Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.141
Filtrar
1.
Appl Neuropsychol Adult ; : 1-15, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39087520

RESUMO

The cognitive impairment known as dementia affects millions of individuals throughout the globe. The use of machine learning (ML) and deep learning (DL) algorithms has shown great promise as a means of early identification and treatment of dementia. Dementias such as Alzheimer's Dementia, frontotemporal dementia, Lewy body dementia, and vascular dementia are all discussed in this article, along with a literature review on using ML algorithms in their diagnosis. Different ML algorithms, such as support vector machines, artificial neural networks, decision trees, and random forests, are compared and contrasted, along with their benefits and drawbacks. As discussed in this article, accurate ML models may be achieved by carefully considering feature selection and data preparation. We also discuss how ML algorithms can predict disease progression and patient responses to therapy. However, overreliance on ML and DL technologies should be avoided without further proof. It's important to note that these technologies are meant to assist in diagnosis but should not be used as the sole criteria for a final diagnosis. The research implies that ML algorithms may help increase the precision with which dementia is diagnosed, especially in its early stages. The efficacy of ML and DL algorithms in clinical contexts must be verified, and ethical issues around the use of personal data must be addressed, but this requires more study.

2.
J Alzheimers Dis Rep ; 8(1): 971-980, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39114555

RESUMO

Background: The Montreal Cognitive Assessment (MoCA) is recommended by the Movement Disorder Society for cognitive testing in movement disorders including Parkinson's disease (PD) and lewy body dementia. Few studies have compared cognitive screening instruments in these diseases, which overlap clinically. Objective: To compare the MoCA and Quick Mild Cognitive Impairment (Qmci) screen in this population. Methods: Patients attending memory and movement disorder clinics associated with a university hospital had the MoCA and Qmci screen performed and diagnostic accuracy compared with the area under the receiver operating characteristic curve (AUC). Duration and severity of movement disorders was assessed using the Unified PD Rating Scale (UPDRS). Results: In total, 133 assessments were available, median age 74±5. Median education was 11±4 years and 65% were male. Median total UPDRS score was 37±26. Median Qmci screen was 51±27, median MoCA was 19±10. There were statistically significant differences in test scores between those with subjective symptoms but normal cognition, mild cognitive impairment (MCI) and dementia (p < 0.001). The Qmci screen had significantly greater accuracy differentiating normal cognition from MCI versus the MoCA (AUC 0.90 versus 0.72, p = 0.01). Both instruments had similar accuracy in identifying cognitive impairment and separating MCI from dementia. The median administration time for the Qmci screen and MoCA were 5.19 and 9.24 minutes (p < 0.001), respectively. Conclusions: Both the MoCA and Qmci screen have good to excellent accuracy in a population with movement disorders experiencing cognitive symptoms. The Qmci screen was significantly more accurate for those with early symptoms and had a shorter administration time.

3.
Front Aging Neurosci ; 16: 1369733, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39104707

RESUMO

Cognitive impairments are a common feature of synucleinopathies such as Parkinson's Disease Dementia and Dementia with Lewy Bodies. These pathologies are characterized by accumulation of Lewy bodies and Lewy neurites as well as neuronal cell death. Alpha-synuclein is the main proteinaceous component of Lewy bodies and Lewy neurites. To model these pathologies in vivo, toxins that selectively target certain neuronal populations or different means of inducing alpha-synuclein aggregation can be used. Alpha-synuclein accumulation can be induced by genetic manipulation, viral vector overexpression or the use of preformed fibrils of alpha-synuclein. In this review, we summarize the cognitive impairments associated with different models of synucleinopathies and relevance to observations in human diseases.

4.
Free Neuropathol ; 52024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39118598

RESUMO

Background: The postmortem diagnostic of individuals having suffered presumptive neurodegenerative disease comprises exclusion of a prion disease, extensive brain sampling and histopathological evaluation, which are resource-intensive and time consuming. To exclude prion disease and to achieve prompt accurate preliminary diagnosis, we developed a fast-track procedure for the histopathological assessment of brains from patients with suspected neurodegenerative disease. Methods: Based on the screening of two brain regions (frontal cortex and cerebellum) with H&E and six immunohistochemical stainings in 133 brain donors, a main histopathological diagnosis was established and compared to the final diagnosis made after a full histopathological work-up according to our brain bank standard procedure. Results: In over 96 % of cases there was a concordance between the fast-track and the final main neuropathological diagnosis. A prion disease was identified in four cases without prior clinical suspicion of a prion infection. Conclusion: The fast-track screening approach relying on two defined, easily accessible brain regions is sufficient to obtain a reliable tentative main diagnosis in individuals with neurodegenerative disease and thus allows for a prompt feedback to the physicians. However, a more thorough histological work-up taking into account the clinical history and the working diagnosis from fast-track screening is necessary for accurate staging and for assessment of co-pathologies.

5.
J Alzheimers Dis ; 2024 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-39121116

RESUMO

Background: Lewy body dementia (LBD) is the second most common neurodegenerative dementia in the US, presenting unique end-of-life challenges. Objective: This study examined healthcare utilization and care continuity in the last year of life in LBD. Methods: Medicare claims for enrollees with LBD, continuously enrolled in the year preceding death, were examined from 2011-2018. We assessed hospital stays, emergency department (ED) visits, intensive care unit (ICU) admissions, life-extending procedures, medications, and care continuity. Results: We identified 45,762 LBD decedents, predominantly female (51.8%), White (85.9%), with average age of 84.1 years (SD 7.5). There was a median of 2 ED visits (IQR 1-5) and 1 inpatient stay (IQR 0-2). Higher age was inversely associated with ICU stays (Odds Ratio [OR] 0.96; 95% Confidence Interval [CI] 0.96-0.97) and life-extending procedures (OR 0.96; 95% CI 0.95-0.96). Black and Hispanic patients experienced higher rates of ED visits, inpatient hospitalizations, ICU admissions, life-extending procedures, and in-hospital deaths relative to White patients. On average, 15 (7.5) medications were prescribed in the last year. Enhanced care continuity correlated with reduced hospital (OR 0.72; 95% CI 0.70-0.74) and ED visits (OR 0.71; 95% CI 0.69-0.87) and fewer life-extending procedures (OR 0.71; 95% CI 0.64-0.79). Conclusions: This study underscored the complex healthcare needs of people with LBD during their final year, which was influenced by age and race. Care continuity may reduce hospital and ED visits and life-extending procedures.

6.
J Parkinsons Dis ; 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39121136

RESUMO

Parkinson's disease (PD) and dementia with Lewy bodies (DLB) share underlying neuropathology. Despite overlapping biology, therapeutic development has been approached separately for these clinical syndromes and there remains no treatment to slow, stop or prevent progression of clinical symptoms and development disability for people living with PD or DLB. Recent advances in biomarker tools, however, have paved new paths for biologic definition and staging of PD and DLB under a shared research framework. Patient-centered research funding organizations see the opportunity for a novel biological staging system for PD and DLB to accelerate and increase success of therapeutic development for the patient communities they serve. Amid growing momentum in the field to develop biological definitions for these neurodegenerative diseases, 7 international nonprofit organizations focused on PD and DLB came together to drive multistakeholder discussion and input on a biological staging system for research. The impact of these convenings to date can be seen in changes incorporated into a proposed biological staging system and growing alignment within the field to rapidly apply new scientific knowledge and biomarker tools to inform clinical trial design. In working together, likeminded nonprofit partners who were initially catalyzed by the significant potential for a biological staging system also realized the power of a shared voice in calling the field to action and have since worked together to establish a coalition to advance precompetitive progress and reduce hurdles to developing better treatments for PD, DLB and biologically related disorders. Plain Language Summary. Disease-focused nonprofit organizations serve to speed new treatments for patients through research funding and advocacy. In the Parkinson's disease (PD) and dementia with Lewy bodies (DLB) fields, several international nonprofit organizations came together to facilitate multistakeholder input on a new biological staging system for research. Stakeholders gathered included researchers, clinicians, drug developers, regulatory agencies, additional nonprofits, and people affected by PD and DLB. This example, fueled by a shared perspective that new drug development tools will improve clinical trials and get better treatments to patients sooner, serves as a model for continued collaborations across the PD and DLB fields. A new, international coalition of nonprofit organizations has emerged to support advancement of treatments to slow, stop, and one day prevent PD, DLB and related disorders, in part, by facilitating future multistakeholder collaborations.

7.
Mol Neurodegener ; 19(1): 60, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39107789

RESUMO

Lewy body dementia (LBD), a class of disorders comprising Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB), features substantial clinical and pathological overlap with Alzheimer's disease (AD). The identification of biomarkers unique to LBD pathophysiology could meaningfully advance its diagnosis, monitoring, and treatment. Using quantitative mass spectrometry (MS), we measured over 9,000 proteins across 138 dorsolateral prefrontal cortex (DLPFC) tissues from a University of Pennsylvania autopsy collection comprising control, Parkinson's disease (PD), PDD, and DLB diagnoses. We then analyzed co-expression network protein alterations in those with LBD, validated these disease signatures in two independent LBD datasets, and compared these findings to those observed in network analyses of AD cases. The LBD network revealed numerous groups or "modules" of co-expressed proteins significantly altered in PDD and DLB, representing synaptic, metabolic, and inflammatory pathophysiology. A comparison of validated LBD signatures to those of AD identified distinct differences between the two diseases. Notably, synuclein-associated presynaptic modules were elevated in LBD but decreased in AD relative to controls. We also found that glial-associated matrisome signatures consistently elevated in AD were more variably altered in LBD, ultimately stratifying those LBD cases with low versus high burdens of concurrent beta-amyloid deposition. In conclusion, unbiased network proteomic analysis revealed diverse pathophysiological changes in the LBD frontal cortex distinct from alterations in AD. These results highlight the LBD brain network proteome as a promising source of biomarkers that could enhance clinical recognition and management.


Assuntos
Doença de Alzheimer , Doença por Corpos de Lewy , Proteômica , Humanos , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Proteômica/métodos , Idoso , Feminino , Masculino , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Córtex Pré-Frontal/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
8.
Am J Med ; 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39151681

RESUMO

This narrative review takes a personal approach in detailing the progression of cognitive decline in a loved one, and the measures taken to care for the patient. The author provides suggestions for a compassionate care plan and advice for clinicians helpful to both patient and caregiver.

9.
Acta Neuropathol ; 148(1): 18, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39141121

RESUMO

Lewy body disorders are heterogeneous neurological conditions defined by intracellular inclusions composed of misshapen α-synuclein protein aggregates. Although α-synuclein aggregates are only one component of inclusions and not strictly coupled to neurodegeneration, evidence suggests they seed the propagation of Lewy pathology within and across cells. Genetic mutations, genomic multiplications, and sequence polymorphisms of the gene encoding α-synuclein are also causally linked to Lewy body disease. In nonfamilial cases of Lewy body disease, the disease trigger remains unidentified but may range from industrial/agricultural toxicants and natural sources of poisons to microbial pathogens. Perhaps due to these peripheral exposures, Lewy inclusions appear at early disease stages in brain regions connected with cranial nerves I and X, which interface with inhaled and ingested environmental elements in the nasal or gastrointestinal cavities. Irrespective of its identity, a stealthy disease trigger most likely shifts soluble α-synuclein (directly or indirectly) into insoluble, cross-ß-sheet aggregates. Indeed, ß-sheet-rich self-replicating α-synuclein multimers reside in patient plasma, cerebrospinal fluid, and other tissues, and can be subjected to α-synuclein seed amplification assays. Thus, clinicians should be able to capitalize on α-synuclein seed amplification assays to stratify patients into potential responders versus non-responders in future clinical trials of α-synuclein targeted therapies. Here, we briefly review the current understanding of α-synuclein in Lewy body disease and speculate on pathophysiological processes underlying the potential transmission of α-synucleinopathy across the neuraxis.


Assuntos
Doença por Corpos de Lewy , alfa-Sinucleína , Humanos , Doença por Corpos de Lewy/patologia , Doença por Corpos de Lewy/metabolismo , alfa-Sinucleína/metabolismo , Animais , Encéfalo/patologia , Encéfalo/metabolismo , Corpos de Lewy/patologia , Corpos de Lewy/metabolismo
10.
Neuron ; 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39079530

RESUMO

The heterogeneity of protein-rich inclusions and its significance in neurodegeneration is poorly understood. Standard patient-derived iPSC models develop inclusions neither reproducibly nor in a reasonable time frame. Here, we developed screenable iPSC "inclusionopathy" models utilizing piggyBac or targeted transgenes to rapidly induce CNS cells that express aggregation-prone proteins at brain-like levels. Inclusions and their effects on cell survival were trackable at single-inclusion resolution. Exemplar cortical neuron α-synuclein inclusionopathy models were engineered through transgenic expression of α-synuclein mutant forms or exogenous seeding with fibrils. We identified multiple inclusion classes, including neuroprotective p62-positive inclusions versus dynamic and neurotoxic lipid-rich inclusions, both identified in patient brains. Fusion events between these inclusion subtypes altered neuronal survival. Proteome-scale α-synuclein genetic- and physical-interaction screens pinpointed candidate RNA-processing and actin-cytoskeleton-modulator proteins like RhoA whose sequestration into inclusions could enhance toxicity. These tractable CNS models should prove useful in functional genomic analysis and drug development for proteinopathies.

11.
J Alzheimers Dis ; 100(3): 761-769, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38968048

RESUMO

This manuscript reviews the significant skin manifestations of Lewy body disease, including Parkinson's disease and dementia with Lewy bodies, and the diagnostic utility of skin biopsy. Besides classic motor and cognitive symptoms, non-motor manifestations, particularly dermatologic disorders, can play a crucial role in disease presentation and diagnosis. This review explores the intricate relationship between the skin and Lewy body disease. Seborrheic dermatitis, autoimmune blistering diseases (bullous pemphigoid and pemphigus), rosacea, and melanoma are scrutinized for their unique associations with Parkinson's disease, revealing potential links through shared pathophysiological mechanisms. Advances in diagnostic techniques allow the identification of promising biomarkers such as α-synuclein in samples obtained by skin punch biopsy. Understanding the dermatologic aspects of Lewy body disease not only contributes to its holistic characterization but also holds implications for innovative diagnostic approaches.


Assuntos
Doença por Corpos de Lewy , Dermatopatias , Pele , Humanos , Doença por Corpos de Lewy/patologia , Doença por Corpos de Lewy/diagnóstico , Dermatopatias/patologia , Dermatopatias/diagnóstico , Pele/patologia , Doença de Parkinson/patologia , Doença de Parkinson/diagnóstico
12.
Geriatr Psychol Neuropsychiatr Vieil ; 22(2): 241-253, 2024 Jun 01.
Artigo em Francês | MEDLINE | ID: mdl-39023159

RESUMO

Lewy body dementia (LBD) is the second most frequent neurodegenerative disorder after Alzheimer disease (AD). In this study, we compared functional decline between LBD and AD patients, considering motor dysfunction, over an 18-month follow-up period. We included all patients >70 years of age, with initial MMSE ≥ 20 and a diagnosis of possible or probable LBD or AD, who consulted at the memory centre of the Pitié-Salpêtrière hospital. Statistical analyses were performed using univariate tests and multivariate linear regression. Thirty-seven AD and 36 LBD patients were included, with a median age of 81 and a median MMSE score of 24/30. Global ADL Katz score decreased significantly for LBD people, compared to AD patients: -0.40 ± 0.75 versus 0 ± 0.24; p=0.003. Global IADL score decreased in the two populations but without a significant difference between the two groups: -1.71 ± 2.19 in LBD versus -1.32 (± 1.55); p=0.38. This study shows a significant decrease in autonomy in LBD patients over time that was faster than that in AD patients, related, in particular, to bathing, dressing and personal care.


Assuntos
Atividades Cotidianas , Doença de Alzheimer , Doença por Corpos de Lewy , Humanos , Doença por Corpos de Lewy/psicologia , Doença por Corpos de Lewy/fisiopatologia , Doença de Alzheimer/psicologia , Masculino , Feminino , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença
13.
Open Biol ; 14(7): 230419, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39013416

RESUMO

The mechanisms responsible for neuronal death causing cognitive loss in Alzheimer's disease (AD) and many other dementias are not known. Serum amyloid P component (SAP) is a constitutive plasma protein, which is cytotoxic for cerebral neurones and also promotes formation and persistence of cerebral Aß amyloid and neurofibrillary tangles. Circulating SAP, which is produced exclusively by the liver, is normally almost completely excluded from the brain. Conditions increasing brain exposure to SAP increase dementia risk, consistent with a causative role in neurodegeneration. Furthermore, neocortex content of SAP is strongly and independently associated with dementia at death. Here, seeking genomic evidence for a causal link of SAP with neurodegeneration, we meta-analysed three genome-wide association studies of 44 288 participants, then conducted cis-Mendelian randomization assessment of associations with neurodegenerative diseases. Higher genetically instrumented plasma SAP concentrations were associated with AD (odds ratio 1.07, 95% confidence interval (CI) 1.02; 1.11, p = 1.8 × 10-3), Lewy body dementia (odds ratio 1.37, 95%CI 1.19; 1.59, p = 1.5 × 10-5) and plasma tau concentration (0.06 log2(ng l-1) 95%CI 0.03; 0.08, p = 4.55 × 10-6). These genetic findings are consistent with neuropathogenicity of SAP. Depletion of SAP from the blood and the brain, by the safe, well tolerated, experimental drug miridesap may thus be neuroprotective.


Assuntos
Estudo de Associação Genômica Ampla , Doenças Neurodegenerativas , Componente Amiloide P Sérico , Humanos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/etiologia , Polimorfismo de Nucleotídeo Único , Predisposição Genética para Doença , Análise da Randomização Mendeliana , Biomarcadores , Proteínas tau/metabolismo , Proteínas tau/genética , Doença por Corpos de Lewy/genética , Doença por Corpos de Lewy/metabolismo , Masculino , Feminino
14.
Rinsho Shinkeigaku ; 2024 Jul 27.
Artigo em Japonês | MEDLINE | ID: mdl-39069492

RESUMO

The patient was an 85-year-old man with a one-year history of difficulty reading kana. Neuropsychological evaluation revealed kana (phonogram)-selective reading impairment and kanji (ideogram)-dominant writing impairment. MRI revealed significant cerebral atrophy in the left occipital cortex, leading to the clinical diagnosis of posterior cortical atrophy (PCA). Cerebrospinal fluid amyloid ß1-42 levels were reduced, and amyloid PET showed accumulation in the posterior cingulate cortex, precuneus, and frontal lobe. In contrast, tau PET showed no accumulation in the atrophied brain areas. Episodes of REM sleep behavior disorder and decreased uptake on meta-iodobenzylguanidine (MIBG) myocardial scintigraphy suggested the involvement of Lewy body pathology. PCA with distinct laterality has been rarely reported, and |this is the first case to present Kana-selective reading impairment and Kanji-dominant writing impairment with neurodegenerative background.

15.
Brain Commun ; 6(4): fcae190, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38978726

RESUMO

Up to 80% of Parkinson's disease patients develop dementia, but time to dementia varies widely from motor symptom onset. Dementia with Lewy bodies presents with clinical features similar to Parkinson's disease dementia, but cognitive impairment precedes or coincides with motor onset. It remains controversial whether dementia with Lewy bodies and Parkinson's disease dementia are distinct conditions or represent part of a disease spectrum. The biological mechanisms underlying disease heterogeneity, in particular the development of dementia, remain poorly understood, but will likely be the key to understanding disease pathways and, ultimately, therapy development. Previous genome-wide association studies in Parkinson's disease and dementia with Lewy bodies/Parkinson's disease dementia have identified risk loci differentiating patients from controls. We collated data for 7804 patients of European ancestry from Tracking Parkinson's, The Oxford Discovery Cohort, and Accelerating Medicine Partnership-Parkinson's Disease Initiative. We conducted a discrete phenotype genome-wide association study comparing Lewy body diseases with and without dementia to decode disease heterogeneity by investigating the genetic drivers of dementia in Lewy body diseases. We found that risk allele rs429358 tagging APOEe4 increases the odds of developing dementia, and that rs7668531 near the MMRN1 and SNCA-AS1 genes and an intronic variant rs17442721 tagging LRRK2 G2019S on chromosome 12 are protective against dementia. These results should be validated in autopsy-confirmed cases in future studies.

16.
Neuropathol Appl Neurobiol ; 50(4): e12999, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39036837

RESUMO

AIMS: Endoplasmic reticulum stress followed by the unfolded protein response is one of the cellular mechanisms contributing to the progression of α-synuclein pathology in Parkinson's disease and other Lewy body diseases. We aimed to investigate the activation of endoplasmic reticulum stress and its correlation with α-synuclein pathology in human post-mortem brain tissue. METHODS: We analysed brain tissue from 45 subjects-14 symptomatic patients with Lewy body disease, 19 subjects with incidental Lewy body disease, and 12 healthy controls. The analysed brain regions included the medulla, pons, midbrain, striatum, amygdala and entorhinal, temporal, frontal and occipital cortex. We analysed activation of endoplasmic reticulum stress via levels of the unfolded protein response-related proteins (Grp78, eIF2α) and endoplasmic reticulum stress-regulating neurotrophic factors (MANF, CDNF). RESULTS: We showed that regional levels of two endoplasmic reticulum-localised neurotrophic factors, MANF and CDNF, did not change in response to accumulating α-synuclein pathology. The concentration of MANF negatively correlated with age in specific regions. eIF2α was upregulated in the striatum of Lewy body disease patients and correlated with increased α-synuclein levels. We found the upregulation of chaperone Grp78 in the amygdala and nigral dopaminergic neurons of Lewy body disease patients. Grp78 levels in the amygdala strongly correlated with soluble α-synuclein levels. CONCLUSIONS: Our data suggest a strong but regionally specific change in Grp78 and eIF2α levels, which positively correlates with soluble α-synuclein levels. Additionally, MANF levels decreased in dopaminergic neurons in the substantia nigra. Our research suggests that endoplasmic reticulum stress activation is not associated with Lewy pathology but rather with soluble α-synuclein concentration and disease progression.


Assuntos
Chaperona BiP do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos , Proteínas de Choque Térmico , Doença por Corpos de Lewy , Resposta a Proteínas não Dobradas , Regulação para Cima , alfa-Sinucleína , Humanos , Doença por Corpos de Lewy/patologia , Doença por Corpos de Lewy/metabolismo , Chaperona BiP do Retículo Endoplasmático/metabolismo , alfa-Sinucleína/metabolismo , Masculino , Idoso , Feminino , Resposta a Proteínas não Dobradas/fisiologia , Idoso de 80 Anos ou mais , Proteínas de Choque Térmico/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Pessoa de Meia-Idade , Fatores de Crescimento Neural/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Biomarcadores/metabolismo
17.
J Clin Exp Neuropsychol ; : 1-11, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38949538

RESUMO

OBJECTIVE: Prior research on the Noise Pareidolia Test (NPT) has demonstrated its clinical utility in detecting patients with mild cognitive impairment and dementia due to Lewy Body Disease (LBD). However, few studies to date have investigated the neuropsychological factors underlying pareidolia errors on the NPT across the clinical spectrum of LBD. Furthermore, to our knowledge, no research has examined the relationship between cortical thickness using MRI data and NPT subscores. As such, this study sought to explore the neuropsychological and neuroanatomical factors influencing performance on the NPT utilizing the National Alzheimer's Coordinating Center Lewy Body Dementia Module. METHODS: Our sample included participants with normal cognition (NC; n = 56), LBD with mild cognitive impairment (LBD-MCI; n = 97), and LBD with dementia (LBD-Dementia; n = 94). Archival data from NACC were retrospectively analyzed for group differences in neuropsychological test scores and cognitive and psychiatric predictors of NPT scores. Clinicoradiological correlates between NPT subscores and a small subsample of the above LBD participants were also examined. RESULTS: Analyses revealed significant differences in NPT scores among groups. Regression analysis demonstrated that dementia severity, attention, and visuospatial processing contributed approximately 24% of NPT performance in LBD groups. Clinicoradiological analysis suggests a potential contribution of the right fusiform gyrus, but not the inferior occipital gyrus, to NPT pareidolia error scores. CONCLUSIONS: Our findings highlight the interplay of attention and visuoperceptual functions in complex pareidolia in LBD. Further investigation is needed to refine the utility of NPT scores in clinical settings, including identifying patients at risk for visual illusions and hallucinations.

18.
Brain ; 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38991041

RESUMO

Concomitant Alzheimer's disease (AD) pathology is a frequent event in the context of Lewy body disease (LBD), occurring in approximately half of all cases. Evidence shows that LBD patients with AD copathology show an accelerated disease course, a greater risk of cognitive decline and an overall poorer prognosis. However, LBD-AD cases may show heterogeneous motor and non-motor phenotypes with higher risk of dementia, and, consequently, be not rarely misdiagnosed. In this review, we summarize the state-of-the-art on LBD-AD by discussing the synergistic effects between AD neuropathological changes and Lewy pathology and their clinical relevance. Furthermore, we provide an extensive overview of neuroimaging and fluid biomarkers under assessment in LBD-AD and their possible diagnostic and prognostic value. AD pathology can be suspected in vivo by means of CSF, MRI and PET markers, whereas α-synuclein seed amplification assays (SAAs) represent to date the most promising technique to identify Lewy pathology in different biological tissues. Pathological imaging and CSF AD biomarkers are associated with a higher likelihood of cognitive decline in LBD but do not always mirror the neuropathological severity like in pure AD. The implementation of blood-based biomarkers of AD might allow the fast screening of LBD patients for AD copathology, thus improving the overall diagnostic sensitivity for LBD-AD. Finally, we discuss the literature on novel candidate biomarkers being exploited in LBD-AD to investigate other aspects of neurodegeneration, such as neuroaxonal injury, glial activation and synaptic dysfunction. The thorough characterization of AD copathology in LBD should be taken into account for the differential diagnosis of dementia syndromes, for the prognostic evaluation on an individual level and for guiding symptomatic and disease-modifying therapies.

19.
Alzheimers Dement ; 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38955137

RESUMO

INTRODUCTION: The recent introduction of seed amplification assays (SAAs) detecting misfolded α-synuclein, a pathology-specific marker for Lewy body disease (LBD), has allowed the in vivo identification and phenotypic characterization of patients with co-occurring Alzheimer's disease (AD) and LBD since the early clinical or even preclinical stage. METHODS: We reviewed studies with an in vivo biomarker-based diagnosis of AD-LBD copathology. RESULTS: Studies in large cohorts of cognitively impaired individuals have shown that cerebrospinal fluid (CSF) biomarkers detect the coexistence of AD and LB pathology in approximately 20%-25% of them, independently of the primary clinical diagnosis. Compared to those with pure AD, AD-LBD patients showed worse global cognition, especially in attentive/executive and visuospatial functions, and worse motor functions. In cognitively unimpaired individuals, concurrent AD-LBD pathologies predicted longitudinal cognitive progression with faster worsening of global cognition, memory, and attentive/executive functions. DISCUSSION: Future research studies aiming for a better precision medicine approach should develop SAAs further to reach a quantitative evaluation or staging of each underlying pathology using a single biofluid sample. HIGHLIGHTS: α-Synuclein seed amplification assays (SAAs) provide a specific marker for Lewy body disease (LBD). SAAs allow for the in vivo identification of co-occurring LBD in patients with Alzheimer's disease (AD). AD-LBD coexist in 20-25% of cognitively impaired elderly individuals, and ∼8% of those asymptomatic. Compared to pure AD, AD-LBD causes a faster worsening of cognitive functions. AD-LBD is associated with worse attentive/executive, memory, visuospatial and motor functions.

20.
J Alzheimers Dis ; 100(2): 579-601, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38875040

RESUMO

Background: Alzheimer's disease (AD) and Lewy body disease (LBD) are characterized by early and gradual worsening perturbations in speeded cognitive responses. Objective: Using simple and choice reaction time tasks, we compared two indicators of cognitive speed within and across the AD and LBD spectra: mean rate (average reaction time across trials) and inconsistency (within person variability). Methods: The AD spectrum cohorts included subjective cognitive impairment (SCI, n = 28), mild cognitive impairment (MCI, n = 121), and AD (n = 45) participants. The LBD spectrum included Parkinson's disease (PD, n = 32), mild cognitive impairment in PD (PD-MCI, n = 21), and LBD (n = 18) participants. A cognitively unimpaired (CU, n = 39) cohort served as common benchmark. We conducted multivariate analyses of variance and discrimination analyses. Results: Within the AD spectrum, the AD cohort was slower and more inconsistent than the CU, SCI, and MCI cohorts. The MCI cohort was slower than the CU cohort. Within the LBD spectrum, the LBD cohort was slower and more inconsistent than the CU, PD, and PD-MCI cohorts. The PD-MCI cohort was slower than the CU and PD cohorts. In cross-spectra (corresponding cohort) comparisons, the LBD cohort was slower and more inconsistent than the AD cohort. The PD-MCI cohort was slower than the MCI cohort. Discrimination analyses clarified the group difference patterns. Conclusions: For both speed tasks, mean rate and inconsistency demonstrated similar sensitivity to spectra-related comparisons. Both dementia cohorts were slower and more inconsistent than each of their respective non-dementia cohorts.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doença por Corpos de Lewy , Testes Neuropsicológicos , Tempo de Reação , Humanos , Feminino , Masculino , Idoso , Doença por Corpos de Lewy/psicologia , Doença de Alzheimer/psicologia , Disfunção Cognitiva/psicologia , Tempo de Reação/fisiologia , Testes Neuropsicológicos/estatística & dados numéricos , Estudos de Coortes , Doença de Parkinson/psicologia , Idoso de 80 Anos ou mais , Cognição/fisiologia , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA