This editorial discusses the challenges associated with transgenic models of AD and ADRD and offers some alternative approaches.


Assuntos
Doença de Alzheimer , Animais , Humanos , Doença de Alzheimer/patologia , Animais Geneticamente Modificados , Encéfalo/patologia
15.
bioRxiv ; 2023 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-37398330

RESUMO

ITM2B/BRI2 mutations cause familial forms of Alzheimer's disease (AD)-related dementias by disrupting BRI2's protein function and leading to the accumulation of amyloidogenic peptides. Although typically studied in neurons, our findings show that BRI2 is highly expressed in microglia, which are crucial in AD pathogenesis due to the association of variants in the microglial gene TREM2 with increased AD risk. Our single-cell RNAseq (scRNAseq) analysis revealed a microglia cluster that depends on a Trem2 activity that is inhibited by Bri2, pointing to a functional interaction between Itm2b/Bri2 and Trem2. Given that the AD-related Amyloid-ß Precursor protein (APP) and TREM2 undergo similar proteolytic processing, and that BRI2 inhibits APP processing, we hypothesized that BRI2 may also regulate TREM2 processing. We found that BRI2 interacts with Trem2 and inhibits its processing by α-secretase in transfected cells. In mice lacking Bri2 expression, we observed increased central nervous system (CNS) levels of Trem2-CTF and sTrem2, which are the products of α-secretase processing of Trem2, indicating increased Trem2 processing by α-secretase in vivo. Reducing Bri2 expression only in microglia resulted in increased sTrem2 levels, suggesting a cell-autonomous effect of Bri2 on α-secretase processing of Trem2. Our study reveals a previously unknow role of BRI2 in regulating TREM2-related neurodegenerative mechanisms. The ability of BRI2 to regulate the processing of both APP and TREM2, combined with its cell-autonomous role in neurons and microglia, makes it a promising candidate for the development of AD and AD-related dementias therapeutics.

16.
Prog Neurobiol ; 227: 102482, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37321444

RESUMO

Several studies including ours reported the detrimental effects of extracellular tau oligomers (ex-oTau) on glutamatergic synaptic transmission and plasticity. Astrocytes greatly internalize ex-oTau whose intracellular accumulation alters neuro/gliotransmitter handling thereby negatively affecting synaptic function. Both amyloid precursor protein (APP) and heparan sulfate proteoglycans (HSPGs) are required for oTau internalization in astrocytes but the molecular mechanisms underlying this phenomenon have not been clearly identified yet. Here we found that a specific antibody anti-glypican 4 (GPC4), a receptor belonging to the HSPG family, significantly reduced oTau uploading from astrocytes and prevented oTau-induced alterations of Ca2+-dependent gliotransmitter release. As such, anti-GPC4 spared neurons co-cultured with astrocytes from the astrocyte-mediated synaptotoxic action of ex-oTau, thus preserving synaptic vesicular release, synaptic protein expression and hippocampal LTP at CA3-CA1 synapses. Of note, the expression of GPC4 depended on APP and, in particular, on its C-terminal domain, AICD, that we found to bind Gpc4 promoter. Accordingly, GPC4 expression was significantly reduced in mice in which either APP was knocked-out or it contained the non-phosphorylatable amino acid alanine replacing threonine 688, thus becoming unable to produce AICD. Collectively, our data indicate that GPC4 expression is APP/AICD-dependent, it mediates oTau accumulation in astrocytes and the resulting synaptotoxic effects.


Assuntos
Precursor de Proteína beta-Amiloide , Glipicanas , Animais , Camundongos , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Astrócitos/metabolismo , Glipicanas/metabolismo , Glipicanas/farmacologia , Neurônios/metabolismo , Transmissão Sináptica/fisiologia
17.
J Neurosci ; 31(33): 11756-61, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21849536

RESUMO

The pathogenic model of Alzheimer's disease (AD) posits that aggregates of amyloid ß, a product of amyloid precursor protein (APP) processing, cause dementia. However, alterations of normal APP functions could contribute to AD pathogenesis, and it is therefore important to understand the role of APP. APP is a member of a gene family that shows functional redundancy as documented by the evidence that single knock-out mice are viable, whereas mice with combined deletions of APP family genes die shortly after birth. A residue in the APP intracellular region, Y(682), is indispensable for these essential functions of APP. It is therefore important to identify pathways that regulate phosphorylation of Y(682) as well as the role of Y(682) in vivo. TrkA is associated with both phosphorylation of APP-Y(682) and alteration of APP processing, suggesting that tyrosine phosphorylation of APP links APP processing and neurotrophic signaling to intracellular pathways associated with cellular differentiation and survival. Here we have tested whether the NGF/TrkA signaling pathway is a physiological regulator of APP phosphorylation. We find that NGF induces tyrosine phosphorylation of APP, and that APP interacts with TrkA and this interaction requires Y(682). Unpredictably, we also uncover that APP, and specifically Y(682), regulates activation of the NGF/TrkA signaling pathway in vivo, the subcellular distribution of TrkA and the sensitivity of neurons to the trophic action of NGF. This evidence suggests that these two membrane protein's functions are strictly interconnected and that the NGF/TrkA signaling pathway is involved in AD pathogenesis and can be used as a therapeutic target.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Fator de Crescimento Neural/metabolismo , Receptor trkA/fisiologia , Transdução de Sinais/fisiologia , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Células Cultivadas , Feminino , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator de Crescimento Neural/fisiologia , Fosforilação/fisiologia , Receptor trkA/metabolismo , Tirosina/fisiologia
18.
J Biol Chem ; 286(11): 8717-21, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21266574

RESUMO

The Aß-precursor protein (APP) intracellular domain is highly conserved and contains many potentially important residues, in particular the (682)YENPTY(687) motif. To dissect the functions of this sequence in vivo, we created an APP knock-in allele mutating Tyr(682) to Gly (Y682G). Crossing this allele to APP-like protein 2 (APLP2) knock-out background showed that mutation of Tyr(682) results in postnatal lethality and neuromuscular synapse defects similar to doubly deficient APP/APLP2 mice. Our results demonstrate that a single residue in the APP intracellular region, Tyr(682), is indispensable for the essential function of APP in developmental regulation.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Junção Neuromuscular/metabolismo , Tirosina/metabolismo , Alelos , Motivos de Aminoácidos , Precursor de Proteína beta-Amiloide/genética , Animais , Técnicas de Introdução de Genes , Camundongos , Camundongos Knockout , Junção Neuromuscular/genética , Estrutura Terciária de Proteína , Tirosina/genética
19.
J Biomed Biotechnol ; 2012: 728178, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22619496

RESUMO

A dominant mutation in the ITM2B/BRI2 gene causes familial Danish dementia (FDD) in humans. To model FDD in animal systems, a knock-in approach was recently implemented in mice expressing a wild-type and mutant allele, which bears the FDD-associated mutation. Since these FDD(KI) mice show behavioural alterations and impaired synaptic function, we characterized their synaptosomal proteome via two-dimensional differential in-gel electrophoresis. After identification by nanoliquid chromatography coupled to electrospray-linear ion trap tandem mass spectrometry, the differentially expressed proteins were classified according to their gene ontology descriptions and their predicted functional interactions. The Dlg4/Psd95 scaffold protein and additional signalling proteins, including protein phosphatases, were revealed by STRING analysis as potential players in the altered synaptic function of FDD(KI) mice. Immunoblotting analysis finally demonstrated the actual downregulation of the synaptosomal scaffold protein Dlg4/Psd95 and of the dual-specificity phosphatase Dusp3 in the synaptosomes of FDD(KI) mice.


Assuntos
Catarata/metabolismo , Ataxia Cerebelar/metabolismo , Surdez/metabolismo , Demência/metabolismo , Modelos Animais de Doenças , Proteoma/análise , Animais , Western Blotting , Eletroforese em Gel Bidimensional , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteômica , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray
20.
PLoS One ; 17(5): e0263546, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35507596

RESUMO

Model organisms mimicking the pathogenesis of human diseases are useful for identifying pathogenic mechanisms and testing therapeutic efficacy of compounds targeting them. Models of Alzheimer's disease (AD) and related dementias (ADRD) aim to reproduce the brain pathology associated with these neurodegenerative disorders. Transgenic models, which involve random insertion of disease-causing genes under the control of artificial promoters, are efficient means of doing so. There are confounding factors associated with transgenic approaches, however, including target gene overexpression, dysregulation of endogenous gene expression at transgenes' integration sites, and limitations in mimicking loss-of-function mechanisms. Furthermore, the choice of species is important, and there are anatomical, physiological, and cognitive reasons for favoring the rat over the mouse, which has been the standard for models of neurodegeneration and dementia. We report an initial assessment of the spatial learning, reversal, and sequencing task capabilities of knock-in (KI) Long-Evans rats with humanizing mutations in the Aß-coding region of App, which encodes amyloid precursor protein (Apph/h rats), using the IntelliCage, an automated operant social home cage system, at 6-8 weeks of age, then again at 4-5 months of age. These rats were previously generated as control organisms for studies on neurodegeneration involving other knock-in rat models from our lab. Apph/h rats of either sex can acquire place learning and reversal tasks. They can also acquire a diagonal sequencing task by 6-8 weeks of age, but not a more advanced serial reversal task involving alternating diagonals, even by 4-5 months of age. Thus, longitudinal behavioral analysis with the IntelliCage system can be useful to determine, in follow-up studies, whether KI rat models of Familial AD (FAD), sporadic late onset AD (LOAD), and of ADRD develop aging-dependent learning and memory deficits.


Assuntos
Doença de Alzheimer , Aplicativos Móveis , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Camundongos Transgênicos , Mutação , Ratos , Ratos Long-Evans , Reversão de Aprendizagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA