Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cureus ; 16(2): e54004, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38476804

ABSTRACT

An infected (mycotic) aneurysm of the visceral arteries is an uncommon entity, which may arise from a secondary infection of a preexisting aneurysm or be due to degeneration from a primary infection. Mycotic aneurysms require prompt recognition and definitive treatment; otherwise, there can be devastating morbidity and mortality. We present the case of a 51-year-old female with HIV and Crohn's disease who presented with subacute abdominal pain, nausea, and vomiting and was found to have an ultimately fatal mycotic aneurysm of the superior mesenteric artery. In addition, we discuss the characteristic imaging features of mycotic aneurysms on computed tomography and magnetic resonance imaging.

2.
JMIR Mhealth Uhealth ; 9(4): e19564, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33861208

ABSTRACT

BACKGROUND: Although fatigue is one of the most debilitating symptoms in patients with multiple sclerosis (MS), its pathogenesis is not well understood. Neurogenic, inflammatory, endocrine, and metabolic mechanisms have been proposed. Taking into account the temporal dynamics and comorbid mood symptoms of fatigue may help differentiate fatigue phenotypes. These phenotypes may reflect different pathogeneses and may respond to different mechanism-specific treatments. Although several tools have been developed to assess various symptoms (including fatigue), monitor clinical status, or improve the perceived level of fatigue in patients with MS, options for a detailed, real-time assessment of MS-related fatigue and relevant comorbidities are still limited. OBJECTIVE: This study aims to present a novel mobile app specifically designed to differentiate fatigue phenotypes using circadian symptom monitoring and state-of-the-art characterization of MS-related fatigue and its related symptoms. We also aim to report the first findings regarding patient compliance and the relationship between compliance and patient characteristics, including MS disease severity. METHODS: After developing the app, we used it in a prospective study designed to investigate the brain magnetic resonance imaging correlates of MS-related fatigue. In total, 64 patients with MS were recruited into this study and asked to use the app over a 2-week period. The app features the following modules: Visual Analogue Scales (VASs) to assess circadian changes in fatigue, depression, anxiety, and pain; daily sleep diaries (SLDs) to assess sleep habits and quality; and 10 one-time questionnaires to assess fatigue, depression, anxiety, sleepiness, physical activity, and motivation, as well as several other one-time questionnaires that were created to assess those relevant aspects of fatigue that were not captured by existing fatigue questionnaires. The app prompts subjects to assess their symptoms multiple times a day and enables real-time symptom monitoring through a web-accessible portal. RESULTS: Of 64 patients, 56 (88%) used the app, of which 51 (91%) completed all one-time questionnaires and 47 (84%) completed all one-time questionnaires, VASs, and SLDs. Patients reported no issues with the usage of the app, and there were no technical issues with our web-based data collection system. The relapsing-remitting MS to secondary-progressive MS ratio was significantly higher in patients who completed all one-time questionnaires, VASs, and SLDs than in those who completed all one-time questionnaires but not all VASs and SLDs (P=.01). No other significant differences in demographics, fatigue, or disease severity were observed between the degrees of compliance. CONCLUSIONS: The app can be used with reasonable compliance across patients with relapsing-remitting and secondary-progressive MS irrespective of demographics, fatigue, or disease severity.


Subject(s)
Mobile Applications , Multiple Sclerosis , Fatigue/diagnosis , Fatigue/epidemiology , Fatigue/etiology , Humans , Multiple Sclerosis/complications , Multiple Sclerosis/diagnosis , Prospective Studies , Surveys and Questionnaires
3.
Neuropharmacology ; 119: 170-181, 2017 06.
Article in English | MEDLINE | ID: mdl-28363773

ABSTRACT

Amylin is an important gut-brain axis hormone. Since amylin and amyloid-ß peptide (Aß) share similar ß sheet secondary structure despite not having the same primary sequences, we hypothesized that the accumulation of Aß in the brains of subjects with Alzheimer's disease (AD) might compete with amylin for binding to the amylin receptor (AmR). If true, adding exogenous amylin type peptides would compete with Aß and reduce the AD pathological cascade, improving cognition. Here we report that a 10-week course of peripheral treatment with human amylin significantly reduced multiple different markers associated with AD pathology, including reducing levels of phospho-tau, insoluble tau, two inflammatory markers (Iba1 and CD68), as well as cerebral Aß. Amylin treatment also led to improvements in learning and memory in two AD mouse models. Mechanistic studies showed that an amylin receptor antagonist successfully antagonized some protective effects of amylin in vivo, suggesting that the protective effects of amylin require interaction with its cognate receptor. Comparison of signaling cascades emanating from AmR suggest that amylin electively suppresses activation of the CDK5 pathway by Aß. Treatment with amylin significantly reduced CDK5 signaling in a receptor dependent manner, dramatically decreasing the levels of p25, the active form of CDK5 with a corresponding reduction in tau phosphorylation. This is the first report documenting the ability of amylin treatment to reduce tauopathy and inflammation in animal models of AD. The data suggest that the clinical analog of amylin, pramlintide, might exhibit utility as a therapeutic agent for AD and other neurodegenerative diseases.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Receptors, Islet Amyloid Polypeptide/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/pharmacology , Ligands , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Mutation/genetics , Peptide Fragments/therapeutic use , Presenilin-1/genetics , Presenilin-1/metabolism , Receptors, Islet Amyloid Polypeptide/antagonists & inhibitors , Spatial Memory/drug effects , tau Proteins/genetics , tau Proteins/metabolism
4.
J Alzheimers Dis ; 56(1): 47-61, 2017.
Article in English | MEDLINE | ID: mdl-27911303

ABSTRACT

Our recent study has demonstrated that peripheral amylin treatment reduces the amyloid pathology in the brain of Alzheimer's disease (AD) mouse models, and improves their learning and memory. We hypothesized that the beneficial effects of amylin for AD was beyond reducing the amyloids in the brain, and have now directly tested the actions of amylin on other aspects of AD pathogenesis, especially neuroinflammation. A 10-week course of peripheral amylin treatment significantly reduced levels of cerebral inflammation markers, Cd68 and Iba1, in amyloid precursor protein (APP) transgenic mice. Mechanistic studies indicated the protective effect of amylin required interaction with its cognate receptor because silencing the amylin receptor expression blocked the amylin effect on Cd68 in microglia. Using weighted gene co-expression network analysis, we discovered that amylin treatment influenced two gene modules linked with amyloid pathology: 1) a module related to proinflammation and transport/vesicle process that included a hub gene of Cd68, and 2) a module related to mitochondria function that included a hub gene of Atp5b. Amylin treatment restored the expression of most genes in the APP cortex toward levels observed in the wild-type (WT) cortex in these two modules including Cd68 and Atp5b. Using a human dataset, we found that the expression levels of Cd68 and Atp5b were significantly correlated with the neurofibrillary tangle burden in the AD brain and with their cognition. These data suggest that amylin acts on the pathological cascade in animal models of AD, and further supports the therapeutic potential of amylin-type peptides for AD.


Subject(s)
Alzheimer Disease/complications , Anti-Inflammatory Agents/therapeutic use , Cerebral Cortex/drug effects , Encephalitis , Gene Expression Regulation/drug effects , Islet Amyloid Polypeptide/therapeutic use , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Calcium-Binding Proteins/metabolism , Cell Line, Transformed , Disease Models, Animal , Encephalitis/drug therapy , Encephalitis/etiology , Encephalitis/pathology , Gene Expression Regulation/genetics , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Mitochondrial Proton-Translocating ATPases/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Receptors, Islet Amyloid Polypeptide/metabolism
5.
J Alzheimers Dis ; 49(1): 129-37, 2016.
Article in English | MEDLINE | ID: mdl-26444783

ABSTRACT

Age is the major risk factor for developing Alzheimer's disease (AD), and modifying age-related factors may help to delay the onset of the disease. The goal of this study was to investigate the relationship between age and the metabolic factors related to the risk of developing AD. The concentrations of insulin, amylin, and amyloid-ß peptide (Aß) in plasma were measured. We further measured the activity of serum Aß degradation by using fluorescein- and biotin-labeled Aß40. Apolipoprotein E4 allele (ApoE4) and cognitive impairment were characterized. Subjects were divided into three age groups: 60-70, 70-80, and ≥80 years old. We found that the older the subjects, the lower the concentration of insulin (p = 0.001) and the higher the concentration of Aß1-40 (p = 0.004) in plasma. However, age was not associated with the concentration of another pancreatic peptide, amylin, and only marginally with Aß1-42. These relationships remained in the absence of diabetes, cardiovascular disease, and stroke, and regardless of the presence of ApoE4 and cognitive impairment. Both age and ApoE4 were inversely associated with, while insulin was positively associated with, the activities of Aß degradation in serum. Our study suggested that low concentration of insulin and high concentration of Aß40 are aging factors related to the risk of AD.


Subject(s)
Aging , Alzheimer Disease/blood , Amyloid beta-Peptides/blood , Apolipoprotein E4/blood , Cognition Disorders/blood , Insulin/blood , Islet Amyloid Polypeptide/blood , Peptide Fragments/blood , Age Distribution , Aged , Aged, 80 and over , Biomarkers/blood , Cross-Sectional Studies , Female , Humans , Linear Models , Male , Middle Aged , Multivariate Analysis
6.
J Alzheimers Dis ; 42(2): 555-63, 2014.
Article in English | MEDLINE | ID: mdl-24898659

ABSTRACT

Our recent study reported that amylin, a pancreatic peptide that readily crosses the blood-brain barrier, improves learning and memory in Alzheimer's disease mouse models. However, the relationship between peripheral amylin and cognition in humans is unknown. In this follow-up study, using a cross-sectional, homebound elderly population, improvement in cognitive function with increasing quartiles of plasma amylin was suggested by positive association with verbal memory (p = 0.0002) and visuoconstruction tasks (p = 0.004), and inverse association with timed measures of attention (p < 0.0001) and executive function (p = 0.04). After adjusting for demographic information, apolipoprotein E4 allele, diabetes, stroke, kidney function, and lipid profile, log10 of plasma amylin remained associated with these cognitive domains. In contrast, plasma amyloid-ß peptide was not associated with these specific cognitive domains. Our study suggests that peripheral amylin may be protective for cognitive decline, especially in the domains affected by Alzheimer's disease.


Subject(s)
Cognition/physiology , Islet Amyloid Polypeptide/blood , Aged , Aged, 80 and over , Amyloidosis, Familial/blood , Apolipoproteins E/genetics , Attention/physiology , Community Health Planning , Corneal Dystrophies, Hereditary/blood , Female , Genotype , Geriatric Assessment , Home Care Services/statistics & numerical data , Humans , Learning/physiology , Male , Mental Status Schedule , Neuropsychological Tests , Verbal Behavior/physiology
7.
PLoS One ; 9(2): e88063, 2014.
Article in English | MEDLINE | ID: mdl-24520345

ABSTRACT

Amylin, a pancreatic peptide that readily crosses the blood brain barrier (BBB), and amyloid-beta peptide (Aß), the main component of amyloid plaques and a major component of Alzheimer's disease (AD) pathology in the brain, share several features. These include having similar ß-sheet secondary structures, binding to the same receptor, and being degraded by the same protease. Thus, amylin may be associated with Aß, but the nature of their relationship remains unclear. In this study, we used human samples to study the relationship between plasma amylin and Aß in the context of the apolipoprotein E alleles (ApoE). We found that concentrations of Aß1-42 (P<0.0001) and Aß1-40 (P<0.0001) increased with each quartile increase of amylin. Using multivariate regression analysis, the study sample showed that plasma amylin was associated with Aß1-42 (ß = +0.149, SE = 0.025, P<0.0001) and Aß1-40 (ß = +0.034, SE = 0.016, P = 0.04) as an outcome after adjusting for age, gender, ethnicity, ApoE4, BMI, diabetes, stroke, kidney function and lipid profile. This positive association between amylin and Aß1-42 in plasma was found regardless of the ApoE genotype. In contrast, the relationship between amylin and Aß1-40 in plasma seen in ApoE4 non-carriers disappeared in the presence of ApoE4. Using AD mouse models, our recent study demonstrates that intraperitoneal (i.p.) injection of synthetic amylin enhances the removal of Aß from the brain into blood, thus resulting in increased blood levels of both amylin and Aß. The positive association between amylin and Aß, especially Aß1-42, in human blood samples is probably relevant to the findings in the AD mouse models. The presence of ApoE4 may attenuate amylin's capacity to remove Aß, especially Aß1-40, from the AD brain.


Subject(s)
Alleles , Amyloid beta-Peptides/blood , Apolipoprotein E4/genetics , Islet Amyloid Polypeptide/blood , Aged , Animals , Biomarkers/blood , Female , Humans , Male , Mice , Multivariate Analysis , Regression Analysis
8.
J Alzheimers Dis ; 37(2): 421-8, 2013.
Article in English | MEDLINE | ID: mdl-23948883

ABSTRACT

Our cross-sectional study showed that the interaction between apolipoprotein E4 (ApoE4) and angiotensin converting enzyme (ACE) inhibitors was associated with Alzheimer's disease (AD). The aim of this longitudinal study was to differentiate whether ACE inhibitors accelerate or reduce the risk of AD in the context of ApoE alleles. Using the longitudinal data from the National Alzheimer's Coordinating Center (NACC) with ApoE genotyping and documentation of ACE inhibitors use, we found that in the absence of ApoE4, subjects who had been taking central ACE inhibitor use (χ2 test: 21% versus 27%, p = 0.0002) or peripheral ACE inhibitor use (χ2 test: 13% versus 27%, p < 0.0001) had lower incidence of AD compared with those who had not been taking an ACE inhibitor. In contrast, in the presence of ApoE4, there was no such association between ACE inhibitor use and the risk of AD. After adjusting for the confounders, central ACE inhibitor use (OR = 0.68, 95% CI = 0.55, 0.83, p = 0.0002) or peripheral ACE inhibitor use (OR = 0.33, 95% CI = 0.33, 0.68, p < 0.0001) still remained inversely associated with a risk of developing AD in ApoE4 non-carriers. In conclusion, ACE inhibitors, especially peripherally acting ones, were associated with a reduced risk of AD in the absence of ApoE4, but had no such effect in those carrying the ApoE4 allele. A double-blind clinical trial should be considered to determine the effect of ACE inhibitors on prevention of AD in the context of ApoE genotype.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Apolipoprotein E4/genetics , Pharmacogenetics , Aged , Aged, 80 and over , Chi-Square Distribution , Databases, Factual/statistics & numerical data , Female , Genotype , Humans , Logistic Models , Longitudinal Studies , Male , Retrospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...