Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Acta Neuropathol ; 144(5): 911-938, 2022 11.
Article in English | MEDLINE | ID: mdl-36104602

ABSTRACT

The mechanistic relationship between amyloid-beta precursor protein (APP) processing and mitochondrial dysfunction in Alzheimer's disease (AD) has long eluded the field. Here, we report that coiled-coil-helix-coiled-coil-helix domain containing 6 (CHCHD6), a core protein of the mammalian mitochondrial contact site and cristae organizing system, mechanistically connects these AD features through a circular feedback loop that lowers CHCHD6 and raises APP processing. In cellular and animal AD models and human AD brains, the APP intracellular domain fragment inhibits CHCHD6 transcription by binding its promoter. CHCHD6 and APP bind and stabilize one another. Reduced CHCHD6 enhances APP accumulation on mitochondria-associated ER membranes and accelerates APP processing, and induces mitochondrial dysfunction and neuronal cholesterol accumulation, promoting amyloid pathology. Compensation for CHCHD6 loss in an AD mouse model reduces AD-associated neuropathology and cognitive impairment. Thus, CHCHD6 connects APP processing and mitochondrial dysfunction in AD. This provides a potential new therapeutic target for patients.


Subject(s)
Alzheimer Disease , Amyloidosis , Alzheimer Disease/pathology , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/metabolism , Animals , Disease Models, Animal , Humans , Mammals/metabolism , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins
2.
J Biomed Inform ; 133: 104164, 2022 09.
Article in English | MEDLINE | ID: mdl-35985621

ABSTRACT

Combination pharmacotherapy targets key disease pathways in a synergistic or additive manner and has high potential in treating complex diseases. Computational methods have been developed to identifying combination pharmacotherapy by analyzing large amounts of biomedical data. Existing computational approaches are often underpowered due to their reliance on our limited understanding of disease mechanisms. On the other hand, observable phenotypic inter-relationships among thousands of diseases often reflect their underlying shared genetic and molecular underpinnings, therefore can offer unique opportunities to design computational models to discover novel combinational therapies by automatically transferring knowledge among phenotypically related diseases. We developed a novel phenome-driven drug discovery system, named TuSDC, which leverages knowledge of existing drug combinations, disease comorbidities, and disease treatments of thousands of disease and drug entities extracted from over 31.5 million biomedical research articles using natural language processing techniques. TuSDC predicts combination pharmacotherapy by extracting representations of diseases and drugs using tensor factorization approaches. In external validation, TuSDC achieved an average precision of 0.77 for top ranked candidates, outperforming a state of art mechanism-based method for discovering drug combinations in treating hypertension. We evaluated top ranked anti-hypertension drug combinations using electronic health records of 84.7 million unique patients and showed that a novel drug combination hydrochlorothiazide-digoxin was associated with significantly lower hazards of subsequent hypertension as compared to the monotherapy hydrochlorothiazide alone (HR: 0.769, 95% CI [0.732, 0.807]) and digoxin alone (0.857, 95% CI [0.785, 0.936]). Data-driven informatics analyses reveal that the renin-angiotensin system is involved in the synergistical interactions of hydrochlorothiazide and digoxin on regulating hypertension. The prediction model's code with PyTorch version 1.5 is available at http://nlp.case.edu/public/data/TuSDC/.


Subject(s)
Hypertension , Natural Language Processing , Digoxin , Drug Combinations , Electronic Health Records , Humans , Hydrochlorothiazide , Hypertension/drug therapy , Machine Learning , Phenotype
3.
Nat Commun ; 13(1): 1121, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35236834

ABSTRACT

Predisposition to Alzheimer's disease (AD) may arise from lipid metabolism perturbation, however, the underlying mechanism remains elusive. Here, we identify ATPase family AAA-domain containing protein 3A (ATAD3A), a mitochondrial AAA-ATPase, as a molecular switch that links cholesterol metabolism impairment to AD phenotypes. In neuronal models of AD, the 5XFAD mouse model and post-mortem AD brains, ATAD3A is oligomerized and accumulated at the mitochondria-associated ER membranes (MAMs), where it induces cholesterol accumulation by inhibiting gene expression of CYP46A1, an enzyme governing brain cholesterol clearance. ATAD3A and CYP46A1 cooperate to promote APP processing and synaptic loss. Suppressing ATAD3A oligomerization by heterozygous ATAD3A knockout or pharmacological inhibition with DA1 restores neuronal CYP46A1 levels, normalizes brain cholesterol turnover and MAM integrity, suppresses APP processing and synaptic loss, and consequently reduces AD neuropathology and cognitive deficits in AD transgenic mice. These findings reveal a role for ATAD3A oligomerization in AD pathogenesis and suggest ATAD3A as a potential therapeutic target for AD.


Subject(s)
ATPases Associated with Diverse Cellular Activities , Alzheimer Disease , Mitochondrial Proteins , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Alzheimer Disease/metabolism , Animals , Cognition , Disease Models, Animal , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
5.
World Psychiatry ; 21(1): 124-132, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34612005

ABSTRACT

Individuals with substance use disorders (SUDs) are at increased risk for COVID-19 infection and for adverse outcomes of the infection. Though vaccines are highly effective against COVID-19, their effectiveness in individuals with SUDs might be curtailed by compromised immune status and a greater likelihood of exposures, added to the waning vaccine immunity and the new SARS-CoV-2 variants. In a population-based cohort study, we assessed the risk, time trends, outcomes and disparities of COVID-19 breakthrough infection in fully vaccinated SUD patients starting 14 days after completion of vaccination. The study included 579,372 individuals (30,183 with a diagnosis of SUD and 549,189 without such a diagnosis) who were fully vaccinated between December 2020 and August 2021, and had not contracted COVID-19 infection prior to vaccination. We used the TriNetX Analytics network platform to access de-identified electronic health records from 63 health care organizations in the US. Among SUD patients, the risk for breakthrough infection ranged from 6.8% for tobacco use disorder to 7.8% for cannabis use disorder, all significantly higher than the 3.6% in non-SUD population (p<0.001). Breakthrough infection risk remained significantly higher after controlling for demographics (age, gender, ethnicity) and vaccine types for all SUD subtypes, except for tobacco use disorder, and was highest for cocaine and cannabis use disorders (hazard ratio, HR=2.06, 95% CI: 1.30-3.25 for cocaine; HR=1.92, 95% CI: 1.39-2.66 for cannabis). When we matched SUD and non-SUD individuals for lifetime comorbidities and adverse socioeconomic determinants of health, the risk for breakthrough infection no longer differed between these populations, except for patients with cannabis use disorder, who remained at increased risk (HR=1.55, 95% CI: 1.22-1.99). The risk for breakthrough infection was higher in SUD patients who received the Pfizer than the Moderna vaccine (HR=1.49, 95% CI: 1.31-1.69). In the vaccinated SUD population, the risk for hospitalization was 22.5% for the breakthrough cohort and 1.6% for the non-breakthrough cohort (risk ratio, RR=14.4, 95% CI: 10.19-20.42), while the risk for death was 1.7% and 0.5% respectively (RR=3.5, 95% CI: 1.74-7.05). No significant age, gender and ethnic disparities for breakthrough infection were observed in vaccinated SUD patients. These data suggest that fully vaccinated SUD individuals are at higher risk for breakthrough COVID-19 infection, and this is largely due to their higher prevalence of comorbidities and adverse socioeconomic determinants of health compared with non-SUD individuals. The high frequency of comorbidities in SUD patients is also likely to contribute to their high rates of hospitalization and death following breakthrough infection.

6.
Alzheimers Res Ther ; 13(1): 177, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34670619

ABSTRACT

BACKGROUND: Interactions between the gut microbiota, microglia, and aging may modulate Alzheimer's disease (AD) pathogenesis but the precise nature of such interactions is not known. METHODS: We developed an integrated multi-dimensional, knowledge-driven, systems approach to identify interactions among microbial metabolites, microglia, and AD. Publicly available datasets were repurposed to create a multi-dimensional knowledge-driven pipeline consisting of an integrated network of microbial metabolite-gene-pathway-phenotype (MGPPN) consisting of 34,509 nodes (216 microbial metabolites, 22,982 genes, 1329 pathways, 9982 mouse phenotypes) and 1,032,942 edges. RESULTS: We evaluated the network-based ranking algorithm by showing that abnormal microglia function and physiology are significantly associated with AD pathology at both genetic and phenotypic levels: AD risk genes were ranked at the top 6.4% among 22,982 genes, P < 0.001. AD phenotypes were ranked at the top 11.5% among 9982 phenotypes, P < 0.001. A total of 8094 microglia-microbial metabolite-gene-pathway-phenotype-AD interactions were identified for top-ranked AD-associated microbial metabolites. Short-chain fatty acids (SCFAs) were ranked at the top among prioritized AD-associated microbial metabolites. Through data-driven analyses, we provided evidence that SCFAs are involved in microglia-mediated gut-microbiota-brain interactions in AD at both genetic, functional, and phenotypic levels. CONCLUSION: Our analysis produces a novel framework to offer insights into the mechanistic links between gut microbial metabolites, microglia, and AD, with the overall goal to facilitate disease mechanism understanding, therapeutic target identification, and designing confirmatory experimental studies.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Alzheimer Disease/genetics , Animals , Brain , Mice , Microglia , Phenotype
8.
EClinicalMedicine ; 31: 100688, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33521611

ABSTRACT

BACKGROUND: Scientific evidence is lacking regarding the risk of patients with chronic liver disease (CLD) for COVID-19, and how these risks are affected by age, gender and race. METHODS: We performed a case-control study of electronic health records of 62.2 million patients (age >18 years) in the US up to October 1st, 2020, including 1,034,270 patients with CLD, 16,530 with COVID-19, and 820 with both COVID-19 and CLD. We assessed the risk, disparities, and outcomes of COVID-19 in patients with six major CLDs. FINDINGS: Patients with a recent medical encounter for CLD were at significantly increased risk for COVID-19 compared with patients without CLD, with the strongest effect in patients with chronic non-alcoholic liver disease [adjusted odd ratio (AOR)=13.11, 95% CI: 12.49-13.76, p < 0.001] and non-alcoholic cirrhosis (AOR=11.53, 95% CI: 10.69-12.43, p < 0.001), followed by chronic hepatitis C (AOR=8.93, 95% CI:8.25-9.66, p < 0.001), alcoholic liver damage (AOR=7.05, 95% CI:6.30-7.88, p < 0.001), alcoholic liver cirrhosis (AOR=7.00, 95% CI:6.15-7.97, p < 0.001) and chronic hepatitis B (AOR=4.37, 95% CI:3.35-5.69, p < 0.001). African Americans with CLD were twice more likely to develop COVID-19 than Caucasians. Patients with COVID-19 and a recent encounter for CLD had a death rate of 10.3% (vs. 5.5% among COVID-19 patients without CLD, p < 0.001) and a hospitalization rate of 41.0% (vs. 23.9% among COVID-19 patients without CLD, p < 0.001). INTERPRETATION: Patients with CLD, especially African Americans, were at increased risk for COVID-19, highlighting the need to protect these patients from exposure to virus infection. FUNDING: National Institutes of Health (AG057557, AG061388, AG062272, 1UL1TR002548-01), American Cancer Society (RSG-16-049-01-MPC).

9.
Alzheimers Dement ; 17(8): 1297-1306, 2021 08.
Article in English | MEDLINE | ID: mdl-33559975

ABSTRACT

INTRODUCTION: At present, there is limited data on the risks, disparity, and outcomes for COVID-19 in patients with dementia in the United States. METHODS: This is a retrospective case-control analysis of patient electronic health records (EHRs) of 61.9 million adult and senior patients (age ≥ 18 years) in the United States up to August 21, 2020. RESULTS: Patients with dementia were at increased risk for COVID-19 compared to patients without dementia (adjusted odds ratio [AOR]: 2.00 [95% confidence interval (CI), 1.94-2.06], P < .001), with the strongest effect for vascular dementia (AOR: 3.17 [95% CI, 2.97-3.37], P < .001), followed by presenile dementia (AOR: 2.62 [95% CI, 2.28-3.00], P < .001), Alzheimer's disease (AOR: 1.86 [95% CI, 1.77-1.96], P < .001), senile dementia (AOR: 1.99 [95% CI, 1.86-2.13], P < .001) and post-traumatic dementia (AOR: 1.67 [95% CI, 1.51-1.86] P < .001). Blacks with dementia had higher risk of COVID-19 than Whites (AOR: 2.86 [95% CI, 2.67-3.06], P < .001). The 6-month mortality and hospitalization risks in patients with dementia and COVID-19 were 20.99% and 59.26%, respectively. DISCUSSION: These findings highlight the need to protect patients with dementia as part of the strategy to control the COVID-19 pandemic.


Subject(s)
COVID-19/complications , Dementia/complications , Adolescent , Adult , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/epidemiology , Black People , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/epidemiology , COVID-19/epidemiology , Case-Control Studies , Dementia/epidemiology , Dementia, Vascular/complications , Dementia, Vascular/epidemiology , Demography , Electronic Health Records , Female , Healthcare Disparities , Hospitalization/statistics & numerical data , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , United States/epidemiology , White People , Young Adult
10.
Mol Psychiatry ; 26(9): 5286-5296, 2021 09.
Article in English | MEDLINE | ID: mdl-33432189

ABSTRACT

Morbidity and mortality from opioid use disorders (OUD) and other substance use disorders (SUD) is a major public health crisis, yet there are few medications to treat them. There is an urgency to accelerate SUD medication development. We present an integrated drug repurposing strategy that combines computational prediction, clinical corroboration using electronic health records (EHRs) of over 72.9 million patients and mechanisms of action analysis. Among top-ranked repurposed candidate drugs, tramadol, olanzapine, mirtazapine, bupropion, and atomoxetine were associated with increased odds of OUD remission (adjusted odds ratio: 1.51 [1.38-1.66], 1.90 [1.66-2.18], 1.38 [1.31-1.46], 1.37 [1.29-1.46], 1.48 [1.25-1.76], p value < 0.001, respectively). Genetic and functional analyses showed these five candidate drugs directly target multiple OUD-associated genes including BDNF, CYP2D6, OPRD1, OPRK1, OPRM1, HTR1B, POMC, SLC6A4 and OUD-associated pathways, including opioid signaling, G-protein activation, serotonin receptors, and GPCR signaling. In summary, we developed an integrated drug repurposing approach and identified five repurposed candidate drugs that might be of value for treating OUD patients, including those suffering from comorbid conditions.


Subject(s)
Drug Repositioning , Opioid-Related Disorders , Analgesics, Opioid/therapeutic use , Electronic Health Records , Humans , Odds Ratio , Opioid-Related Disorders/drug therapy , Serotonin Plasma Membrane Transport Proteins
11.
World Psychiatry ; 20(1): 124-130, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33026219

ABSTRACT

Concerns have been expressed that persons with a pre-existing mental disorder may represent a population at increased risk for COVID-19 infec-tion and with a higher likelihood of adverse outcomes of the infection, but there is no systematic research evidence in this respect. This study assessed the impact of a recent (within past year) diagnosis of a mental disorder - including attention-deficit/hyperactivity disorder (ADHD), bipolar disorder, depression and schizophrenia - on the risk for COVID-19 infection and related mortality and hospitalization rates. We analyzed a nation-wide database of electronic health records of 61 million adult patients from 360 hospitals and 317,000 providers, across 50 states in the US, up to July 29, 2020. Patients with a recent diagnosis of a mental disorder had a significantly increased risk for COVID-19 infection, an effect strongest for depression (adjusted odds ratio, AOR=7.64, 95% CI: 7.45-7.83, p<0.001) and schizophrenia (AOR=7.34, 95% CI: 6.65-8.10, p<0.001). Among patients with a recent diagnosis of a mental disorder, African Americans had higher odds of COVID-19 infection than Caucasians, with the strongest ethnic disparity for depression (AOR=3.78, 95% CI: 3.58-3.98, p<0.001). Women with mental disorders had higher odds of COVID-19 infection than males, with the strongest gender disparity for ADHD (AOR=2.03, 95% CI: 1.73-2.39, p<0.001). Patients with both a recent diagnosis of a mental disorder and COVID-19 infection had a death rate of 8.5% (vs. 4.7% among COVID-19 patients with no mental disorder, p<0.001) and a hospitalization rate of 27.4% (vs. 18.6% among COVID-19 patients with no mental disorder, p<0.001). These findings identify individuals with a recent diagnosis of a mental disorder as being at increased risk for COVID-19 infection, which is further exacerbated among African Americans and women, and as having a higher frequency of some adverse outcomes of the infection. This evidence highlights the need to identify and address modifiable vulnerability factors for COVID-19 infection and to prevent delays in health care provision in this population.

12.
Blood Rev ; 47: 100775, 2021 05.
Article in English | MEDLINE | ID: mdl-33187811

ABSTRACT

Scientific data is limited on the risks, adverse outcomes and racial disparities for COVID-19 illness in individuals with hematologic malignancies in the United States. To fill this void, we screened and analyzed a nation-wide database of patient electronic health records (EHRs) of 73 million patients in the US (up to September 1st) for COVID-19 and eight major types of hematologic malignancies. Patients with hematologic malignancies had increased odds of COVID-19 infection compared with patients without hematologic malignancies for both all-time diagnosis (malignancy diagnosed in the past year or prior) (adjusted Odds ratio or AOR: 2.27 [2.17-2.36], p < 0.001) and recent diagnosis (malignancy diagnosed in the past year) (AOR:11.91 [11.31-12.53], p < 0.001), with strongest effect for recently diagnosed acute lymphoid leukemia (AOR: 31.03 [25.87-37.27], p < 0.001), essential thrombocythemia (AOR: 20.65 [19.10-22.32], p < 0.001), acute myeloid leukemia (AOR: 18.94 [15.79-22.73], p < 0.001), multiple myeloma (AOR: 14.21 [12.72-15.89], p < 0.001). Among patients with hematologic malignancies, African Americans had higher odds of COVID-19 infection than Caucasians with largest racial disparity for multiple myeloma (AOR: 4.23 [3.21-5.56], p < 0.001). Patients with recently diagnosed hematologic malignancies had worse outcomes (hospitalization: 51.9%, death: 14.8%) than COVID-19 patients without hematologic malignancies (hospitalization: 23.5%, death: 5.1%) (p < 0.001) and hematologic malignancy patients without COVID-19 (hospitalization: 15.0%, death: 4.1%) (p < 0.001).


Subject(s)
COVID-19/epidemiology , Hematologic Neoplasms/complications , Adolescent , Adult , Aged , Female , Health Status Disparities , Hematologic Neoplasms/epidemiology , Hospitalization , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/epidemiology , Male , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/epidemiology , Odds Ratio , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Risk Factors , SARS-CoV-2/isolation & purification , United States/epidemiology , Young Adult
13.
JAMA Oncol ; 7(2): 220-227, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33300956

ABSTRACT

Importance: Patients with specific cancers may be at higher risk than those without cancer for coronavirus disease 2019 (COVID-19) and its severe outcomes. At present, limited data are available on the risk, racial disparity, and outcomes for COVID-19 illness in patients with cancer. Objectives: To investigate how patients with specific types of cancer are at risk for COVID-19 infection and its adverse outcomes and whether there are cancer-specific race disparities for COVID-19 infection. Design, Setting, and Participants: This retrospective case-control analysis of patient electronic health records included 73.4 million patients from 360 hospitals and 317 000 clinicians across 50 US states to August 14, 2020. The odds of COVID-19 infections for 13 common cancer types and adverse outcomes were assessed. Exposures: The exposure groups were patients diagnosed with a specific cancer, whereas the unexposed groups were patients without the specific cancer. Main Outcomes and Measures: The adjusted odds ratio (aOR) and 95% CI were estimated using the Cochran-Mantel-Haenszel test for the risk of COVID-19 infection. Results: Among the 73.4 million patients included in the analysis (53.6% female), 2 523 920 had at least 1 of the 13 common cancers diagnosed (all cancer diagnosed within or before the last year), and 273 140 had recent cancer (cancer diagnosed within the last year). Among 16 570 patients diagnosed with COVID-19, 1200 had a cancer diagnosis and 690 had a recent cancer diagnosis of at least 1 of the 13 common cancers. Those with recent cancer diagnosis were at significantly increased risk for COVID-19 infection (aOR, 7.14 [95% CI, 6.91-7.39]; P < .001), with the strongest association for recently diagnosed leukemia (aOR, 12.16 [95% CI, 11.03-13.40]; P < .001), non-Hodgkin lymphoma (aOR, 8.54 [95% CI, 7.80-9.36]; P < .001), and lung cancer (aOR, 7.66 [95% CI, 7.07-8.29]; P < .001) and weakest for thyroid cancer (aOR, 3.10 [95% CI, 2.47-3.87]; P < .001). Among patients with recent cancer diagnosis, African Americans had a significantly higher risk for COVID-19 infection than White patients; this racial disparity was largest for breast cancer (aOR, 5.44 [95% CI, 4.69-6.31]; P < .001), followed by prostate cancer (aOR, 5.10 [95% CI, 4.34-5.98]; P < .001), colorectal cancer (aOR, 3.30 [95% CI, 2.55-4.26]; P < .001), and lung cancer (aOR, 2.53 [95% CI, 2.10-3.06]; P < .001). Patients with cancer and COVID-19 had significantly worse outcomes (hospitalization, 47.46%; death, 14.93%) than patients with COVID-19 without cancer (hospitalization, 24.26%; death, 5.26%) (P < .001) and patients with cancer without COVID-19 (hospitalization, 12.39%; death, 4.03%) (P < .001). Conclusions and Relevance: In this case-control study, patients with cancer were at significantly increased risk for COVID-19 infection and worse outcomes, which was further exacerbated among African Americans. These findings highlight the need to protect and monitor patients with cancer as part of the strategy to control the pandemic.


Subject(s)
Black or African American/statistics & numerical data , COVID-19/epidemiology , Hospitalization/statistics & numerical data , Mortality , Neoplasms/epidemiology , White People/statistics & numerical data , Adult , Aged , Breast Neoplasms/epidemiology , COVID-19/ethnology , Case-Control Studies , Colorectal Neoplasms/epidemiology , Female , Humans , Lung Neoplasms/epidemiology , Male , Middle Aged , Odds Ratio , Prostatic Neoplasms/epidemiology , Risk Factors , SARS-CoV-2
14.
Sci Adv ; 6(49)2020 12.
Article in English | MEDLINE | ID: mdl-33277246

ABSTRACT

Myelin degeneration and white matter loss resulting from oligodendrocyte (OL) death are early events in Alzheimer's disease (AD) that lead to cognitive deficits; however, the underlying mechanism remains unknown. Here, we find that mature OLs in both AD patients and an AD mouse model undergo NLR family pyrin domain containing 3 (NLRP3)-dependent Gasdermin D-associated inflammatory injury, concomitant with demyelination and axonal degeneration. The mature OL-specific knockdown of dynamin-related protein 1 (Drp1; a mitochondrial fission guanosine triphosphatase) abolishes NLRP3 inflammasome activation, corrects myelin loss, and improves cognitive ability in AD mice. Drp1 hyperactivation in mature OLs induces a glycolytic defect in AD models by inhibiting hexokinase 1 (HK1; a mitochondrial enzyme that initiates glycolysis), which triggers NLRP3-associated inflammation. These findings suggest that OL glycolytic deficiency plays a causal role in AD development. The Drp1-HK1-NLRP3 signaling axis may be a key mechanism and therapeutic target for white matter degeneration in AD.


Subject(s)
Alzheimer Disease , Inflammasomes , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Glycolysis , Humans , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oligodendroglia/metabolism
15.
J Biomed Inform ; 109: 103524, 2020 09.
Article in English | MEDLINE | ID: mdl-32791237

ABSTRACT

MOTIVATION: Trillions of bacteria in human body (human microbiota) affect human health and diseases by controlling host functions through small molecule metabolites.An accurate and comprehensive catalog of the metabolic output from human microbiota is critical for our deep understanding of how microbial metabolism contributes to human health.The large number of published biomedical research articles is a rich resource of microbiome studies.However, automatically extracting microbial metabolites from free-text documents and differentiating them from other human metabolites is a challenging task.Here we developed an integrated approach called Co-occurrence Metabolite Network Ranking (CoMNRank) by combining named entity extraction, network construction and topic sensitive network-based prioritization to extract and prioritize microbial metabolites from biomedical articles. METHODS: The text data included 28,851,232 MEDLINE records.CoMNRank consists of three steps: (1) extraction of human metabolites from MEDLINE records; (2) construction of a weighted co-occurrence metabolite network (CoMN); (3) prioritization and differentiation of microbial metabolites from other human metabolites. RESULTS: For the first step of CoMNRank, we extracted 11,846 human metabolites from MEDLINE articles, with a baseline performance of precision of 0.014, recall of 0.959 and F1 of 0.028.We then constructed a weighted CoMN of 6,996 nodes and 986,186 edges.CoMNRank effectively prioritized microbial metabolites: the precision of top ranked metabolites is 0.45, a 31-fold enrichment as compared to the overall precision of 0.014.Manual curation of top 100 metabolites showed a true precision of 0.67, among which 48% true positives are not captured by existing databases. CONCLUSION: Our study sets the foundation for future tasks of microbial entity and relationship extractions as well as data-driven studies of how microbial metabolism contributes to human health and diseases.


Subject(s)
Data Mining , Publications , Databases, Factual , Humans , MEDLINE
16.
Sci Rep ; 10(1): 9996, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561832

ABSTRACT

Many diseases are driven by gene-environment interactions. One important environmental factor is the metabolic output of human gut microbiota. A comprehensive catalog of human metabolites originated in microbes is critical for data-driven approaches to understand how microbial metabolism contributes to human health and diseases. Here we present a novel integrated approach to automatically extract and analyze microbial metabolites from 28 million published biomedical records. First, we classified 28,851,232 MEDLINE records into microbial metabolism-related or not. Second, candidate microbial metabolites were extracted from the classified texts. Third, we developed signal prioritization algorithms to further differentiate microbial metabolites from metabolites originated from other resources. Finally, we systematically analyzed the interactions between extracted microbial metabolites and human genes. A total of 11,846 metabolites were extracted from 28 million MEDLINE articles. The combined text classification and signal prioritization significantly enriched true positives among top: manual curation of top 100 metabolites showed a true precision of 0.55, representing a significant 38.3-fold enrichment as compared to the precision of 0.014 for baseline extraction. More importantly, 29% extracted microbial metabolites have not been captured by existing databases. We performed data-driven analysis of the interactions between the extracted microbial metabolite and human genetics. This study represents the first effort towards automatically extracting and prioritizing microbial metabolites from published biomedical literature, which can set a foundation for future tasks of microbial metabolite relationship extraction from literature and facilitate data-driven studies of how microbial metabolism contributes to human diseases.


Subject(s)
Data Mining , Gastrointestinal Microbiome/physiology , Databases, Factual , Humans , MEDLINE
17.
JAMIA Open ; 2(1): 173-178, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30976759

ABSTRACT

OBJECTIVES: Immune checkpoint inhibitors (ICIs) have dramatically improved outcomes in cancer patients. However, ICIs are associated with significant immune-related adverse events (irAEs) and the underlying biological mechanisms are not well-understood. To ensure safe cancer treatment, research efforts are needed to comprehensively detect and understand irAEs. MATERIALS AND METHODS: We manually extracted and standardized irAEs from The U.S Food and Drug Administration (FDA) drug labels for six FDA-approved ICIs. We compared irAE profile similarities among ICIs and 1507 FDA-approved non-ICI drugs. We investigated how irAEs have differential effects on human organs by classifying irAEs based on their targeted organ systems. Finally, we identified broad-spectrum (nontarget-specific) and narrow-spectrum (target-specific) irAEs. RESULTS: A total of 893 irAEs were extracted. 31.4% irAEs were shared among ICIs as compared to the 8.0% between ICIs and non-ICI drugs. irAEs were resulted from both on- and off-target effects: irAE profiles were more similar for ICIs with same target than different targets, demonstrating the on-target effects; irAE profile similarity for ICIs with the same target is less than 50%, demonstrating unknown off-target effects. ICIs significantly target many organ systems, including endocrine system (3.4-fold enrichment), metabolism (3.7-fold enrichment), immune system (3.6-fold enrichment), and autoimmune system (4.8-fold enrichment). We identified 21 broad-spectrum irAEs shared among all ICIs, 20 irAEs specific for PD-L1/PD-1 inhibition, and 28 irAEs specific for CTLA-4 inhibition. DISCUSSION AND CONCLUSION: Our study presents the first effort toward building a standardized database of irAEs. The extracted irAEs can serve as the gold standard for automatic irAE extractions from other data resources and set a foundation to understand biological mechanisms of irAEs.

18.
BMC Genomics ; 20(1): 124, 2019 Feb 11.
Article in English | MEDLINE | ID: mdl-30744546

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is the most common autoimmune disease and affects about 1% of the population. The cause of RA remains largely unknown and could result from a complex interaction between genes and environment factors. Recent studies suggested that gut microbiota and their collective metabolic outputs exert profound effects on the host immune system and are implicated in RA. However, which and how gut microbial metabolites interact with host genetics in contributing to RA pathogenesis remains unknown. In this study, we present a data-driven study to understand how gut microbial metabolites contribute to RA at the genetic, functional and phenotypic levels. RESULTS: We used publicly available disease genetics, chemical genetics, human metabolome, genetic signaling pathways, mouse genome-wide mutation phenotypes, and mouse phenotype ontology data. We identified RA-associated microbial metabolites and prioritized them based on their genetic, functional and phenotypic relevance to RA. We evaluated the prioritization methods using short-chain fatty acids (SCFAs), which were previously shown to be involved in RA etiology. We validate the algorithms by showing that SCFAs are highly associated with RA at genetic, functional and phenotypic levels: SCFAs ranked at top 3.52% based on shared genes with RA, top 5.69% based on shared genetic pathways, and top 16.94% based on shared phenotypes. Based on the genetic-level analysis, human gut microbial metabolites directly interact with many RA-associated genes (as many as 18.1% of all 166 RA genes). Based on the functional-level analysis, human gut microbial metabolites participate in many RA-associated genetic pathways (as many as 71.4% of 311 genetic pathways significantly enriched for RA), including immune system pathways. Based on the phenotypic-level analysis, gut microbial metabolites affect many RA-related phenotypes (as many as 51.3% of 978 phenotypes significantly enriched for RA), including many immune system phenotypes. CONCLUSIONS: Our study demonstrates strong gut-microbiome-immune-joint interactions in RA, which converged on both genetic, functional and phenotypic levels.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/microbiology , Computational Biology , Gastrointestinal Microbiome , Joints/metabolism , Joints/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Humans , Metabolomics , Mutation , Phenotype , Signal Transduction
19.
Gynecol Oncol ; 151(3): 525-532, 2018 12.
Article in English | MEDLINE | ID: mdl-30301560

ABSTRACT

OBJECTIVE: To evaluate the utility of amiodarone and its derivative dronedarone as novel drug repositioning candidates in EOC and to determine the potential pathways targeted by these drugs. METHODS: Drug-predict bioinformatics platform was used to assess the utility of amiodarone as a novel drug-repurposing candidate in EOC. EOC cells were treated with amiodarone and dronedarone. Cell death was assessed by Annexin V staining. Cell viability and cell survival were assessed by MTT and clonogenics assays respectively. c-MYC and mTOR/Akt axis were evaluated as potential targets. Effect on autophagy was determined by autophagy flux flow cytometry. RESULTS: "DrugPredict" bioinformatics platform ranked Class III antiarrhythmic drug amiodarone within the top 3.9% of potential EOC drug repositioning candidates which was comparable to carboplatin ranking in the top 3.7%. Amiodarone and dronedarone were the only Class III antiarrhythmic drugs that decreased the cellular survival of both cisplatin-sensitive and cisplatin-resistant primary EOC cells. Interestingly, both drugs induced degradation of c-MYC protein and decreased the expression of known transcriptional targets of c-MYC. Furthermore, stable overexpression of non-degradable c-MYC partially rescued the effects of amiodarone and dronedarone induced cell death. Dronedarone induced higher autophagy flux in EOC cells as compared to amiodarone with decreased phospho-AKT and phospho-4EBP1 protein expression, suggesting autophagy induction due to inhibition of AKT/mTOR axis with these drugs. Lastly, both drugs also inhibited the survival of EOC tumor-initiating cells (TICs). CONCLUSIONS: We provide the first evidence of class III antiarrhythmic agents as novel c-MYC targeting drugs and autophagy inducers in EOC. Since c-MYC is amplified in >40% ovarian tumors, our results provide the basis for repositioning amiodarone and dronedarone as novel c-MYC targeting drugs in EOC with potential extension to other cancers.


Subject(s)
Amiodarone/therapeutic use , Anti-Arrhythmia Agents/therapeutic use , Dronedarone/therapeutic use , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/drug therapy , Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Dronedarone/pharmacology , Female , Humans , Ovarian Neoplasms/pathology
20.
Sci Rep ; 8(1): 6225, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29670137

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer-related deaths. It is estimated that about half the cases of CRC occurring today are preventable. Recent studies showed that human gut microbiota and their collective metabolic outputs play important roles in CRC. However, the mechanisms by which human gut microbial metabolites interact with host genetics in contributing CRC remain largely unknown. We hypothesize that computational approaches that integrate and analyze vast amounts of publicly available biomedical data have great potential in better understanding how human gut microbial metabolites are mechanistically involved in CRC. Leveraging vast amount of publicly available data, we developed a computational algorithm to predict human gut microbial metabolites for CRC. We validated the prediction algorithm by showing that previously known CRC-associated gut microbial metabolites ranked highly (mean ranking: top 10.52%; median ranking: 6.29%; p-value: 3.85E-16). Moreover, we identified new gut microbial metabolites likely associated with CRC. Through computational analysis, we propose potential roles for tartaric acid, the top one ranked metabolite, in CRC etiology. In summary, our data-driven computation-based study generated a large amount of associations that could serve as a starting point for further experiments to refute or validate these microbial metabolite associations in CRC cancer.


Subject(s)
Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Energy Metabolism , Gastrointestinal Microbiome , Systems Biology , Animals , Cell Culture Techniques , Disease Susceptibility , Humans , Metabolome , Metabolomics/methods , Mice , Microbiota , Phenotype , Systems Biology/methods
SELECTION OF CITATIONS
SEARCH DETAIL