Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Crit Care ; 28(1): 63, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38414082

ABSTRACT

RATIONALE: Acute respiratory distress syndrome (ARDS) is a life-threatening critical care syndrome commonly associated with infections such as COVID-19, influenza, and bacterial pneumonia. Ongoing research aims to improve our understanding of ARDS, including its molecular mechanisms, individualized treatment options, and potential interventions to reduce inflammation and promote lung repair. OBJECTIVE: To map and compare metabolic phenotypes of different infectious causes of ARDS to better understand the metabolic pathways involved in the underlying pathogenesis. METHODS: We analyzed metabolic phenotypes of 3 ARDS cohorts caused by COVID-19, H1N1 influenza, and bacterial pneumonia compared to non-ARDS COVID-19-infected patients and ICU-ventilated controls. Targeted metabolomics was performed on plasma samples from a total of 150 patients using quantitative LC-MS/MS and DI-MS/MS analytical platforms. RESULTS: Distinct metabolic phenotypes were detected between different infectious causes of ARDS. There were metabolomics differences between ARDSs associated with COVID-19 and H1N1, which include metabolic pathways involving taurine and hypotaurine, pyruvate, TCA cycle metabolites, lysine, and glycerophospholipids. ARDSs associated with bacterial pneumonia and COVID-19 differed in the metabolism of D-glutamine and D-glutamate, arginine, proline, histidine, and pyruvate. The metabolic profile of COVID-19 ARDS (C19/A) patients admitted to the ICU differed from COVID-19 pneumonia (C19/P) patients who were not admitted to the ICU in metabolisms of phenylalanine, tryptophan, lysine, and tyrosine. Metabolomics analysis revealed significant differences between C19/A, H1N1/A, and PNA/A vs ICU-ventilated controls, reflecting potentially different disease mechanisms. CONCLUSION: Different metabolic phenotypes characterize ARDS associated with different viral and bacterial infections.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Pneumonia, Bacterial , Respiratory Distress Syndrome , Humans , COVID-19/complications , Influenza, Human/complications , Influenza, Human/therapy , Tandem Mass Spectrometry , Chromatography, Liquid , Lysine , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/therapy , Pyruvates
2.
Metabolites ; 13(11)2023 Nov 10.
Article in English | MEDLINE | ID: mdl-37999238

ABSTRACT

Sepsis is the result of an uncontrolled host inflammatory response to infection that may lead to septic shock with multiorgan failure and a high mortality rate. There is an urgent need to improve early diagnosis and to find markers identifying those who will develop septic shock and certainly a need to develop targeted treatments to prevent septic shock and its high mortality. Herein, we explore metabolic alterations due to mesenchymal stromal cell (MSC) treatment of septic shock. The clinical findings for this study were already reported; MSC therapy was well-tolerated and safe in patients in this phase I clinical trial. In this exploratory metabolomics study, 9 out of 30 patients received an escalating dose of MSC treatment, while 21 patients were without MSC treatment. Serum metabolomics profiling was performed to detect and characterize metabolite changes due to MSC treatment and to help determine the sample size needed for a phase II clinical trial and to define a metabolomic response to MSC treatment. Serum metabolites were measured using 1H-NMR and HILIC-MS at times 0, 24 and 72 h after MSC infusion. The results demonstrated the significant impact of MSC treatment on serum metabolic changes in a dose- and time-dependent manner compared to non-MSC-treated septic shock patients. This study suggests that plasma metabolomics can be used to assess the response to MSC therapy and that treatment-related metabolomics effects can be used to help determine the sample size needed in a phase II trial. As this study was not powered to detect outcome, how the treatment-induced metabolomic changes described in this study of MSC-treated septic shock patients are related to outcomes of septic shock in the short and long term will need to be explored in a larger adequately powered phase II clinical trial.

3.
BMC Neurosci ; 24(1): 54, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37845610

ABSTRACT

BACKGROUND: Diagnosis and prognostication of severe traumatic brain injury (sTBI) continue to be problematic despite years of research efforts. There are currently no clinically reliable biomarkers, though advances in protein biomarkers are being made. Utilizing Omics technology, particularly metabolomics, may provide new diagnostic biomarkers for sTBI. Several published studies have attempted to determine the specific metabolites and metabolic pathways involved; these studies will be reviewed. AIMS: This scoping review aims to summarize the current literature concerning metabolomics in sTBI, review the comprehensive data, and identify commonalities, if any, to define metabolites with potential clinical use. In addition, we will examine related metabolic pathways through pathway analysis. METHODS: Scoping review methodology was used to examine the current literature published in Embase, Scopus, PubMed, and Medline. An initial 1090 publications were identified and vetted with specific inclusion criteria. Of these, 20 publications were selected for further examination and summary. Metabolic data was classified using the Human Metabolome Database (HMDB) and arranged to determine the 'recurrent' metabolites and classes found in sTBI. To help understand potential mechanisms of injury, pathway analysis was performed using these metabolites and the Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database. RESULTS: Several metabolites related to sTBI and their effects on biological pathways were identified in this review. Across the literature, proline, citrulline, lactate, alanine, valine, leucine, and serine all decreased in adults post sTBI, whereas both octanoic and decanoic acid increased. Hydroxy acids and organooxygen compounds generally increased following sTBI, while most carboxylic acids decreased. Pathway analysis showed significantly affected glycine and serine metabolism, glycolysis, branched-chain amino acid (BCAA) metabolism, and other amino acid metabolisms. Interestingly, no tricarboxylic acid cycle metabolites were affected. CONCLUSION: Aside from a select few metabolites, classification of a metabolic profile proved difficult due to significant ambiguity between study design, sample size, type of sample, metabolomic detection techniques, and other confounding variables found in sTBI literature. Given the trends found in some studies, further metabolomics investigation of sTBI may be useful to identify clinically relevant metabolites.


Subject(s)
Brain Injuries, Traumatic , Adult , Humans , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/metabolism , Metabolomics/methods , Metabolome , Biomarkers/metabolism , Serine/metabolism
4.
Crit Care ; 27(1): 295, 2023 07 22.
Article in English | MEDLINE | ID: mdl-37481590

ABSTRACT

BACKGROUND: Prognostication is very important to clinicians and families during the early management of severe traumatic brain injury (sTBI), however, there are no gold standard biomarkers to determine prognosis in sTBI. As has been demonstrated in several diseases, early measurement of serum metabolomic profiles can be used as sensitive and specific biomarkers to predict outcomes. METHODS: We prospectively enrolled 59 adults with sTBI (Glasgow coma scale, GCS ≤ 8) in a multicenter Canadian TBI (CanTBI) study. Serum samples were drawn for metabolomic profiling on the 1st and 4th days following injury. The Glasgow outcome scale extended (GOSE) was collected at 3- and 12-months post-injury. Targeted direct infusion liquid chromatography-tandem mass spectrometry (DI/LC-MS/MS) and untargeted proton nuclear magnetic resonance spectroscopy (1H-NMR) were used to profile serum metabolites. Multivariate analysis was used to determine the association between serum metabolomics and GOSE, dichotomized into favorable (GOSE 5-8) and unfavorable (GOSE 1-4), outcomes. RESULTS: Serum metabolic profiles on days 1 and 4 post-injury were highly predictive (Q2 > 0.4-0.5) and highly accurate (AUC > 0.99) to predict GOSE outcome at 3- and 12-months post-injury and mortality at 3 months. The metabolic profiles on day 4 were more predictive (Q2 > 0.55) than those measured on day 1 post-injury. Unfavorable outcomes were associated with considerable metabolite changes from day 1 to day 4 compared to favorable outcomes. Increased lysophosphatidylcholines, acylcarnitines, energy-related metabolites (glucose, lactate), aromatic amino acids, and glutamate were associated with poor outcomes and mortality. DISCUSSION: Metabolomic profiles were strongly associated with the prognosis of GOSE outcome at 3 and 12 months and mortality following sTBI in adults. The metabolic phenotypes on day 4 post-injury were more predictive and significant for predicting the sTBI outcome compared to the day 1 sample. This may reflect the larger contribution of secondary brain injury (day 4) to sTBI outcome. Patients with unfavorable outcomes demonstrated more metabolite changes from day 1 to day 4 post-injury. These findings highlighted increased concentration of neurobiomarkers such as N-acetylaspartate (NAA) and tyrosine, decreased concentrations of ketone bodies, and decreased urea cycle metabolites on day 4 presenting potential metabolites to predict the outcome. The current findings strongly support the use of serum metabolomics, that are shown to be better than clinical data, in determining prognosis in adults with sTBI in the early days post-injury. Our findings, however, require validation in a larger cohort of adults with sTBI to be used for clinical practice.


Subject(s)
Brain Injuries, Traumatic , Tandem Mass Spectrometry , Humans , Glasgow Outcome Scale , Chromatography, Liquid , Canada , Brain Injuries, Traumatic/complications , Metabolomics , Lactic Acid
5.
Front Med (Lausanne) ; 10: 1170331, 2023.
Article in English | MEDLINE | ID: mdl-37215714

ABSTRACT

Background: At the end of 2019, the coronavirus disease 2019 (COVID-19) pandemic increased the hospital burden of COVID-19 caused by the SARS-Cov-2 and became the most significant health challenge for nations worldwide. The severity and high mortality of COVID-19 have been correlated with various demographic characteristics and clinical manifestations. Prediction of mortality rate, identification of risk factors, and classification of patients played a crucial role in managing COVID-19 patients. Our purpose was to develop machine learning (ML)-based models for the prediction of mortality and severity among patients with COVID-19. Identifying the most important predictors and unraveling their relationships by classification of patients to the low-, moderate- and high-risk groups might guide prioritizing treatment decisions and a better understanding of interactions between factors. A detailed evaluation of patient data is believed to be important since COVID-19 resurgence is underway in many countries. Results: The findings of this study revealed that the ML-based statistically inspired modification of the partial least square (SIMPLS) method could predict the in-hospital mortality among COVID-19 patients. The prediction model was developed using 19 predictors including clinical variables, comorbidities, and blood markers with moderate predictability (Q2 = 0.24) to separate survivors and non-survivors. Oxygen saturation level, loss of consciousness, and chronic kidney disease (CKD) were the top mortality predictors. Correlation analysis showed different correlation patterns among predictors for each non-survivor and survivor cohort separately. The main prediction model was verified using other ML-based analyses with a high area under the curve (AUC) (0.81-0.93) and specificity (0.94-0.99). The obtained data revealed that the mortality prediction model can be different for males and females with diverse predictors. Patients were classified into four clusters of mortality risk and identified the patients at the highest risk of mortality, which accentuated the most significant predictors correlating with mortality. Conclusion: An ML model for predicting mortality among hospitalized COVID-19 patients was developed considering the interactions between factors that may reduce the complexity of clinical decision-making processes. The most predictive factors related to patient mortality were identified by assessing and classifying patients into different groups based on their sex and mortality risk (low-, moderate-, and high-risk groups).

6.
Sci Rep ; 12(1): 8294, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35585165

ABSTRACT

Follicular lymphoma (FL) is a cancer of B-cells, representing the second most common type of non-Hodgkin lymphoma and typically diagnosed at advanced stage in older adults. In contrast to the wide range of available molecular genetic data, limited data relating the metabolomic features of follicular lymphoma are known. Metabolomics is a promising analytical approach employing metabolites (molecules < 1 kDa in size) as potential biomarkers in cancer research. In this pilot study, we performed proton nuclear magnetic resonance spectroscopy (1H-NMR) on 29 cases of FL and 11 control patient specimens. The resulting spectra were assessed by both unsupervised and supervised statistical methods. We report significantly discriminant metabolomic models of common metabolites distinguishing FL from control tissues. Within our FL case series, we also report discriminant metabolomic signatures predictive of progression-free survival.


Subject(s)
Lymphoma, Follicular , Aged , Humans , Lymph Nodes , Lymphoma, Follicular/diagnosis , Magnetic Resonance Spectroscopy/methods , Metabolomics/methods , Pilot Projects
7.
Physiol Rep ; 10(9): e15286, 2022 05.
Article in English | MEDLINE | ID: mdl-35510328

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a lung injury characterized by noncardiogenic pulmonary edema and hypoxic respiratory failure. The purpose of this study was to investigate the effects of therapeutic hypothermia on short-term experimental ARDS. Twenty adult female Yorkshire pigs were divided into four groups (n = 5 each): normothermic control (C), normothermic injured (I), hypothermic control (HC), and hypothermic injured (HI). Acute respiratory distress syndrome was induced experimentally via intrapulmonary injection of oleic acid. Target core temperature was achieved in the HI group within 1 h of injury induction. Cardiorespiratory, histologic, cytokine, and metabolomic data were collected on all animals prior to and following injury/sham. All data were collected for approximately 12 h from the beginning of the study until euthanasia. Therapeutic hypothermia reduced injury in the HI compared to the I group (histological injury score = 0.51 ± 0.18 vs. 0.76 ± 0.06; p = 0.02) with no change in gas exchange. All groups expressed distinct phenotypes, with a reduction in pro-inflammatory metabolites, an increase in anti-inflammatory metabolites, and a reduction in inflammatory cytokines observed in the HI group compared to the I group. Changes to respiratory system mechanics in the injured groups were due to increases in lung elastance (E) and resistance (R) (ΔE from pre-injury = 46 ± 14 cmH2 O L-1 , p < 0.0001; ΔR from pre-injury: 3 ± 2 cmH2 O L-1  s- , p = 0.30) rather than changes to the chest wall (ΔE from pre-injury: 0.7 ± 1.6 cmH2 O L-1 , p = 0.99; ΔR from pre-injury: 0.6 ± 0.1 cmH2 O L-1  s- , p = 0.01). Both control groups had no change in respiratory mechanics. In conclusion, therapeutic hypothermia can reduce markers of injury and inflammation associated with experimentally induced short-term ARDS.


Subject(s)
Hypothermia, Induced , Lung Injury , Respiratory Distress Syndrome , Animals , Biomarkers , Cytokines , Female , Lung/pathology , Respiratory Distress Syndrome/therapy , Respiratory Mechanics , Swine
8.
Crit Care ; 25(1): 328, 2021 09 08.
Article in English | MEDLINE | ID: mdl-34496940

ABSTRACT

BACKGROUND: The coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-Cov2 virus has become the greatest health and controversial issue for worldwide nations. It is associated with different clinical manifestations and a high mortality rate. Predicting mortality and identifying outcome predictors are crucial for COVID patients who are critically ill. Multivariate and machine learning methods may be used for developing prediction models and reduce the complexity of clinical phenotypes. METHODS: Multivariate predictive analysis was applied to 108 out of 250 clinical features, comorbidities, and blood markers captured at the admission time from a hospitalized cohort of patients (N = 250) with COVID-19. Inspired modification of partial least square (SIMPLS)-based model was developed to predict hospital mortality. Prediction accuracy was randomly assigned to training and validation sets. Predictive partition analysis was performed to obtain cutting value for either continuous or categorical variables. Latent class analysis (LCA) was carried to cluster the patients with COVID-19 to identify low- and high-risk patients. Principal component analysis and LCA were used to find a subgroup of survivors that tends to die. RESULTS: SIMPLS-based model was able to predict hospital mortality in patients with COVID-19 with moderate predictive power (Q2 = 0.24) and high accuracy (AUC > 0.85) through separating non-survivors from survivors developed using training and validation sets. This model was obtained by the 18 clinical and comorbidities predictors and 3 blood biochemical markers. Coronary artery disease, diabetes, Altered Mental Status, age > 65, and dementia were the topmost differentiating mortality predictors. CRP, prothrombin, and lactate were the most differentiating biochemical markers in the mortality prediction model. Clustering analysis identified high- and low-risk patients among COVID-19 survivors. CONCLUSIONS: An accurate COVID-19 mortality prediction model among hospitalized patients based on the clinical features and comorbidities may play a beneficial role in the clinical setting to better management of patients with COVID-19. The current study revealed the application of machine-learning-based approaches to predict hospital mortality in patients with COVID-19 and identification of most important predictors from clinical, comorbidities and blood biochemical variables as well as recognizing high- and low-risk COVID-19 survivors.


Subject(s)
COVID-19/mortality , Hospital Mortality/trends , Machine Learning/standards , Severity of Illness Index , COVID-19/epidemiology , Cohort Studies , Female , Humans , Male , Prognosis , Respiration, Artificial/statistics & numerical data , Risk Assessment/methods , Risk Factors
9.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L79-L90, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33949201

ABSTRACT

In this study, we aimed to identify acute respiratory distress syndrome (ARDS) metabolic fingerprints in selected patient cohorts and compare the metabolic profiles of direct versus indirect ARDS and hypoinflammatory versus hyperinflammatory ARDS. We hypothesized that the biological and inflammatory processes in ARDS would manifest as unique metabolomic fingerprints that set ARDS apart from other intensive care unit (ICU) conditions and could help examine ARDS subphenotypes and clinical subgroups. Patients with ARDS (n = 108) and ICU ventilated controls (n = 27) were included. Samples were randomly divided into 2/3 training and 1/3 test sets. Samples were analyzed using 1H nuclear magnetic resonance spectroscopy and gas chromatography-mass spectrometry. Twelve proteins/cytokines were also measured. Orthogonal partial least squares discriminant analysis (OPLS-DA) was used to select the most differentiating ARDS metabolites and protein/cytokines. Predictive performance of OPLS-DA models was measured in the test set. Temporal changes of metabolites were examined as patients progressed through ARDS until clinical recovery. Metabolic profiles of direct versus indirect ARDS subgroups and hypoinflammatory versus hyperinflammatory ARDS subgroups were compared. Serum metabolomics and proteins/cytokines had similar area under receiver operator curves when distinguishing ARDS from ICU controls. Pathway analysis of ARDS differentiating metabolites identified a dominant involvement of serine-glycine metabolism. In longitudinal tracking, the identified pathway metabolites generally exhibited correction by 7-14 days, coinciding with clinical improvement. ARDS subphenotypes and clinical subgroups were metabolically distinct. However, our identified metabolic fingerprints are not ARDS diagnostic biomarkers, and further research is required to ascertain generalizability. In conclusion, patients with ARDS are metabolically different from ICU controls. ARDS subphenotypes and clinical subgroups are metabolically distinct.


Subject(s)
Benchmarking/methods , Biomarkers/metabolism , Metabolome , Respiratory Distress Syndrome/pathology , Aged , Biomarkers/analysis , Case-Control Studies , Discriminant Analysis , Female , Humans , Male , Middle Aged , Respiratory Distress Syndrome/metabolism
10.
Crit Care ; 24(1): 461, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32718333

ABSTRACT

INTRODUCTION: Pneumonia is the most common cause of mortality from infectious diseases, the second leading cause of nosocomial infection, and the leading cause of mortality among hospitalized adults. To improve clinical management, metabolomics has been increasingly applied to find specific metabolic biopatterns (profiling) for the diagnosis and prognosis of various infectious diseases, including pneumonia. METHODS: One hundred fifty bacterial community-acquired pneumonia (CAP) patients whose plasma samples were drawn within the first 24 h of hospital admission were enrolled in this study and separated into two age- and sex-matched cohorts: non-survivors (died ≤ 90 days) and survivors (survived > 90 days). Three analytical tools, 1H-NMR spectroscopy, GC-MS, and targeted DI-MS/MS, were used to prognosticate non-survivors from survivors by means of metabolic profiles. RESULTS: We show that quantitative lipid profiling using DI-MS/MS can predict the 90-day mortality and in-hospital mortality among patients with bacterial CAP compared to 1H-NMR- and GC-MS-based metabolomics. This study showed that the decreased lysophosphatidylcholines and increased acylcarnitines are significantly associated with increased mortality in bacterial CAP. Additionally, we found that decreased lysophosphatidylcholines and phosphatidylcholines (> 36 carbons) and increased acylcarnitines may be used to predict the prognosis of in-hospital mortality for bacterial CAP as well as the need for ICU admission and severity of bacterial CAP. DISCUSSION: This study demonstrates that lipid-based plasma metabolites can be used for the prognosis of 90-day mortality among patients with bacterial CAP. Moreover, lipid profiling can be utilized to identify patients with bacterial CAP who are at the highest risk of dying in hospital and who need ICU admission as well as the severity assessment of CAP.


Subject(s)
Hospital Mortality/trends , Lipids/analysis , Pneumonia/blood , Prognosis , Aged , Aged, 80 and over , Alberta , Case-Control Studies , Community-Acquired Infections/blood , Community-Acquired Infections/mortality , Female , Hospitalization/statistics & numerical data , Humans , Intensive Care Units/organization & administration , Intensive Care Units/statistics & numerical data , Lipids/blood , Male , Middle Aged , Pennsylvania , Pneumonia/mortality , Retrospective Studies
11.
Article in English | MEDLINE | ID: mdl-31797714

ABSTRACT

Breast cancer is the most common malignancy and the second leading cause of cancer deaths among women worldwide after lung cancer. Mitochondria play a central role in the regulation of cellular function, metabolism, and cell death in cancer cells. We aim to examine the mitochondrial polymorphisms of complex I in association with breast cancer in an Iranian cohort.This experimental study includes 53 patients with breast cancer and 35 healthy control patients. In addition, tumor-adjacent normal breast tissue was obtained from each patient. The DNA of the tissue cells was extracted and analyzed for complex I mutations using a PCR sequencing method. Our results show 94 mtDNA complex I variants in tumor tissues. A10398G was the most prevalent polymorphism and strongly correlated with Her2 receptor in tumor tissue samples. Mitochondrial DNA (mtDNA) mutations have been widely linked to the etiology of numerous disorders. The mtDNA mutations screening on A10398G along with other mutations might provide insight on the role of mitochondrial mutations in breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA, Mitochondrial/genetics , Genetic Predisposition to Disease , Mutation , Receptor, ErbB-2/genetics , Adult , Aged , Female , Humans , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors
12.
Sci Rep ; 9(1): 19584, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31863066

ABSTRACT

Sarcoidosis is a disorder characterized by granulomatous inflammation of unclear etiology. In this study we evaluated whether veterans with sarcoidosis exhibited different plasma metabolomic and metallomic profiles compared with civilians with sarcoidosis. A case control study was performed on veteran and civilian patients with confirmed sarcoidosis. Proton nuclear magnetic resonance spectroscopy (1H NMR), hydrophilic interaction liquid chromatography mass spectrometry (HILIC-MS) and inductively coupled plasma mass spectrometry (ICP-MS) were applied to quantify metabolites and metal elements in plasma samples. Our results revealed that the veterans with sarcoidosis significantly differed from civilians, according to metabolic and metallomics profiles. Moreover, the results showed that veterans with sarcoidosis and veterans with COPD were similar to each other in metabolomics and metallomics profiles. This study suggests the important role of environmental risk factors in the development of different molecular phenotypic responses of sarcoidosis. In addition, this study suggests that sarcoidosis in veterans may be an occupational disease.


Subject(s)
Metabolomics , Metals/chemistry , Pulmonary Disease, Chronic Obstructive/metabolism , Sarcoidosis, Pulmonary/metabolism , Case-Control Studies , Chromatography, Liquid , Discriminant Analysis , Female , Humans , Least-Squares Analysis , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Phenotype , Protons , Pulmonary Disease, Chronic Obstructive/diagnosis , Retrospective Studies , Sarcoidosis, Pulmonary/diagnosis , Translational Research, Biomedical , United States , Veterans
13.
Neurocrit Care ; 30(1): 22-32, 2019 02.
Article in English | MEDLINE | ID: mdl-29569129

ABSTRACT

This scoping review will discuss the basic functions and prognostic significance of the commonly researched cytokines implicated in severe traumatic brain injury (sTBI), including tumour necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), IL-6, tissue inhibitor of matrix metalloproteinases-1 (TIMP-1), transforming growth factor-ß (TGF-ß), substance P, and soluble CD40 ligand (sCD40L). A scoping review was undertaken with an electronic search for articles from the Ovid MEDLINE, PUBMED and EMBASE databases from 1995 to 2017. Inclusion criteria were original research articles, and reviews including both animal models and human clinical studies of acute (< 3 months) sTBI. Selected articles included both isolated sTBI and sTBI with systemic injury. After applying the inclusion criteria and removing duplicates, 141 full-text articles, 126 original research articles and 15 review articles, were evaluated in compiling this review paper. A single reviewer, CC, completed the review in two phases. During the first phase, titles and abstracts of selected articles were reviewed for inclusion. A second evaluation was then conducted on the full text of all selected articles to ensure relevancy. From our current understanding of the literature, it is unlikely a single biomarker will be sufficient in accurately prognosticating patients with sTBI. Intuitively, a more severe injury will demonstrate higher levels of inflammatory cytokines which may correlate as a marker of severe injury. This does not mean, necessarily, these cytokines have a direct and causal role in the poor outcome of the patient. Further research is required to better delineate the complex systemic inflammatory and CNS interactions that occur during sTBI before they can be applied as a reliable prognostic tool.


Subject(s)
Biomarkers/metabolism , Brain Injuries, Traumatic/diagnosis , Cytokines/metabolism , Animals , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/metabolism , Humans
14.
Am J Physiol Lung Cell Mol Physiol ; 315(4): L526-L534, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29952222

ABSTRACT

To date, there is no clinically agreed-upon diagnostic test for acute respiratory distress syndrome (ARDS): the condition is still diagnosed on the basis of a constellation of clinical findings, laboratory tests, and radiological images. Development of ARDS biomarkers has been in a state of continuous flux during the past four decades. To address ARDS heterogeneity, several studies have recently focused on subphenotyping the disease on the basis of observable clinical characteristics and associated blood biomarkers. However, the strong correlation between identified biomarkers and ARDS subphenotypes has yet to establish etiology; hence, there is a need for the adoption of other methodologies for studying ARDS. In this review, we will shed light on ARDS metabolomics research in the literature and discuss advances and major obstacles encountered in ARDS metabolomics research. Generally, the ARDS metabolomics studies focused on identification of differentiating metabolites for diagnosing ARDS, but they were performed to different standards in terms of sample size, selection of control cohort, type of specimens collected, and measuring technique utilized. Virtually none of these studies have been properly validated to identify true metabolomics biomarkers of ARDS. Though in their infancy, metabolomics studies exhibit promise to unfold the biological processes underlying ARDS and, in our opinion, have great potential for pushing forward our present understanding of ARDS.


Subject(s)
Biomarkers/metabolism , Metabolome , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/metabolism , Humans
15.
J Neurotrauma ; 35(16): 1831-1848, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29587568

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of disability and mortality worldwide. The TBI pathogenesis can induce broad pathophysiological consequences and clinical outcomes attributed to the complexity of the brain. Thus, the diagnosis and prognosis are important issues for the management of mild, moderate, and severe forms of TBI. Metabolomics of readily accessible biofluids is a promising tool for establishing more useful and reliable biomarkers of TBI than using clinical findings alone. Metabolites are an integral part of all biochemical and pathophysiological pathways. Metabolomic processes respond to the internal and external stimuli resulting in an alteration of metabolite concentrations. Current high-throughput and highly sensitive analytical tools are capable of detecting and quantifying small concentrations of metabolites, allowing one to measure metabolite alterations after a pathological event when compared to a normal state or a different pathological process. Further, these metabolic biomarkers could be used for the assessment of injury severity, discovery of mechanisms of injury, and defining structural damage in the brain in TBI. Metabolic biomarkers can also be used for the prediction of outcome, monitoring treatment response, in the assessment of or prognosis of post-injury recovery, and potentially in the use of neuroplasticity procedures. Metabolomics can also enhance our understanding of the pathophysiological mechanisms of TBI, both in primary and secondary injury. Thus, this review presents the promising application of metabolomics for the assessment of TBI as a stand-alone platform or in association with proteomics in the clinical setting.


Subject(s)
Biomarkers/metabolism , Brain Injuries, Traumatic/diagnosis , Brain Injuries, Traumatic/metabolism , Metabolomics/methods , Animals , Humans
16.
Crit Care ; 21(1): 97, 2017 Apr 19.
Article in English | MEDLINE | ID: mdl-28424077

ABSTRACT

BACKGROUND: Metabolomics is a tool that has been used for the diagnosis and prognosis of specific diseases. The purpose of this study was to examine if metabolomics could be used as a potential diagnostic and prognostic tool for H1N1 pneumonia. Our hypothesis was that metabolomics can potentially be used early for the diagnosis and prognosis of H1N1 influenza pneumonia. METHODS: 1H nuclear magnetic resonance spectroscopy and gas chromatography-mass spectrometry were used to profile the metabolome in 42 patients with H1N1 pneumonia, 31 ventilated control subjects in the intensive care unit (ICU), and 30 culture-positive plasma samples from patients with bacterial community-acquired pneumonia drawn within the first 24 h of hospital admission for diagnosis and prognosis of disease. RESULTS: We found that plasma-based metabolomics from samples taken within 24 h of hospital admission can be used to discriminate H1N1 pneumonia from bacterial pneumonia and nonsurvivors from survivors of H1N1 pneumonia. Moreover, metabolomics is a highly sensitive and specific tool for the 90-day prognosis of mortality in H1N1 pneumonia. CONCLUSIONS: This study demonstrates that H1N1 pneumonia can create a quite different plasma metabolic profile from bacterial culture-positive pneumonia and ventilated control subjects in the ICU on the basis of plasma samples taken within 24 h of hospital/ICU admission, early in the course of disease.


Subject(s)
Influenza, Human/diagnosis , Metabolomics/methods , Plasma/metabolism , APACHE , Adult , Aged , Female , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/pathogenicity , Intensive Care Units , Magnetic Resonance Spectroscopy/methods , Male , Middle Aged , Multivariate Analysis , Prognosis , Severity of Illness Index
17.
Med J Islam Repub Iran ; 30: 320, 2016.
Article in English | MEDLINE | ID: mdl-27390690

ABSTRACT

BACKGROUND: Visfatin is an adipocytokine secreted by visceral adipose tissue. It has been shown that adipocytokines may contribute to the induction of carcinogens and progression of tumors. Previously, we found a significant increase in the visfatin serum level in colorectal cancer patients. Herein, we investigated if this cytokine increases in patients with colorectal adenoma as a precursor of colorectal cancer. METHODS: In this case-control analytic study, a total of 34 patients diagnosed with colorectal adenoma and 35 disease-free controls were included. Adenomas were also categorized based on their location within the colon. Visfatin serum levels were measured in all cases and controls using enzyme- linked immunosorbent assay kits. In order to compare visfatin levels between groups a twotailed t-test was considered. Pearson correlation was also used to assess the relationship between visfatin levels and other measured variables. RESULTS: Patients included 18 male (53%) and 16 female (47%) with a mean±SD age of 48.3±10.96 years and controls were 18 male (51%) and 17 female (49%) with a mean±SD age of 51.6±12.52 years. There were no significant difference in terms of the visfatin level between the two groups (6.7±3.01 ng/ml for patients and 6.8±2.49 ng/ml for controls, p>0.05). Except for a significant correlation between the BMI and visfatin level (p=0.041), no other correlation was detected. We found no significant difference between the levels of visfatin in each location of adenoma comparing the healthy controls (p>0.05 in all comparisons). There was no statistical difference between the locations groups in terms of visfatin level as well (p>0.05). CONCLUSION: Visfatin serum level does not significantly increase in patients with colorectal adenoma. Site of adenoma within the colon or rectum does not seem to play an important role in this regard as well.

18.
Sarcoidosis Vasc Diffuse Lung Dis ; 33(1): 29-38, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-27055833

ABSTRACT

BACKGROUND: There is no known marker to screen patients with sarcoidosis to determine the risk of progression to pulmonary fibrosis. We aimed to identify potential noninvasive biomarkers for early detection of pulmonary fibrosing sarcoidosis. METHODS: A case-control study was performed on African Americans with confirmed sarcoidosis included 31 subjects with pulmonary fibrosis vs. 36 without pulmonary fibrosis. Plasma samples were analyzed by liquid chromatography-mass spectrum. Multivariate statistical analysis models were developed in a training set based on 50 age- and sex-matched samples to identify metabolites involved in the discrimination. Principal component analysis and orthogonal partial least squares-discriminant (OPLS) analysis coupled to the most influential variables were used to derive significant metabolic discriminations. RESULTS: Of the datasets from 171 feature peaks, 14 features including p-coumaroylagmatine and palmitoylcarnitine showed significant differences between fibrosing and non-fibrosing pulmonary sarcoidosis (p = 0.001). OPLS analysis presented clear separation between two groups with an acceptable goodness of fit (R(2) = 0.522) and predictive power (Q(2)=0.322). Discriminating metabolites involved collagen pathway metabolites especially those in the arginine-proline pathway. CONCLUSIONS: Metabolomics can provide a useful tool to detect pulmonary fibrosis in patients with sarcoidosis. Two discriminating metabolites, p-coumaroylagmatine and palmitoylcarnitine may be potential markers for fibrosing pulmonary sarcoidosis.


Subject(s)
Metabolome , Sarcoidosis, Pulmonary/blood , Case-Control Studies , Female , Humans , Male , Metabolomics , Middle Aged , Retrospective Studies , Sarcoidosis, Pulmonary/metabolism
20.
Mitochondrial DNA A DNA Mapp Seq Anal ; 27(3): 1693-6, 2016 05.
Article in English | MEDLINE | ID: mdl-25230702

ABSTRACT

Beta-thalassemia, one of the most common single-gene disorders, is the result of reduced or absent production of ß-globin chains. Patients with ß-thalassemia show weak genotype-phenotype correlations. Mitochondrial DNA polymorphisms are a potential source for different physiological and pathological characteristics and have been found to be associated as genetic modifiers with various pathophysiologies, including cancers and neurodegenerative diseases. A group of 35 patients with ß-thalassemia was investigated for the presence of mtDNA D-loop polymorphisms in comparison with 504 normal controls. We found four mtDNA D-loop polymorphisms at nucleotides 16,069C > T, 16,189T > C, 16,319G > A, and 16,519T > C that showed significant differences between patients and normal subjects. There is no strong proof for the association of these polymorphisms with ß-thalassemia. It is hypothesized that iron overload or its effects on sequestration of calcium or zinc can lead to oxidative stress and ROS production inside the mitochondria. Therefore, possible accompanying of mtDNA polymorphisms with ß-thalassemia disease may complicate the genotype-phenotype correlation and could affect the clinical outcomes in the patients.


Subject(s)
DNA, Mitochondrial/chemistry , DNA, Mitochondrial/genetics , Genetic Association Studies , Genetic Variation , Nucleic Acid Conformation , beta-Thalassemia/genetics , Case-Control Studies , Humans , Mutation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...