Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Morad K. Nakhleh is active.

Publication


Featured researches published by Morad K. Nakhleh.


ACS Nano | 2017

Diagnosis and Classification of 17 Diseases from 1404 Subjects via Pattern Analysis of Exhaled Molecules

Morad K. Nakhleh; Haitham Amal; Raneen Jeries; Yoav Y. Broza; Manal Aboud; Alaa Gharra; Hodaya Ivgi; Salam Khatib; Shifaa Badarneh; Lior Har-Shai; Lea Glass-Marmor; Izabella Lejbkowicz; Ariel Miller; Samih Badarny; Raz Winer; John Finberg; Sylvia Cohen-Kaminsky; Frédéric Perros; David Montani; Barbara Girerd; Gilles Garcia; Gérald Simonneau; Farid Nakhoul; Shira Baram; Raed Salim; Marwan Hakim; Maayan Gruber; Ohad Ronen; Tal Marshak; Ilana Doweck

We report on an artificially intelligent nanoarray based on molecularly modified gold nanoparticles and a random network of single-walled carbon nanotubes for noninvasive diagnosis and classification of a number of diseases from exhaled breath. The performance of this artificially intelligent nanoarray was clinically assessed on breath samples collected from 1404 subjects having one of 17 different disease conditions included in the study or having no evidence of any disease (healthy controls). Blind experiments showed that 86% accuracy could be achieved with the artificially intelligent nanoarray, allowing both detection and discrimination between the different disease conditions examined. Analysis of the artificially intelligent nanoarray also showed that each disease has its own unique breathprint, and that the presence of one disease would not screen out others. Cluster analysis showed a reasonable classification power of diseases from the same categories. The effect of confounding clinical and environmental factors on the performance of the nanoarray did not significantly alter the obtained results. The diagnosis and classification power of the nanoarray was also validated by an independent analytical technique, i.e., gas chromatography linked with mass spectrometry. This analysis found that 13 exhaled chemical species, called volatile organic compounds, are associated with certain diseases, and the composition of this assembly of volatile organic compounds differs from one disease to another. Overall, these findings could contribute to one of the most important criteria for successful health intervention in the modern era, viz. easy-to-use, inexpensive (affordable), and miniaturized tools that could also be used for personalized screening, diagnosis, and follow-up of a number of diseases, which can clearly be extended by further development.


Nanomedicine: Nanotechnology, Biology and Medicine | 2014

Monolayer-capped gold nanoparticles for disease detection from breath

Morad K. Nakhleh; Yoav Y. Broza; Hossam Haick

The recognition of volatile organic compounds in breath samples is a promising approach for noninvasive safe diagnosis of disease. Spectrometry and spectroscopy methods used for breath analysis suffer from suboptimal accuracy, are expensive and are unsuitable for diagnostics. This article presents a concise review on arrays of monolayer-capped gold nanoparticle (GNP) sensors in conjugation with pattern recognition methods for cost-effective, fast and high-throughput point-of-care diagnostic results from exhaled breath samples. The article starts with a general introduction to the rationale and advantages of breath analysis as well as with a presentation of the utility of monolayer-capped GNP sensors in this field. The article continues with a presentation of the main fabrication and operation principles of these GNP sensors and concludes with selected examples regarding their utility in different fields of medicine, particularly in neurology, infectiology, respiratory medicine and oncology.


European Respiratory Journal | 2014

Detecting active pulmonary tuberculosis with a breath test using nanomaterial-based sensors

Morad K. Nakhleh; Raneen Jeries; Alaa Gharra; Anke Binder; Yoav Y. Broza; Mellissa Pascoe; Keertan Dheda; Hossam Haick

To the Editor: Detecting active tuberculosis (TB) remains a major global public health challenge [1]. The tuberculin skin test does not distinguish latent from active TB [2]. The interferon-γ release assays have similar limitations [3]. Acid-fast bacilli staining of sputum has a high false-negative rate (up to 50%) [4]. Nucleic acid amplification tests (NAATs), such as GeneXpert MTB/RIF (Cepheid, Sunnyvale, CA, USA), are accurate but require a good infrastructure and the necessity to obtain a good quality sputum sample, which is often unobtainable in more than a third of HIV-infected persons [5, 6]. Given these unmet needs, we explored the use of a novel, rapid, simple and inexpensive point-of-care test for the diagnosis of TB [7]. The approach is based on the detection of volatile organic compounds (VOCs) that are emitted from infected cells and released in exhaled breath [8, 9]. Using gas chromatography linked with mass spectrometry, researchers have previously reported identification of TB-related VOCs in the exhaled breath, though there has been low accuracy in detection (80–85%) [10]. In this study, we explore the possibility of active TB detection via the analysis of exhaled breath using a novel technology of organically modified nanomaterial-based sensors. Such cross-reactive sensors are highly sensitive to the collective changes in the VOCs spectrum [8]. Therefore, we designed a case–control study, in which breath and sputum sampling was performed in 210 adult participants, after informed consent, at three sites in Cape Town, South Africa, between November 2011 and March 2012. The study population consisted of two main subgroups. The first subgroup included those in …


Nanomedicine: Nanotechnology, Biology and Medicine | 2014

Sensor arrays based on nanoparticles for early detection of kidney injury by breath samples

Morad K. Nakhleh; Haitham Amal; Hoda Awad; Alaa Gharra; Niroz Abu-Saleh; Raneen Jeries; Hossam Haick; Zaid Abassi

UNLABELLED The outcomes of acute kidney injury (AKI) could be severe and even lethal, if not diagnosed in its early stages and treated appropriately. Blood and urine biomarkers, currently in use as indicators for kidney function, are either inaccurate in various cases or not timely. We report on dramatic changes in exhaled breath composition, associated with kidney dysfunction after ischemic insult in rat models. Gas chromatography linked mass spectrometry examination of breath samples indicated significant elevations in the concentration of three exhaled volatile organic compounds, two to six hours after AKI was surgically induced. Relying on these findings, we introduce an array of sensors, based on organic-layer capped gold nanoparticles, sensitive to odor changes. The ability of the array to detect AKI via breath testing was examined and scored a sensitivity of 96%, only one hour after disease induction. FROM THE CLINICAL EDITOR In this study, organic-layer capped gold nanoparticle-based biosensors are used to analyse breath samples in an acute kidney injury model, capitalizing on the observation that specific volatile organic compounds are present in breath samples in that condition. The authors report excellent sensitivity in as little as one hour after acute kidney injury. This method, if commercialized, may replace the current blood and urine sample analysis-based tests with a more convenient, rapid and accurate nanotechnology-based method.


Parkinsonism & Related Disorders | 2015

Distinguishing idiopathic Parkinson's disease from other parkinsonian syndromes by breath test

Morad K. Nakhleh; Samih Badarny; R. Winer; Raneen Jeries; John P. M. Finberg; Hossam Haick

INTRODUCTION Diagnosis of different parkinsonian syndromes is linked with high misdiagnosis rates and various confounding factors. This is particularly problematic in its early stages. With this in mind, the current pilot study aimed to distinguish between Idiopathic Parkinsons Disease (iPD), other Parkinsonian syndromes (non-iPD) and healthy subjects, by a breath test that analyzes the exhaled volatile organic compounds using a highly sensitive nanoarray. METHODS Breath samples of 44 iPD, 16 non-iPD patients and 37 healthy controls were collected. The samples were passed over a nanoarray and the resulting electrical signals were analyzed with discriminant factor analysis as well as by a K-fold cross-validation method, to test the accuracy of the model. RESULTS Comparison of non-iPD with iPD states yielded 88% sensitivity, 88% accuracy, and 88% Receiver Operating Characteristic area under the curve in the training set samples with known identity. The validation set of this comparison scored 81% sensitivity and accuracy and 92% negative predictive value. Comparison between atypical parkinsonism states and healthy subjects scored 94% sensitivity and 85% accuracy in the training set samples with known identity. The validation set of this comparison scored 81% sensitivity and 78% accuracy. The obtained results were not affected by l-Dopa or MAO-B inhibitor treatment. CONCLUSIONS Exhaled breath analysis with nanoarray is a promising approach for a non-invasive, inexpensive, and portable technique for differentiation between different Parkinsonian states. A larger cohort is required in order to establish the clinical usefulness of the method.


Advanced Healthcare Materials | 2016

Programmed Nanoparticles for Tailoring the Detection of Inflammatory Bowel Diseases and Irritable Bowel Syndrome Disease via Breathprint

Amir Karban; Morad K. Nakhleh; John C. Cancilla; Rotem Vishinkin; Tova Rainis; Eduard Koifman; Raneen Jeries; Hodaya Ivgi; José S. Torrecilla; Hossam Haick

Chemical sensors based on programmable molecularly modified gold nanoparticles are tailored for the detection and discrimination between the breathprint of irritable bowel syndrome (IBS) and inflammatory bowel diseases (IBD). The sensors are examined in both lab- and real-world clinical conditions. The results reveal a discriminative power accuracy of 81% between IBD and IBS and 75% between Crohns and Colitis states.


European Respiratory Journal | 2017

Volatolomics of breath as an emerging frontier in pulmonary arterial hypertension

Morad K. Nakhleh; Hossam Haick; Marc Humbert; Sylvia Cohen-Kaminsky

There is accumulating evidence in support of the significant improvement in survival rates and clinical outcomes when pulmonary arterial hypertension (PAH) is diagnosed at early stages. Nevertheless, it remains a major clinical challenge and the outcomes are dependent on invasive right heart catheterisation. Resulting from pathophysiological processes and detectable in exhaled breath, volatile organic compounds (VOCs) have been proposed as noninvasive biomarkers for PAH. Studies have confirmed significant alterations of the exhaled VOCs among PAH patients when compared to controls and/or patients with other respiratory diseases. This suggests exhaled breath analysis as a potential noninvasive medical application in the field of PAH. In this article, we review and discuss the progress made so far in the field of exhaled volatolomics (the omics of VOCs) as a potential noninvasive diagnostics of PAH. In addition, we propose a model including possible biochemical pathways on the level of the remodelled artery, in which specific VOCs could be detectable in exhaled breath during the early phases of PAH. We debate the different analytical approaches used and recommend a diagram including a “bottom–top” strategy, from basic to translational studies, required for promoting the field. Volatile organic compounds: exhaled volatolomics analysis for noninvasive early diagnosis of PAH http://ow.ly/4R6Q306A0zJ


Circulation | 2018

NMDA-Type Glutamate Receptor Activation Promotes Vascular Remodeling and Pulmonary Arterial Hypertension

Sébastien J. Dumas; Gilles Bru-Mercier; Audrey Courboulin; Marceau Quatredeniers; Catherine Rucker-Martin; Fabrice Antigny; Morad K. Nakhleh; Benoit Ranchoux; Elodie Gouadon; Maria-Candida Vinhas; Matthieu Vocelle; Nicolas Raymond; Peter Dorfmüller; Elie Fadel; Frédéric Perros; Marc Humbert; Sylvia Cohen-Kaminsky

Background: Excessive proliferation and apoptosis resistance in pulmonary vascular cells underlie vascular remodeling in pulmonary arterial hypertension (PAH). Specific treatments for PAH exist, mostly targeting endothelial dysfunction, but high pulmonary arterial pressure still causes heart failure and death. Pulmonary vascular remodeling may be driven by metabolic reprogramming of vascular cells to increase glutaminolysis and glutamate production. The N-methyl-D-aspartate receptor (NMDAR), a major neuronal glutamate receptor, is also expressed on vascular cells, but its role in PAH is unknown. Methods: We assessed the status of the glutamate-NMDAR axis in the pulmonary arteries of patients with PAH and controls through mass spectrometry imaging, Western blotting, and immunohistochemistry. We measured the glutamate release from cultured pulmonary vascular cells using enzymatic assays and analyzed NMDAR regulation/phosphorylation through Western blot experiments. The effect of NMDAR blockade on human pulmonary arterial smooth muscle cell proliferation was determined using a BrdU incorporation assay. We assessed the role of NMDARs in vascular remodeling associated to pulmonary hypertension, in both smooth muscle-specific NMDAR knockout mice exposed to chronic hypoxia and the monocrotaline rat model of pulmonary hypertension using NMDAR blockers. Results: We report glutamate accumulation, upregulation of the NMDAR, and NMDAR engagement reflected by increases in GluN1-subunit phosphorylation in the pulmonary arteries of human patients with PAH. Kv channel inhibition and type A-selective endothelin receptor activation amplified calcium-dependent glutamate release from human pulmonary arterial smooth muscle cell, and type A-selective endothelin receptor and platelet-derived growth factor receptor activation led to NMDAR engagement, highlighting crosstalk between the glutamate-NMDAR axis and major PAH-associated pathways. The platelet-derived growth factor-BB-induced proliferation of human pulmonary arterial smooth muscle cells involved NMDAR activation and phosphorylated GluN1 subunit localization to cell-cell contacts, consistent with glutamatergic communication between proliferating human pulmonary arterial smooth muscle cells via NMDARs. Smooth-muscle NMDAR deficiency in mice attenuated the vascular remodeling triggered by chronic hypoxia, highlighting the role of vascular NMDARs in pulmonary hypertension. Pharmacological NMDAR blockade in the monocrotaline rat model of pulmonary hypertension had beneficial effects on cardiac and vascular remodeling, decreasing endothelial dysfunction, cell proliferation, and apoptosis resistance while disrupting the glutamate-NMDAR pathway in pulmonary arteries. Conclusions: These results reveal a dysregulation of the glutamate-NMDAR axis in the pulmonary arteries of patients with PAH and identify vascular NMDARs as targets for antiremodeling treatments in PAH.


ACS Chemical Neuroscience | 2018

Sensor Array for Detection of Early Stage Parkinson’s Disease before Medication

John P. M. Finberg; Miguel Schwartz; Raneen Jeries; Samih Badarny; Morad K. Nakhleh; Enas Abu Daoud; Yelena Ayubkhanov; Manal Aboud-Hawa; Yoav Y. Broza; Hossam Haick

Early diagnosis of Parkinsons disease (PD) is important because it affects the choice of therapy and is subject to a relatively high degree of error. In addition, early detection of PD can potentially enable the start of neuroprotective therapy before extensive loss of dopaminergic neurons of the substantia nigra occurs. However, until now, studies for early detection of PD using volatile biomarkers sampled only treated and medicated patients. Therefore, there is a great need to evaluate untreated patients for establishing a real world screening and diagnostic technology. Here we describe for the first time a clinical trial to distinguish between de novo PD and control subjects using an electronic system for detection of volatile molecules in exhaled breath (sensor array). We further determine for the first time the association to other common tests for PD diagnostics as smell, ultrasound, and nonmotor symptoms. The test group consisted of 29 PD patients after initial diagnosis by an experienced neurologist, compared with 19 control subjects of similar age. The sensitivity, specificity, and accuracy values of the sensor array to detect PD from controls were 79%, 84%, and 81% respectively, in comparison with midbrain ultrasonography (93%, 90%, 92%) and smell detection (62%, 89%, 73%). The results confirm previous data showing the potential of sensor arrays to detect PD.


ACS Chemical Neuroscience | 2017

Altered Volatile Organic Compound Profile in Transgenic Rats Bearing A53T Mutation of Human α-Synuclein: Comparison with Dopaminergic and Serotonergic Denervation

John P M Finberg; Yuval Aluf; Yelena Loboda; Morad K. Nakhleh; Raneen Jeries; Manal Abud-Hawa; Salman Zubedat; Avi Avital; Soliman Khatib; Jacob Vaya; Hossam Haick

Early diagnosis of Parkinsons disease (PD) is of great importance due its progressive phenotype. Neuroprotective drugs could potentially slow down disease progression if used at early stages. Previously, we have reported an altered content of volatile organic compounds (VOCs) in the breath of rats following a 50% reduction in striatal dopamine (DA) content induced by 6-hydroxydopamine. We now report on the difference in the breath-print and content of VOCs between rats with mild and severe lesions of DA neurons, serotonergic neuronal lesions, and transgenic (Tg) rats carrying the PD-producing A53T mutation of the SNCA (α-synuclein) gene. The Tg rats had an increased content of 3-octen-1-ol and 4-chloro-3-methyl phenol in blood, while in brain tissue, hexanal, hexanol, and 2,3-octanedione were present in controls but absent in Tg rats. Levels of 1-heptyl-2-methyl cyclopropane were increased in brain tissue of Tg rats. The data confirm the potential of breath analysis for detection of human idiosyncratic as well as autosomal dominant PD.

Collaboration


Dive into the Morad K. Nakhleh's collaboration.

Top Co-Authors

Avatar

Hossam Haick

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Raneen Jeries

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Yoav Y. Broza

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Marc Humbert

Université Paris-Saclay

View shared research outputs
Top Co-Authors

Avatar

Alaa Gharra

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

John P. M. Finberg

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Frédéric Perros

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar

Haitham Amal

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Hodaya Ivgi

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge