1
|
Sala S, Nitschke P, Masuda R, Gray N, Lawler NG, Wood JM, Buckler JN, Berezhnoy G, Bolaños J, Boughton BA, Lonati C, Rössler T, Singh Y, Wilson ID, Lodge S, Morillon AC, Loo RL, Hall D, Whiley L, Evans GB, Grove TL, Almo SC, Harris LD, Holmes E, Merle U, Trautwein C, Nicholson JK, Wist J. Integrative Molecular Structure Elucidation and Construction of an Extended Metabolic Pathway Associated with an Ancient Innate Immune Response in COVID-19 Patients. J Proteome Res 2024; 23:956-970. [PMID: 38310443 PMCID: PMC10913068 DOI: 10.1021/acs.jproteome.3c00654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/01/2023] [Accepted: 12/29/2023] [Indexed: 02/05/2024]
Abstract
We present compelling evidence for the existence of an extended innate viperin-dependent pathway, which provides crucial evidence for an adaptive response to viral agents, such as SARS-CoV-2. We show the in vivo biosynthesis of a family of novel endogenous cytosine metabolites with potential antiviral activities. Two-dimensional nuclear magnetic resonance (NMR) spectroscopy revealed a characteristic spin-system motif, indicating the presence of an extended panel of urinary metabolites during the acute viral replication phase. Mass spectrometry additionally enabled the characterization and quantification of the most abundant serum metabolites, showing the potential diagnostic value of the compounds for viral infections. In total, we unveiled ten nucleoside (cytosine- and uracil-based) analogue structures, eight of which were previously unknown in humans allowing us to propose a new extended viperin pathway for the innate production of antiviral compounds. The molecular structures of the nucleoside analogues and their correlation with an array of serum cytokines, including IFN-α2, IFN-γ, and IL-10, suggest an association with the viperin enzyme contributing to an ancient endogenous innate immune defense mechanism against viral infection.
Collapse
Affiliation(s)
- Samuele Sala
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Philipp Nitschke
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Reika Masuda
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Nicola Gray
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Nathan G. Lawler
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - James M. Wood
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Joshua N. Buckler
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
| | - Georgy Berezhnoy
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Jose Bolaños
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Berin A. Boughton
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Caterina Lonati
- Center
for Preclinical Research, Fondazione IRCCS
Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Titus Rössler
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Yogesh Singh
- Institute
of Medical Genetics and Applied Genomics, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Ian D. Wilson
- Division
of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, Burlington Danes Building, Du Cane Road, London W12 0NN, U.K.
| | - Samantha Lodge
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Aude-Claire Morillon
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Ruey Leng Loo
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Drew Hall
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Luke Whiley
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
| | - Gary B. Evans
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Tyler L. Grove
- Department
of Biochemistry, Albert Einstein College
of Medicine, Bronx, New York 10461, United States
| | - Steven C. Almo
- Department
of Biochemistry, Albert Einstein College
of Medicine, Bronx, New York 10461, United States
| | - Lawrence D. Harris
- Ferrier
Research Institute, Victoria University
of Wellington, Wellington 6012, New Zealand
- The
Maurice Wilkins Centre for Molecular Biodiscovef Wellington, Welry, The University of Auckland, Auckland 1010, New Zealand
| | - Elaine Holmes
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Division
of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, Burlington Danes Building, Du Cane Road, London W12 0NN, U.K.
| | - Uta Merle
- Department
of Internal Medicine IV, University Hospital
Heidelberg, 69120 Heidelberg, Germany
| | - Christoph Trautwein
- Department
of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, University Hospital Tübingen, 72074 Tübingen, Germany
| | - Jeremy K. Nicholson
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Institute
of Global Health Innovation, Faculty of
Medicine, Imperial College London, Level 1, Faculty Building, South Kensington Campus, London SW7 2NA, U.K.
| | - Julien Wist
- The
Australian National Phenome Centre and Computational and Systems Medicine,
Health Futures Institute, Murdoch University, Harry Perkins Building, Perth WA6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
- Faculty of Medicine, Department of Metabolism,
Digestion and Reproduction,
Division of Digestive Diseases at Imperial College, London SW7 2AZ, U.K.
| |
Collapse
|
2
|
Dai T, Yan F, Gong J, Liu S. Anti-heart failure mechanism of saponin extract of black ginseng based on metabolomics. J Pharm Biomed Anal 2023; 236:115738. [PMID: 37742504 DOI: 10.1016/j.jpba.2023.115738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
OBJECTIVE This study aimed to explore the mechanism of total saponin of black ginseng (TSBG) in treating heart failure (HF) in DOX-induced HF model rats. METHODS Rats with HF induced by the intraperitoneal injection of DOX were treated with TSBG (low dose, 30 mg/kg/day; medium dose, 60 mg/kg/day; high dose, 120 mg/kg/day) and shakubar trivalsartan (80 mg/kg/day, positive control) for four weeks. Serum BNP and ANP levels were tested by ELISA, and pathological tissue sections were examined. Serum metabolites were measured using nontargeted metabolomic techniques. The expression of Akt/mTOR autophagy-associated proteins in heart tissue was detected using Western blot, including Beclin1, p62, LCII and LC3I. RESULTS Compared with the model group, rats in the TSBG-H group had a significantly lower heart index (p < 0.05), significantly lower serum levels of BNP (p < 0.01) and ANP (p < 0.01) and significantly fewer cardiac histopathological changes. Metabolomic results showed that TSBG significantly back-regulated 12 metabolites (p < 0.05), including cholesterol, histamine, sphinganine, putrescine, arachidonic acid, 3-sulfinoalanine, hypotaurine, gluconic acid and lysoPC (18:0:0). These metabolite changes were involved in taurine and hypotaurine metabolism, arachidonic acid metabolism, sphingolipid metabolism, etc. The protein expression level of p-Akt/Akt and p-mTOR/mTOR was significantly up-regulated (p < 0.001), whereas that of Beclin1, p62 (p < 0.001) and LCII/LC3I was down-regulated (p < 0.05). CONCLUSION TSBG has an excellent therapeutic effect on DOX-induced HF in rats, probably by regulating the Akt/mTOR autophagy signalling pathway, resulting in the improvement of taurine and hypotaurine metabolism, arachidonic acid metabolism and sphingolipid metabolism, which may provide a reference for elucidating the potential mechanism of action of TSBG against HF.
Collapse
Affiliation(s)
- Tingting Dai
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Fuyuan Yan
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiyu Gong
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Shuying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
3
|
D’Amato M, Campagnoli M, Iadarola P, Bignami PM, Fumagalli M, Chiarelli LR, Stelitano G, Meloni F, Linciano P, Collina S, Pietrocola G, Vertui V, Aliberti A, Fossali T, Viglio S. Could the Oxidation of α1-Antitrypsin Prevent the Binding of Human Neutrophil Elastase in COVID-19 Patients? Int J Mol Sci 2023; 24:13533. [PMID: 37686340 PMCID: PMC10488172 DOI: 10.3390/ijms241713533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Human neutrophil elastase (HNE) is involved in SARS-CoV-2 virulence and plays a pivotal role in lung infection of patients infected by COVID-19. In healthy individuals, HNE activity is balanced by α1-antitrypsin (AAT). This is a 52 kDa glycoprotein, mainly produced and secreted by hepatocytes, that specifically inhibits HNE by blocking its activity through the formation of a stable complex (HNE-AAT) in which the two proteins are covalently bound. The lack of this complex, together with the detection of HNE activity in BALf/plasma samples of COVID-19 patients, leads us to hypothesize that potential functional deficiencies should necessarily be attributed to possible structural modifications of AAT. These could greatly diminish its ability to inhibit neutrophil elastase, thus reducing lung protection. The aim of this work was to explore the oxidation state of AAT in BALf/plasma samples from these patients so as to understand whether the deficient inhibitory activity of AAT was somehow related to possible conformational changes caused by the presence of abnormally oxidized residues.
Collapse
Affiliation(s)
- Maura D’Amato
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy; (M.C.); (G.P.); (S.V.)
| | - Monica Campagnoli
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy; (M.C.); (G.P.); (S.V.)
| | - Paolo Iadarola
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (P.I.); (P.M.B.); (M.F.); (L.R.C.); (G.S.)
| | - Paola Margherita Bignami
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (P.I.); (P.M.B.); (M.F.); (L.R.C.); (G.S.)
| | - Marco Fumagalli
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (P.I.); (P.M.B.); (M.F.); (L.R.C.); (G.S.)
| | - Laurent Roberto Chiarelli
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (P.I.); (P.M.B.); (M.F.); (L.R.C.); (G.S.)
| | - Giovanni Stelitano
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (P.I.); (P.M.B.); (M.F.); (L.R.C.); (G.S.)
| | - Federica Meloni
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, 27100 Pavia, Italy; (F.M.); (V.V.)
- Transplant Unit, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Pasquale Linciano
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (P.L.); (S.C.)
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (P.L.); (S.C.)
| | - Giampiero Pietrocola
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy; (M.C.); (G.P.); (S.V.)
| | - Valentina Vertui
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, 27100 Pavia, Italy; (F.M.); (V.V.)
| | - Anna Aliberti
- Division of Anesthesiology and Intensive Care 1, IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Tommaso Fossali
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, 20157 Milan, Italy;
| | - Simona Viglio
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy; (M.C.); (G.P.); (S.V.)
| |
Collapse
|
4
|
Ciaramelli C, Palmioli A, Brioschi M, Viglio S, D’Amato M, Iadarola P, Tosi S, Zucconi L, Airoldi C. Antarctic Soil Metabolomics: A Pilot Study. Int J Mol Sci 2023; 24:12340. [PMID: 37569716 PMCID: PMC10418359 DOI: 10.3390/ijms241512340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
In Antarctica, ice-free areas can be found along the coast, on mountain peaks, and in the McMurdo Dry Valleys, where microorganisms well-adapted to harsh conditions can survive and reproduce. Metabolic analyses can shed light on the survival mechanisms of Antarctic soil communities from both coastal sites, under different plant coverage stages, and inner sites where slow-growing or dormant microorganisms, low water availability, salt accumulation, and a limited number of primary producers make metabolomic profiling difficult. Here, we report, for the first time, an efficient protocol for the extraction and the metabolic profiling of Antarctic soils based on the combination of NMR spectroscopy and mass spectrometry (MS). This approach was set up on samples harvested along different localities of Victoria Land, in continental Antarctica, devoid of or covered by differently developed biological crusts. NMR allowed for the identification of thirty metabolites (mainly sugars, amino acids, and organic acids) and the quantification of just over twenty of them. UPLC-MS analysis identified more than twenty other metabolites, in particular flavonoids, medium- and long-chain fatty acids, benzoic acid derivatives, anthracenes, and quinones. Our results highlighted the complementarity of the two analytical techniques. Moreover, we demonstrated that their combined use represents the "gold standard" for the qualitative and quantitative analysis of little-explored samples, such as those collected from Antarctic soils.
Collapse
Affiliation(s)
- Carlotta Ciaramelli
- Department of Biotechnology and Biosciences, University of Milano—Bicocca, P.zza della Scienza 2, 20126 Milano, Italy; (C.C.); (A.P.); (M.B.)
| | - Alessandro Palmioli
- Department of Biotechnology and Biosciences, University of Milano—Bicocca, P.zza della Scienza 2, 20126 Milano, Italy; (C.C.); (A.P.); (M.B.)
| | - Maura Brioschi
- Department of Biotechnology and Biosciences, University of Milano—Bicocca, P.zza della Scienza 2, 20126 Milano, Italy; (C.C.); (A.P.); (M.B.)
| | - Simona Viglio
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy; (S.V.); (M.D.)
| | - Maura D’Amato
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy; (S.V.); (M.D.)
| | - Paolo Iadarola
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, Via Adolfo Ferrata 9, 27100 Pavia, Italy;
| | - Solveig Tosi
- Department of Earth and Environmental Sciences, University of Pavia, Via S. Epifanio 14, 27100 Pavia, Italy;
| | - Laura Zucconi
- Department of Ecological and Biological Sciences, University of Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milano—Bicocca, P.zza della Scienza 2, 20126 Milano, Italy; (C.C.); (A.P.); (M.B.)
| |
Collapse
|
5
|
Mumcu A. A different approach to the quantification of human seminal plasma metabolites using high-resolution NMR spectroscopy. J Pharm Biomed Anal 2023; 229:115356. [PMID: 37011551 DOI: 10.1016/j.jpba.2023.115356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
In this study, a reliable method was established for the absolute quantification of metabolite concentrations in human seminal plasma using ERETIC2, a quantification tool developed by Bruker based on the PULCON principle. The performance of the ERETIC2 was examined using an AVANCE III HD NMR spectrometer (600 MHz) equipped with a triple inverse 1.7 mm TXI probe in terms of some experimental parameters that may affect the accuracy and precision of the quantitative results. Then, the accuracy, precision, and repeatibility of ERETIC2 were determined using L-asparagine solutions at different concentrations. And it was evaluated by comparing it with the classical internal standard (IS) quantification method. The relative standard deviation (RSD) values for ERETIC2 were calculated in the range of 0.55-1.90% and the minimum recovery value was 99.9%, while the RSD values for the IS method were calculated in the range of 0.88-5.83% and recovery value was minimum 91.0%. Besides, the RSD values of the inter-day precisions for the ERETIC2 and IS methods were obtained to be in the range of 1.25 - 3.03% and 0.97 - 3.46%, respectively. Finally, the concentration values of seminal plasma metabolites were determined using different pulse programs with both methods for samples obtained from normozoospermic control and azoospermic patient groups. The results proved that this quantification method developed using NMR spectroscopy is easy to use in complex sample systems such as biological fluids and is a good alternative to the classical internal standard method in terms of accuracy and sensitivity. In addition, the improvement of the spectral resolution and sensitivity with the microcoil probe technology and the possibility of analyzing with minimum sample quantities has contributed positively to the results of this method.
Collapse
|
6
|
Baciu C, Shin J, Hsin M, Cypel M, Keshavjee S, Liu M. Altered purine metabolism at reperfusion affects clinical outcome in lung transplantation. Thorax 2023; 78:249-257. [PMID: 35450941 DOI: 10.1136/thoraxjnl-2021-217498] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 03/22/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Lung transplantation is an established treatment for patients with end-stage lung disease. However, ischaemia reperfusion injury remains a barrier to achieving better survival outcomes. Here, we aim to investigate the metabolomic and transcriptomic profiles in human lungs before and after reperfusion, to identify mechanisms relevant to clinical outcome. METHODS We analysed 67 paired human lung tissue samples collected from 2008 to 2011, at the end of cold preservation and 2 hours after reperfusion. Gene expression analysis was performed with R. Pathway analysis was conducted with Ingenuity Pathway Analysis. MetaboAnalyst and OmicsNet were used for metabolomics analysis and omics data integration, respectively. Association of identified metabolites with transplant outcome was investigated with Kaplan-Meier estimate and Cox proportional hazard models. RESULTS Activation of energy metabolism and reduced antioxidative biochemicals were found by metabolomics. Upregulation of genes related to cytokines and inflammatory mediators, together with major signalling pathways were revealed by transcriptomics. Purine metabolism was identified as the most significantly enriched pathway at reperfusion, based on integrative analysis of the two omics data sets. Elevated expression of purine nucleoside phosphorylase (PNP) could be attributed to activation of multiple transcriptional pathways. PNP catabolised reactions were evidenced by changes in related metabolites, especially decreased levels of inosine and increased levels of uric acid. Multivariable analyses showed significant association of inosine and uric acid levels with intensive care unit length of stay and ventilation time. CONCLUSION Oxidative stress, especially through purine metabolism pathway, is a major metabolic event during reperfusion and may contribute to the ischaemia reperfusion injury of lung grafts.
Collapse
Affiliation(s)
- Cristina Baciu
- Latner Thoracic Surgical Laboratory, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jason Shin
- Latner Thoracic Surgical Laboratory, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Michael Hsin
- Latner Thoracic Surgical Laboratory, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgical Laboratory, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada.,Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgical Laboratory, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada.,Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgical Laboratory, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada .,Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Markers of Bronchiolitis Obliterans Syndrome after Lung Transplant: Between Old Knowledge and Future Perspective. Biomedicines 2022; 10:biomedicines10123277. [PMID: 36552035 PMCID: PMC9775233 DOI: 10.3390/biomedicines10123277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is the most common form of CLAD and is characterized by airflow limitation and an obstructive spirometric pattern without high-resolution computed tomography (HRCT) evidence of parenchymal opacities. Computed tomography and microCT analysis show abundant small airway obstruction, starting from the fifth generation of airway branching and affecting up to 40-70% of airways. The pathogenesis of BOS remains unclear. It is a multifactorial syndrome that leads to pathological tissue changes and clinical manifestations. Because BOS is associated with the worst long-term survival in LTx patients, many studies are focused on the early identification of BOS. Markers may be useful for diagnosis and for understanding the molecular and immunological mechanisms involved in the onset of BOS. Diagnostic and predictive markers of BOS have also been investigated in various biological materials, such as blood, BAL, lung tissue and extracellular vesicles. The aim of this review was to evaluate the scientific literature on markers of BOS after lung transplant. We performed a systematic review to find all available data on potential prognostic and diagnostic markers of BOS.
Collapse
|
8
|
Ciaramelli C, Palmioli A, Angotti I, Colombo L, De Luigi A, Sala G, Salmona M, Airoldi C. NMR-Driven Identification of Cinnamon Bud and Bark Components With Anti-Aβ Activity. Front Chem 2022; 10:896253. [PMID: 35755250 PMCID: PMC9214034 DOI: 10.3389/fchem.2022.896253] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/26/2022] [Indexed: 11/15/2022] Open
Abstract
The anti-Alzheimer disease (AD) activity reported for an aqueous cinnamon bark extract prompted us to investigate and compare the anti-amyloidogenic properties of cinnamon extracts obtained from both bark and bud, the latter being a very little explored matrix. We prepared the extracts with different procedures (alcoholic, hydroalcoholic, or aqueous extractions). An efficient protocol for the rapid analysis of NMR spectra of cinnamon bud and bark extracts was set up, enabling the automatic identification and quantification of metabolites. Moreover, we exploited preparative reverse-phase (RP) chromatography to prepare fractions enriched in polyphenols, further characterized by UPLC-HR-MS. Then, we combined NMR-based molecular recognition studies, atomic force microscopy, and in vitro biochemical and cellular assays to investigate the anti-amyloidogenic activity of our extracts. Both bud and bark extracts showed a potent anti-amyloidogenic activity. Flavanols, particularly procyanidins, and cinnamaldehydes, are the chemical components of cinnamon hindering Aβ peptide on-pathway aggregation and toxicity in a human neuroblastoma SH-SY5Y cell line. Together with the previously reported ability to hinder tau aggregation and filament formation, these data indicate cinnamon polyphenols as natural products possessing multitarget anti-AD activity. Since cinnamon is a spice increasingly present in the human diet, our results support its use to prepare nutraceuticals useful in preventing AD through an active contrast to the biochemical processes that underlie the onset of this disease. Moreover, the structures of cinnamon components responsible for cinnamon anti-AD activities represent molecular templates for designing and synthesizing new anti-amyloidogenic drugs.
Collapse
Affiliation(s)
- Carlotta Ciaramelli
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy.,Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy
| | - Alessandro Palmioli
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy.,Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy
| | - Irene Angotti
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri"- IRCCS, Milano, Italy
| | - Ada De Luigi
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri"- IRCCS, Milano, Italy
| | - Gessica Sala
- Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy.,School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri"- IRCCS, Milano, Italy
| | - Cristina Airoldi
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy.,Milan Center for Neuroscience (NeuroMI), University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
9
|
D’Amato M, Vertui V, Pandolfi L, Bozzini S, Fossali T, Colombo R, Aliberti A, Fumagalli M, Iadarola P, Didò C, Viglio S, Meloni F. Investigating the Link between Alpha-1 Antitrypsin and Human Neutrophil Elastase in Bronchoalveolar Lavage Fluid of COVID-19 Patients. Curr Issues Mol Biol 2022; 44:2122-2138. [PMID: 35678672 PMCID: PMC9164061 DOI: 10.3390/cimb44050143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 11/26/2022] Open
Abstract
Neutrophils play a pathogenic role in COVID-19 by releasing Neutrophils Extracellular Traps (NETs) or human neutrophil elastase (HNE). Given that HNE is inhibited by α1-antitrypsin (AAT), we aimed to assess the content of HNE, α1-antitrypsin (AAT) and HNE-AAT complexes (the AAT/HNE balance) in 33 bronchoalveolar lavage fluid (BALf) samples from COVID-19 patients. These samples were submitted for Gel-Electrophoresis, Western Blot and ELISA, and proteins (bound to AAT or HNE) were identified by Liquid Chromatography-Mass Spectrometry. NETs' release was analyzed by confocal microscopy. Both HNE and AAT were clearly detectable in BALf at high levels. Contrary to what was previously observed in other settings, the formation of HNE-AAT complex was not detected in COVID-19. Rather, HNE was found to be bound to acute phase proteins, histones and C3. Due to the relevant role of NETs, we assessed the ability of free AAT to bind to histones. While confirming this binding, AAT was not able to inhibit NET formation. In conclusion, despite the finding of a high burden of free and bound HNE, the lack of the HNE-AAT inhibitory complex in COVID-19 BALf demonstrates that AAT is not able to block HNE activity. Furthermore, while binding to histones, AAT does not prevent NET formation nor their noxious activity.
Collapse
Affiliation(s)
- Maura D’Amato
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Valentina Vertui
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, 27100 Pavia, Italy; (V.V.); (L.P.); (C.D.); (F.M.)
| | - Laura Pandolfi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, 27100 Pavia, Italy; (V.V.); (L.P.); (C.D.); (F.M.)
| | - Sara Bozzini
- Laboratory of Respiratory Disease, Cell Biology Section, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Tommaso Fossali
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, 20121 Milan, Italy; (T.F.); (R.C.)
| | - Riccardo Colombo
- Department of Anesthesiology and Intensive Care, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, 20121 Milan, Italy; (T.F.); (R.C.)
| | - Anna Aliberti
- Division of Anesthesiology and Intensive Care 1, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Marco Fumagalli
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (M.F.); (P.I.)
| | - Paolo Iadarola
- Department of Biology and Biotechnologies “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (M.F.); (P.I.)
| | - Camilla Didò
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, 27100 Pavia, Italy; (V.V.); (L.P.); (C.D.); (F.M.)
| | - Simona Viglio
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Federica Meloni
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, 27100 Pavia, Italy; (V.V.); (L.P.); (C.D.); (F.M.)
- Transplant Unit, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
10
|
Watzenboeck ML, Gorki AD, Quattrone F, Gawish R, Schwarz S, Lambers C, Jaksch P, Lakovits K, Zahalka S, Rahimi N, Starkl P, Symmank D, Artner T, Pattaroni C, Fortelny N, Klavins K, Frommlet F, Marsland BJ, Hoetzenecker K, Widder S, Knapp S. Multi-omics profiling predicts allograft function after lung transplantation. Eur Respir J 2022; 59:2003292. [PMID: 34244315 DOI: 10.1183/13993003.03292-2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 06/09/2021] [Indexed: 11/05/2022]
Abstract
RATIONALE Lung transplantation is the ultimate treatment option for patients with end-stage respiratory diseases but bears the highest mortality rate among all solid organ transplantations due to chronic lung allograft dysfunction (CLAD). The mechanisms leading to CLAD remain elusive due to an insufficient understanding of the complex post-transplant adaptation processes. OBJECTIVES To better understand these lung adaptation processes after transplantation and to investigate their association with future changes in allograft function. METHODS We performed an exploratory cohort study of bronchoalveolar lavage samples from 78 lung recipients and donors. We analysed the alveolar microbiome using 16S rRNA sequencing, the cellular composition using flow cytometry, as well as metabolome and lipidome profiling. MEASUREMENTS AND MAIN RESULTS We established distinct temporal dynamics for each of the analysed data sets. Comparing matched donor and recipient samples, we revealed that recipient-specific as well as environmental factors, rather than the donor microbiome, shape the long-term lung microbiome. We further discovered that the abundance of certain bacterial strains correlated with underlying lung diseases even after transplantation. A decline in forced expiratory volume during the first second (FEV1) is a major characteristic of lung allograft dysfunction in transplant recipients. By using a machine learning approach, we could accurately predict future changes in FEV1 from our multi-omics data, whereby microbial profiles showed a particularly high predictive power. CONCLUSION Bronchoalveolar microbiome, cellular composition, metabolome and lipidome show specific temporal dynamics after lung transplantation. The lung microbiome can predict future changes in lung function with high precision.
Collapse
Affiliation(s)
- Martin L Watzenboeck
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- These authors contributed equally
| | - Anna-Dorothea Gorki
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- These authors contributed equally
| | - Federica Quattrone
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- These authors contributed equally
| | - Riem Gawish
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- These authors contributed equally
| | - Stefan Schwarz
- Division of Thoracic Surgery, Dept of Surgery, Medical University of Vienna, Vienna, Austria
- These authors contributed equally
| | - Christopher Lambers
- Division of Thoracic Surgery, Dept of Surgery, Medical University of Vienna, Vienna, Austria
| | - Peter Jaksch
- Division of Thoracic Surgery, Dept of Surgery, Medical University of Vienna, Vienna, Austria
| | - Karin Lakovits
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Sophie Zahalka
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Nina Rahimi
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- Division of Thoracic Surgery, Dept of Surgery, Medical University of Vienna, Vienna, Austria
| | - Philipp Starkl
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Dörte Symmank
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tyler Artner
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Céline Pattaroni
- Dept of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Nikolaus Fortelny
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kristaps Klavins
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Florian Frommlet
- Institute of Medical Statistics, Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | - Konrad Hoetzenecker
- Division of Thoracic Surgery, Dept of Surgery, Medical University of Vienna, Vienna, Austria
| | - Stefanie Widder
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Konrad Lorenz Institute for Evolution and Cognition Research, Klosterneuburg, Austria
- S. Widder and S. Knapp contributed equally to this article as lead authors and supervised the work
| | - Sylvia Knapp
- Research Laboratory of Infection Biology, Dept of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- S. Widder and S. Knapp contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
11
|
Ali S, Nedvědová Š, Badshah G, Afridi MS, Abdullah, Dutra LM, Ali U, Faria SG, Soares FL, Rahman RU, Cançado FA, Aoyanagi MM, Freire LG, Santos AD, Barison A, Oliveira CA. NMR spectroscopy spotlighting immunogenicity induced by COVID-19 vaccination to mitigate future health concerns. CURRENT RESEARCH IN IMMUNOLOGY 2022; 3:199-214. [PMID: 36032416 PMCID: PMC9393187 DOI: 10.1016/j.crimmu.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022] Open
Abstract
In this review, the disease and immunogenicity affected by COVID-19 vaccination at the metabolic level are described considering the use of nuclear magnetic resonance (NMR) spectroscopy for the analysis of different biological samples. Consistently, we explain how different biomarkers can be examined in the saliva, blood plasma/serum, bronchoalveolar-lavage fluid (BALF), semen, feces, urine, cerebrospinal fluid (CSF) and breast milk. For example, the proposed approach for the given samples can allow one to detect molecular biomarkers that can be relevant to disease and/or vaccine interference in a system metabolome. The analysis of the given biomaterials by NMR often produces complex chemical data which can be elucidated by multivariate statistical tools, such as PCA and PLS-DA/OPLS-DA methods. Moreover, this approach may aid to improve strategies that can be helpful in disease control and treatment management in the future. NMR analysis of various bio-samples can explore disease course and vaccine interaction. Immunogenicity and reactogenicity caused by COVID-19 vaccination can be studied by NMR. Vaccine interaction alters metabolic pathway(s) at a certain stage, and this mechanism can be probed at the metabolic level.
Collapse
|
12
|
Dasgupta S, Ghosh N, Choudhury P, Joshi M, Chowdhury SR, Bhattacharyya P, Chaudhury K. NMR metabolomic and microarray-based transcriptomic data integration identifies unique molecular signatures of hypersensitivity pneumonitis. Mol Omics 2021; 18:101-111. [PMID: 34881764 DOI: 10.1039/d1mo00209k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hypersensitivity pneumonitis (HP) is an immune-mediated granulomatous interstitial lung disease (ILD) that results from repeated inhalation of certain antigens. Despite major advances in research, pathophysiology of the disease remains poorly understood. The present study combines metabolomic and transcriptomic data to determine alterations in HP subjects as compared with healthy controls. Metabolic signatures were identified in serum, exhaled breath condensate (EBC) and bronchoalveolar lavage fluid (BALF) of HP patients using proton nuclear magnetic resonance (NMR) metabolomics. The expression of three metabolites, i.e., lactate, pyruvate, and proline, was found to be significantly altered in all three biofluids. The potential of differential diagnosis based on these three metabolites was investigated by including a group of patients with sarcoidosis, which is another type of granulomatous ILD. In addition, differentially expressed transcriptomic fingerprints in blood samples were identified by analyzing a Gene Expression Omnibus (GEO) database. The transcriptomics analysis of these microarray-based data revealed 59 genes to be significantly dysregulated in patients with HP. Over representation analysis of the metabolites and genes of interest was performed using IMPaLA (Integrated Molecular Pathway Level Analysis) version 12. Integrated analysis of serum metabolite signatures and blood gene expression suggests dysregulation of PI3K-AKT signaling and TCA cycle pathways in these patients. This preliminary study is a step towards better understanding of the pathogenesis of HP by identification of differentially expressed metabolites and transcriptomic fingerprints. These molecular signatures may be explored as diagnostic markers for differentiating HP from other lung diseases.
Collapse
Affiliation(s)
- Sanjukta Dasgupta
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Nilanjana Ghosh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Priyanka Choudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Mamata Joshi
- National Facility for High-field NMR, Tata Institute of Fundamental Research, Mumbai, India
| | | | | | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| |
Collapse
|
13
|
Letertre MPM, Giraudeau P, de Tullio P. Nuclear Magnetic Resonance Spectroscopy in Clinical Metabolomics and Personalized Medicine: Current Challenges and Perspectives. Front Mol Biosci 2021; 8:698337. [PMID: 34616770 PMCID: PMC8488110 DOI: 10.3389/fmolb.2021.698337] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Personalized medicine is probably the most promising area being developed in modern medicine. This approach attempts to optimize the therapies and the patient care based on the individual patient characteristics. Its success highly depends on the way the characterization of the disease and its evolution, the patient’s classification, its follow-up and the treatment could be optimized. Thus, personalized medicine must combine innovative tools to measure, integrate and model data. Towards this goal, clinical metabolomics appears as ideally suited to obtain relevant information. Indeed, the metabolomics signature brings crucial insight to stratify patients according to their responses to a pathology and/or a treatment, to provide prognostic and diagnostic biomarkers, and to improve therapeutic outcomes. However, the translation of metabolomics from laboratory studies to clinical practice remains a subsequent challenge. Nuclear magnetic resonance spectroscopy (NMR) and mass spectrometry (MS) are the two key platforms for the measurement of the metabolome. NMR has several advantages and features that are essential in clinical metabolomics. Indeed, NMR spectroscopy is inherently very robust, reproducible, unbiased, quantitative, informative at the structural molecular level, requires little sample preparation and reduced data processing. NMR is also well adapted to the measurement of large cohorts, to multi-sites and to longitudinal studies. This review focus on the potential of NMR in the context of clinical metabolomics and personalized medicine. Starting with the current status of NMR-based metabolomics at the clinical level and highlighting its strengths, weaknesses and challenges, this article also explores how, far from the initial “opposition” or “competition”, NMR and MS have been integrated and have demonstrated a great complementarity, in terms of sample classification and biomarker identification. Finally, a perspective discussion provides insight into the current methodological developments that could significantly raise NMR as a more resolutive, sensitive and accessible tool for clinical applications and point-of-care diagnosis. Thanks to these advances, NMR has a strong potential to join the other analytical tools currently used in clinical settings.
Collapse
Affiliation(s)
| | | | - Pascal de Tullio
- Metabolomics Group, Center for Interdisciplinary Research of Medicine (CIRM), Department of Pharmacy, Université de Liège, Liège, Belgique
| |
Collapse
|
14
|
Amin MS, Wozniak M, Barbaric L, Pickard S, Yerrabelli RS, Christensen A, Coiado OC. Experimental Technologies in the Diagnosis and Treatment of COVID-19 in Patients with Comorbidities. JOURNAL OF HEALTHCARE INFORMATICS RESEARCH 2021; 6:48-71. [PMID: 34541448 PMCID: PMC8442516 DOI: 10.1007/s41666-021-00106-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/05/2021] [Accepted: 09/01/2021] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has impacted the whole world and raised concerns about its effects on different human organ systems. Early detection of COVID-19 may significantly increase the rate of survival; thus, it is critical that the disease is detected early. Emerging technologies have been used to prevent, diagnose, and manage COVID-19 among the populace in the USA and globally. Numerous studies have revealed the growing implementation of novel engineered systems during the intervention at various points of the disease’s pathogenesis, especially as it relates to comorbidities and complications related to cardiovascular and respiratory organ systems. In this review, we provide a succinct, but extensive, review of the pathogenesis of COVID-19, particularly as it relates to angiotensin-converting enzyme 2 (ACE2) as a viral entry point. This is followed by a comprehensive analysis of cardiovascular and respiratory comorbidities of COVID-19 and novel technologies that are used to diagnose and manage hospitalized patients. Continuous cardiorespiratory monitoring systems, novel machine learning algorithms for rapidly triaging patients, various imaging modalities, wearable immunosensors, hotspot tracking systems, and other emerging technologies are reviewed. COVID-19 effects on the immune system, associated inflammatory biomarkers, and innovative therapies are also assessed. Finally, with emphasis on the impact of wearable and non-wearable systems, this review highlights future technologies that could help diagnose, monitor, and mitigate disease progression. Technologies that account for an individual’s health conditions, comorbidities, and even socioeconomic factors can drastically reduce the high mortality seen among many COVID-19 patients, primarily via disease prevention, early detection, and pertinent management.
Collapse
Affiliation(s)
- Md Shahnoor Amin
- Carle Illinois College of Medicine, University of Illinois At Urbana-Champaign, Champaign, IL 61820 USA
| | - Marcin Wozniak
- Beckman Institute for Advanced Science and Technology, Urbana, IL 61801 USA.,Department of Medical Laboratory Diagnostics - Biobank, Medical University of Gdansk, Gdansk, Poland
| | - Lidija Barbaric
- Carle Illinois College of Medicine, University of Illinois At Urbana-Champaign, Champaign, IL 61820 USA
| | - Shanel Pickard
- Carle Illinois College of Medicine, University of Illinois At Urbana-Champaign, Champaign, IL 61820 USA
| | - Rahul S Yerrabelli
- Carle Illinois College of Medicine, University of Illinois At Urbana-Champaign, Champaign, IL 61820 USA
| | - Anton Christensen
- Carle Illinois College of Medicine, University of Illinois At Urbana-Champaign, Champaign, IL 61820 USA
| | - Olivia C Coiado
- Carle Illinois College of Medicine, University of Illinois At Urbana-Champaign, Champaign, IL 61820 USA.,Department of Bioengineering, University of Illinois At Urbana-Champaign, Urbana, IL 61801 USA.,Carle Illinois College of Medicine, 1406 W. Green St, Urbana, IL 61801 USA
| |
Collapse
|
15
|
Manzo G, Gianfanti F, Hind CK, Allison L, Clarke M, Hohenbichler J, Limantoro I, Martin B, Do Carmo Silva P, Ferguson PM, Hodgson-Casson AC, Fleck RA, Sutton JM, Phoenix DA, Mason AJ. Impacts of Metabolism and Organic Acids on Cell Wall Composition and Pseudomonas aeruginosa Susceptibility to Membrane Active Antimicrobials. ACS Infect Dis 2021; 7:2310-2323. [PMID: 34329558 DOI: 10.1021/acsinfecdis.1c00002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Reliable antimicrobial susceptibility testing is essential in informing both clinical antibiotic therapy decisions and the development of new antibiotics. Mammalian cell culture media have been proposed as an alternative to bacteriological media, potentially representing some critical aspects of the infection environment more accurately. Here, we use a combination of NMR metabolomics and electron microscopy to investigate the response of Escherichia coli and Pseudomonas aeruginosa to growth in differing rich media to determine whether and how this determines metabolic strategies, the composition of the cell wall, and consequently susceptibility to membrane active antimicrobials including colistin and tobramycin. The NMR metabolomic approach is first validated by characterizing the expected E. coli acid stress response to fermentation and the accompanying changes in the cell wall composition, when cultured in glucose rich mammalian cell culture media. Glucose is not a major carbon source for P. aeruginosa but is associated with a response to osmotic stress and a modest increase in colistin tolerance. Growth of P. aeruginosa in a range of bacteriological media is supported by consumption of formate, an important electron donor in anaerobic respiration. In mammalian cell culture media, however, the overall metabolic strategy of P. aeruginosa is instead dependent on consumption of glutamine and lactate. Formate doping of mammalian cell culture media does not alter the overall metabolic strategy but is associated with polyamine catabolism, remodelling of both inner and outer membranes, and a modest sensitization of P. aeruginosa PAO1 to colistin. Further, in a panel of P. aeruginosa isolates an increase between 2- and 3-fold in sensitivity to tobramycin is achieved through doping with other organic acids, notably propionate which also similarly enhances the activity of colistin. Organic acids are therefore capable of nonspecifically influencing the potency of membrane active antimicrobials.
Collapse
Affiliation(s)
- Giorgia Manzo
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Federico Gianfanti
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Charlotte K. Hind
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - Leanne Allison
- Centre for Ultrastructural Imaging, Guy’s Campus, King’s College London, London SE1 1UL, United Kingdom
| | - Maria Clarke
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Julia Hohenbichler
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Ilene Limantoro
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Bethany Martin
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - Phoebe Do Carmo Silva
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - Philip M. Ferguson
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Alice C. Hodgson-Casson
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Roland A. Fleck
- Centre for Ultrastructural Imaging, Guy’s Campus, King’s College London, London SE1 1UL, United Kingdom
| | - J. Mark Sutton
- Technology Development Group, National Infection Service, Public Health England, Salisbury SP4 0JG United Kingdom
| | - David A. Phoenix
- School of Applied Science, London South Bank University, 103 Borough Road, London SE1 0AA, United Kingdom
| | - A. James Mason
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| |
Collapse
|
16
|
Mahoney LB, Esther CR, May K, Rosen R. Metabolomic profiling of extraesophageal reflux disease in children. Clin Transl Sci 2021; 14:2025-2033. [PMID: 34058076 PMCID: PMC8504841 DOI: 10.1111/cts.13064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 11/29/2022] Open
Abstract
Although respiratory symptoms in children are often attributed to gastroesophageal reflux disease, establishing a clear diagnosis of extraesophageal reflux disease (EERD) can be challenging, as there are no sensitive or specific EERD biomarkers. The aim of this study was to evaluate the metabolite profile in bronchoalveolar (BAL) fluid from children with suspected EERD and assess the impact of reflux treatment on these metabolites. In this prospective pilot study, we performed nontargeted global metabolomic profiling on BAL fluid from 43 children undergoing testing with bronchoscopy, upper endoscopy, and multichannel intraluminal impedance with pH (pH-MII) for evaluation of chronic respiratory symptoms. Twenty-three (54%) patients had an abnormal pH-MII study. Seventeen (40%) patients were on proton pump inhibitors (PPIs) for testing. Levels of histamine, malate, adenosine 5'-monophosphate, and ascorbate were significantly lower in subjects with abnormal pH-MII studies compared to those normal studies. Furthermore, in children off PPI therapy, those with abnormal pH-MII studies had robust increases in a number of glycerophospholipids within phospholipid metabolic pathways, including derivatives of glycerophosphorylcholine, glycerophosphoglycerol, and glycerophosphoinositol, compared to those with normal pH-MII studies. These findings offer insight into the impact of reflux and PPIs on the lungs and provide a foundation for future studies using targeted metabolomic analysis to identify potential biomarkers of EERD.
Collapse
Affiliation(s)
- Lisa B Mahoney
- Aerodigestive Center, Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Charles R Esther
- Pediatric Pulmonology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Kara May
- Aerodigestive Center, Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rachel Rosen
- Aerodigestive Center, Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Yang S, Abuduwufuer A, Lv W, Bao F, Hu J. [Predictors for the Bronchiolitis Obliterans Syndrome in Lung Transplant Patient]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:496-502. [PMID: 32517455 PMCID: PMC7309540 DOI: 10.3779/j.issn.1009-3419.2020.101.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
肺移植是治疗终末期肺病的有效方法。目前,肺移植术后1年生存率已达到80%,由于闭塞性细支气管炎综合症(bronchiolitis obliterans syndrome, BOS)的发生,5年生存率维持在50%左右。BOS是一个纤维化的过程,最终导致不可逆的气道闭塞。缺血-再灌注损伤、感染、氧化应激以及急性排斥反应等多个因素参与了BOS的发生。研究证实BOS的早期诊断与预后良好相关。因此,寻找灵敏、特异的BOS预测标记物对于提高肺移植患者长期生存具有重要的科学和临床意义。本文就与BOS发生发展相关的免疫调节细胞、分泌性蛋白质、细胞膜蛋白等指标的变化在BOS早期诊断中的作用进行综述。
Collapse
Affiliation(s)
- Sijia Yang
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | - Wang Lv
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Feichao Bao
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian Hu
- The First Affiliated Hospital, Collage of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
18
|
Palmioli A, Alberici D, Ciaramelli C, Airoldi C. Metabolomic profiling of beers: Combining 1H NMR spectroscopy and chemometric approaches to discriminate craft and industrial products. Food Chem 2020; 327:127025. [PMID: 32447135 DOI: 10.1016/j.foodchem.2020.127025] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/10/2020] [Accepted: 05/07/2020] [Indexed: 01/12/2023]
Abstract
The authentication and traceability of craft beers is an important issue for both beer consumers and producers. Reliable analytical methods able to identify and discriminate products are needed to protect the craft brew market against fraud and counterfeit. Here, 1H NMR analysis of 31 beer samples, differing for beer style and brewing method (craft or industrial) was combined with multivariate statistical analysis, following both an untargeted and a targeted approach. NMR-based analysis of beer samples was sped developing a specific protocol enabling the automatic identification and quantification of metabolites in approximately thirty seconds per spectrum. A clear discrimination was achieved by exploiting 1H NMR analysis and multivariate chemometric methods and the targeted approach identified the metabolites responsible for the segregation. Overall, this study reports an analytical approach addressing beer traceability and is the starting point for the development of a standardized protocol for the discrimination of industrial and craft beers.
Collapse
Affiliation(s)
- Alessandro Palmioli
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.zza della Scienza 2, 20126 Milan, Italy.
| | - Diego Alberici
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Carlotta Ciaramelli
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.zza della Scienza 2, 20126 Milan, Italy
| | - Cristina Airoldi
- BioOrgNMR Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, P.zza della Scienza 2, 20126 Milan, Italy.
| |
Collapse
|
19
|
Sharma NS, Vestal G, Wille K, Patel KN, Cheng F, Tipparaju S, Tousif S, Banday MM, Xu X, Wilson L, Nair VS, Morrow C, Hayes D, Seyfang A, Barnes S, Deshane JS, Gaggar A. Differences in airway microbiome and metabolome of single lung transplant recipients. Respir Res 2020; 21:104. [PMID: 32375889 PMCID: PMC7201609 DOI: 10.1186/s12931-020-01367-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022] Open
Abstract
Background Recent studies suggest that alterations in lung microbiome are associated with occurrence of chronic lung diseases and transplant rejection. To investigate the host-microbiome interactions, we characterized the airway microbiome and metabolome of the allograft (transplanted lung) and native lung of single lung transplant recipients. Methods BAL was collected from the allograft and native lungs of SLTs and healthy controls. 16S rRNA microbiome analysis was performed on BAL bacterial pellets and supernatant used for metabolome, cytokines and acetylated proline-glycine-proline (Ac-PGP) measurement by liquid chromatography-high-resolution mass spectrometry. Results In our cohort, the allograft airway microbiome was distinct with a significantly higher bacterial burden and relative abundance of genera Acinetobacter & Pseudomonas. Likewise, the expression of the pro-inflammatory cytokine VEGF and the neutrophil chemoattractant matrikine Ac-PGP in the allograft was significantly higher. Airway metabolome distinguished the native lung from the allografts and an increased concentration of sphingosine-like metabolites that negatively correlated with abundance of bacteria from phyla Proteobacteria. Conclusions Allograft lungs have a distinct microbiome signature, a higher bacterial biomass and an increased Ac-PGP compared to the native lungs in SLTs compared to the native lungs in SLTs. Airway metabolome distinguishes the allografts from native lungs and is associated with distinct microbial communities, suggesting a functional relationship between the local microbiome and metabolome.
Collapse
Affiliation(s)
- Nirmal S Sharma
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida, Tampa, FL, USA. .,Division of Pulmonary, Critical Care & Sleep Medicine, University of South Florida/Tampa General Hospital, University of South Florida, Tampa, FL, USA. .,Division of Cardiothoracic Surgery, University of South Florida, Tampa, FL, USA. .,Brigham and Women's Hospital, Harvard Medical School, Thorn-908 C, 20 Shattuck St, Boston, MA, USA.
| | - Grant Vestal
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida, Tampa, FL, USA
| | - Keith Wille
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kapil N Patel
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida, Tampa, FL, USA.,Division of Pulmonary, Critical Care & Sleep Medicine, University of South Florida/Tampa General Hospital, University of South Florida, Tampa, FL, USA
| | - Feng Cheng
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, USA
| | - Srinivas Tipparaju
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, USA
| | - Sultan Tousif
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Mudassir M Banday
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida, Tampa, FL, USA.,Division of Pulmonary, Critical Care & Sleep Medicine, University of South Florida/Tampa General Hospital, University of South Florida, Tampa, FL, USA
| | - Xin Xu
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Landon Wilson
- Metabolomics Core, Microbiome Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Viswam S Nair
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida, Tampa, FL, USA.,Division of Pulmonary, Critical Care & Sleep Medicine, University of Washington School of Medicine, Washington, USA
| | - Casey Morrow
- Metabolomics Core, Microbiome Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Don Hayes
- Department of Pediatrics, The Ohio State University, Nationwide Children's Hospital, Columbus, OH, USA
| | - Andreas Seyfang
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Stephen Barnes
- Metabolomics Core, Microbiome Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jessy S Deshane
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Amit Gaggar
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.,Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
20
|
A Pilot Study to Investigate the Balance between Proteases and α1-Antitrypsin in Bronchoalveolar Lavage Fluid of Lung Transplant Recipients. High Throughput 2019; 8:ht8010005. [PMID: 30781848 PMCID: PMC6480715 DOI: 10.3390/ht8010005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/30/2019] [Accepted: 02/07/2019] [Indexed: 12/12/2022] Open
Abstract
The neutrophilic component in bronchiolitis obliterans syndrome (BOS, the main form of chronic lung rejection), plays a crucial role in the pathogenesis and maintenance of the disorder. Human Neutrophil Elastase (HNE), a serine protease responsible of elastin degradation whose action is counteracted by α1-antitrypsin (AAT), a serum inhibitor specific for this protease. This work aimed to investigate the relationship between HNE and AAT in bronchoalveolar lavage fluid (BALf) from stable lung transplant recipients and BOS patients to understand whether the imbalance between proteases and inhibitors is relevant to the development of BOS. To reach this goal a multidisciplinary procedure was applied which included: (i) the use of electrophoresis/western blotting coupled with liquid chromatography-mass spectrometric analysis; (ii) the functional evaluation of the residual antiprotease activity, and (iii) a neutrophil count. The results of these experiments demonstrated, for the first time, the presence of the complex between HNE and AAT in a number of BALf samples. The lack of this complex in a few specimens analyzed was investigated in relation to a patient’s lung inflammation. The neutrophil count and the determination of HNE and AAT activities allowed us to speculate that the presence of the complex correlated with the level of lung inflammation.
Collapse
|
21
|
Paris D, Maniscalco M, Motta A. Nuclear magnetic resonance-based metabolomics in respiratory medicine. Eur Respir J 2018; 52:13993003.01107-2018. [PMID: 30115612 DOI: 10.1183/13993003.01107-2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/05/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Debora Paris
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli (Naples), Italy.,Both authors contributed equally
| | - Mauro Maniscalco
- Pulmonary Rehabilitation Unit, ICS Maugeri SpA, IRCCS, Telese Terme (Benevento), Italy.,Both authors contributed equally
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli (Naples), Italy
| |
Collapse
|
22
|
Bazzano M, Laghi L, Zhu C, Magi GE, Serri E, Spaterna A, Tesei B, Laus F. Metabolomics of tracheal wash samples and exhaled breath condensates in healthy horses and horses affected by equine asthma. J Breath Res 2018; 12:046015. [PMID: 30168442 DOI: 10.1088/1752-7163/aade13] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The present work characterized the metabolomic profile of tracheal wash (TW) and exhaled breath condensate (EBC) in healthy horses and horses with respiratory disease. Six asthma-affected horses (group A) and six healthy controls (group H) underwent clinical, endoscopic and cytologic examinations of upper airways to confirm the active phase of asthma. TW and EBC samples were collected from each animal and investigated by proton nuclear magnetic resonance (1H-NMR) metabolomic analysis. A total of ten out of 38 metabolites found in the TW were significantly different between the groups (p < 0.05). Higher concentrations of histamine and oxidant agents, such as glutamate, valine, leucine and isoleucine, as well as lower levels of ascorbate, methylamine, dimethylamine and O-phosphocholine, were found in group A compared to group H. Eight metabolites were found in equine EBC, namely methanol, ethanol, formate, trimethylamine, acetone, acetate, lactate and butanone, previously observed also in human EBC. Despite the fact that this was a pilot study, the results showed that the metabolomic analysis of TW and EBC has the potentiality to serve as a basis for diagnostic tools in horses with asthma.
Collapse
Affiliation(s)
- Marilena Bazzano
- School of Biosciences and Veterinary Medicine, University of Camerino, Macerata, Italy
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Viswan A, Singh C, Rai RK, Azim A, Sinha N, Baronia AK. Metabolomics based predictive biomarker model of ARDS: A systemic measure of clinical hypoxemia. PLoS One 2017; 12:e0187545. [PMID: 29095932 PMCID: PMC5667881 DOI: 10.1371/journal.pone.0187545] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 10/20/2017] [Indexed: 12/25/2022] Open
Abstract
Despite advancements in ventilator technologies, lung supportive and rescue therapies, the outcome and prognostication in acute respiratory distress syndrome (ARDS) remains incremental and ambiguous. Metabolomics is a potential insightful measure to the diagnostic approaches practiced in critical disease settings. In our study patients diagnosed with mild and moderate/severe ARDS clinically governed by hypoxemic P/F ratio between 100-300 but with indistinct molecular phenotype were discriminated employing nuclear magnetic resonance (NMR) based metabolomics of mini bronchoalveolar lavage fluid (mBALF). Resulting biomarker prototype comprising six metabolites was substantiated highlighting ARDS susceptibility/recovery. Both the groups (mild and moderate/severe ARDS) showed distinct biochemical profile based on 83.3% classification by discriminant function analysis and cross validated accuracy of 91% using partial least squares discriminant analysis as major classifier. The predictive performance of narrowed down six metabolites were found analogous with chemometrics. The proposed biomarker model consisting of six metabolites proline, lysine/arginine, taurine, threonine and glutamate were found characteristic of ARDS sub-stages with aberrant metabolism observed mainly in arginine, proline metabolism, lysine synthesis and so forth correlating to diseased metabotype. Thus NMR based metabolomics has provided new insight into ARDS sub-stages and conclusively a precise biomarker model proposed, reflecting underlying metabolic dysfunction aiding prior clinical decision making.
Collapse
Affiliation(s)
- Akhila Viswan
- Centre of Biomedical Research, Lucknow, Uttar Pradesh, India
- Faculty of Engineering and Technology, Dr. A. P. J Abdul Kalam Technical University, Lucknow, Uttar Pradesh, India
| | - Chandan Singh
- Centre of Biomedical Research, Lucknow, Uttar Pradesh, India
| | - Ratan Kumar Rai
- Centre of Biomedical Research, Lucknow, Uttar Pradesh, India
| | - Afzal Azim
- Department of Critical Care Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Neeraj Sinha
- Centre of Biomedical Research, Lucknow, Uttar Pradesh, India
| | - Arvind Kumar Baronia
- Department of Critical Care Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
24
|
Bosso M, Ständker L, Kirchhoff F, Münch J. Exploiting the human peptidome for novel antimicrobial and anticancer agents. Bioorg Med Chem 2017; 26:2719-2726. [PMID: 29122440 DOI: 10.1016/j.bmc.2017.10.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/28/2017] [Indexed: 12/22/2022]
Abstract
Infectious diseases and cancers are leading causes of death and pose major challenges to public health. The human peptidome encompasses millions of compounds that display an enormous structural and functional diversity and represents an excellent source for the discovery of endogenous agents with antimicrobial and/or anticancer activity. Here, we discuss how to exploit the human peptidome for novel antimicrobial and anticancer agents through the generation of peptide libraries from human body fluids and tissues and stepwise purification of bioactive compounds.
Collapse
Affiliation(s)
- Matteo Bosso
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany
| | - Ludger Ständker
- Ulm Peptide Pharmaceuticals, Ulm University, Albert-Einstein-Allee 47, 89081 Ulm, Germany; Core Facility Functional Peptidomics, Ulm University Medical Center, Albert-Einstein-Allee 47, 89081 Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany; Ulm Peptide Pharmaceuticals, Ulm University, Albert-Einstein-Allee 47, 89081 Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstrasse 1, 89081 Ulm, Germany; Ulm Peptide Pharmaceuticals, Ulm University, Albert-Einstein-Allee 47, 89081 Ulm, Germany; Core Facility Functional Peptidomics, Ulm University Medical Center, Albert-Einstein-Allee 47, 89081 Ulm, Germany.
| |
Collapse
|