1
|
Pruekprasert K, Tan M, Ford L, Davies AH, Takats Z, Onida S. Direct Sampling Mass Spectrometry Analysis for the Assessment of Wounds: A Systematic Review. Int Wound J 2025; 22:e70158. [PMID: 40129114 PMCID: PMC11932957 DOI: 10.1111/iwj.70158] [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: 08/16/2024] [Revised: 11/05/2024] [Accepted: 11/21/2024] [Indexed: 03/26/2025] Open
Abstract
Mass spectrometry is increasingly utilised in medicine to identify and quantify small biomarkers for diagnostic and prognostic purposes. Conventional mass spectrometry, however, requires time-consuming sample preparation, hindering its clinical application. Direct sampling mass spectrometry, which allows for direct analysis of patient samples with minimal preparation, offers potential for clinical use. This systematic review examines the utility of direct sampling mass spectrometry for the assessment of external wounds and explores its translational applications in wound care. Out of 2 930 screened abstracts, six studies were included employing various direct sampling mass spectrometry technologies. These studies focused on burn wounds (n = 3), pressure ulcers (n = 2), and acute surgical wounds (n = 1). Both targeted and untargeted molecular profiling methods were used to examine biomarkers related to inflammatory and healing processes, including various proteins, lipid species, and other metabolites. Direct sampling mass spectrometry was found to complement conventional methods such as histology, providing additional insights into the spatial localisation and accumulation of metabolites within wounds. Additionally, imaging techniques equipped with this technology can spatially map wound surfaces and reveal dynamic changes in wounds as they age or progress through different healing processes, with specific metabolite and protein accumulations potentially aiding in prognostication.
Collapse
Affiliation(s)
- Kanin Pruekprasert
- Section of Vascular Surgery, Department of Surgery and CancerImperial College LondonLondonUK
| | - Matthew Tan
- Section of Vascular Surgery, Department of Surgery and CancerImperial College LondonLondonUK
| | - Lauren Ford
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Alun Huw Davies
- Section of Vascular Surgery, Department of Surgery and CancerImperial College LondonLondonUK
| | - Zoltan Takats
- Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Sarah Onida
- Section of Vascular Surgery, Department of Surgery and CancerImperial College LondonLondonUK
| |
Collapse
|
2
|
Heywood WE, Searle J, Collis R, Doykov I, Ashworth M, Sebire N, Bamber A, Gautel M, Eaton S, Coats CJ, Elliott PM, Mills K. A Proof of Principle 2D Spatial Proteome Mapping Analysis Reveals Distinct Regional Differences in the Cardiac Proteome. Life (Basel) 2024; 14:970. [PMID: 39202712 PMCID: PMC11355120 DOI: 10.3390/life14080970] [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: 04/26/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Proteomics studies often explore phenotypic differences between whole organs and systems. Within the heart, more subtle variation exists. To date, differences in the underlying proteome are only described between whole cardiac chambers. This study, using the bovine heart as a model, investigates inter-regional differences and assesses the feasibility of measuring detailed, cross-tissue variance in the cardiac proteome. Using a bovine heart, we created a two-dimensional section through a plane going through two chambers. This plane was further sectioned into 4 × 4 mm cubes and analysed using label-free proteomics. We identified three distinct proteomes. When mapped to the extracted sections, the proteomes corresponded largely to the outer wall of the right ventricle and secondly to the outer wall of the left ventricle, right atrial appendage, tricuspid and mitral valves, modulator band, and parts of the left atrium. The third separate proteome corresponded to the inner walls of the left and right ventricles, septum, and left atrial appendage. Differential protein abundancies indicated differences in energy metabolism between regions. Data analyses of the mitochondrial proteins revealed a variable pattern of abundances of complexes I-V between the proteomes, indicating differences in the bioenergetics of the different cardiac sub-proteomes. Mapping of disease-associated proteins interestingly showed desmoglein-2, for which defects in this protein are known to cause Arrhythmogenic Right Ventricular Dysplasia/Cardiomyopathy, which was present predominantly in the outer wall of the left ventricle. This study highlights that organs can have variable proteomes that do not necessarily correspond to anatomical features.
Collapse
Affiliation(s)
- Wendy E. Heywood
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Jon Searle
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Richard Collis
- Institute of Cardiovascular Science, University College London, Gower Street, London WC1E 6BT, UK; (R.C.); (P.M.E.)
| | - Ivan Doykov
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Michael Ashworth
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK (N.S.)
| | - Neil Sebire
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK (N.S.)
| | - Andrew Bamber
- Histopathology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK (N.S.)
| | - Mathias Gautel
- Randall Division of Cell and Molecular Biophysics, Muscle Signalling Section, King’s College, London WC2E 2LS, UK
| | - Simon Eaton
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| | - Caroline J. Coats
- Institute of Cardiovascular Science, University College London, Gower Street, London WC1E 6BT, UK; (R.C.); (P.M.E.)
| | - Perry M. Elliott
- Institute of Cardiovascular Science, University College London, Gower Street, London WC1E 6BT, UK; (R.C.); (P.M.E.)
- Barts Heart Centre, and the Inherited Cardiovascular Diseases Unit, St Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
| | - Kevin Mills
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (W.E.H.); (I.D.)
| |
Collapse
|
3
|
Wu C, Chen X, Huang W, Yang J, Zhang Z, Liu J, Liu L, Chen Y, Jiang X, Zhang J. Electric fields reverse the differentiation of keratinocyte monolayer by down-regulating E-cadherin through PI3K/AKT/Snail pathway. Heliyon 2024; 10:e33069. [PMID: 39022057 PMCID: PMC11252959 DOI: 10.1016/j.heliyon.2024.e33069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Re-epithelialization is an important step in skin wound healing, referring to the migration, proliferation, and differentiation of keratinocytes around the wound. During this process, the edges of the wound begin to form new epithelial cells, which migrate from the periphery of the wound towards the center, gradually covering the entire wound area. These newly formed epithelial cells proliferate and differentiate, ultimately forming a protective layer over the exposed dermal surface. Wound endogenous electric fields (EFs) are known as the dominant factor to facilitate the epidermal migration to wound center. However, the precise mechanisms by which EFs promote epidermal migration remains elusive. Here, we found that in a model of cultured keratinocyte monolayer in vitro, EFs application reversed the differentiation of cells, as indicated by the reduction of the early differentiation markers K1 and K10. Genetic manipulation confirmed that EFs reversed keratinocyte differentiation through down-regulating the E-cadherin-mediated adhesion. By RNA-sequencing analysis, we screened out Snail as the transcription suppressor of E-cadherin. Snail knockdown abolished the down-regulation of E-cadherin and the reversal of differentiation induced by EFs. KEGG analysis identified PI3K/AKT signaling for Snail induction under EFs. Inhibition of PI3K by LY294002 diminished the EFs-induced AKT activation and Snail augmentation, largely restoring the level of E-cadherin reduced by EFs. Finally, in model of full-thickness skin wounds in pigs, we found that weakening of the wound endogenous EFs by the direction-reversed exogenous EFs resulted in an up-regulation of E-cadherin and earlier differentiation in newly formed epidermis in vivo. Our research suggests that electric fields (EFs) decrease E-cadherin expression by suppressing the PI3K/AKT/Snail pathway, thereby reversing the differentiation of keratinocytes. This discovery provides us with new insights into the role of electric fields in wound healing. EFs intervene in intracellular signaling pathways, inhibiting the expression of E-cadherin, which results in a lower differentiation state of keratinocytes. In this state, keratinocytes exhibit increased migratory capacity, facilitating the migration of epidermal cells and wound reepithelialization.
Collapse
Affiliation(s)
- Chao Wu
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xu Chen
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wanqi Huang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jinrui Yang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Burns and Plastic Surgery Centre, General Hospital of Xinjiang Military Command, Xinjiang, 830000, China
| | - Ze Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jie Liu
- Department of Plastic and Maxillofacial Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Luojia Liu
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ying Chen
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xupin Jiang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiaping Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
4
|
Woodall MNJ, Cujba AM, Worlock KB, Case KM, Masonou T, Yoshida M, Polanski K, Huang N, Lindeboom RGH, Mamanova L, Bolt L, Richardson L, Cakir B, Ellis S, Palor M, Burgoyne T, Pinto A, Moulding D, McHugh TD, Saleh A, Kilich E, Mehta P, O'Callaghan C, Zhou J, Barclay W, De Coppi P, Butler CR, Cortina-Borja M, Vinette H, Roy S, Breuer J, Chambers RC, Heywood WE, Mills K, Hynds RE, Teichmann SA, Meyer KB, Nikolić MZ, Smith CM. Age-specific nasal epithelial responses to SARS-CoV-2 infection. Nat Microbiol 2024; 9:1293-1311. [PMID: 38622380 PMCID: PMC11087271 DOI: 10.1038/s41564-024-01658-1] [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: 03/24/2023] [Accepted: 03/04/2024] [Indexed: 04/17/2024]
Abstract
Children infected with SARS-CoV-2 rarely progress to respiratory failure. However, the risk of mortality in infected people over 85 years of age remains high. Here we investigate differences in the cellular landscape and function of paediatric (<12 years), adult (30-50 years) and older adult (>70 years) ex vivo cultured nasal epithelial cells in response to infection with SARS-CoV-2. We show that cell tropism of SARS-CoV-2, and expression of ACE2 and TMPRSS2 in nasal epithelial cell subtypes, differ between age groups. While ciliated cells are viral replication centres across all age groups, a distinct goblet inflammatory subtype emerges in infected paediatric cultures and shows high expression of interferon-stimulated genes and incomplete viral replication. In contrast, older adult cultures infected with SARS-CoV-2 show a proportional increase in basaloid-like cells, which facilitate viral spread and are associated with altered epithelial repair pathways. We confirm age-specific induction of these cell types by integrating data from in vivo COVID-19 studies and validate that our in vitro model recapitulates early epithelial responses to SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | | | - Kaylee B Worlock
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | | | - Tereza Masonou
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Masahiro Yoshida
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | | | - Ni Huang
- Wellcome Sanger Institute, Cambridge, UK
| | | | | | - Liam Bolt
- Wellcome Sanger Institute, Cambridge, UK
| | | | | | - Samuel Ellis
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Machaela Palor
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Thomas Burgoyne
- UCL Institute of Ophthalmology, University College London, London, UK
- Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Andreia Pinto
- Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Dale Moulding
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Timothy D McHugh
- UCL Centre for Clinical Microbiology, Division of Infection and Immunity, University College London, London, UK
| | - Aarash Saleh
- Royal Free Hospital NHS Foundation Trust, London, UK
| | - Eliz Kilich
- UCL Respiratory, Division of Medicine, University College London, London, UK
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Puja Mehta
- UCL Respiratory, Division of Medicine, University College London, London, UK
- University College London Hospitals NHS Foundation Trust, London, UK
| | | | - Jie Zhou
- Department of Infectious Disease, Imperial College London, London, UK
| | - Wendy Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Paolo De Coppi
- Great Ormond Street UCL Institute of Child Health, London, UK
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Colin R Butler
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, Great Ormond Street UCL Institute of Child Health, University College London, London, UK
| | | | - Heloise Vinette
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Sunando Roy
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Judith Breuer
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Rachel C Chambers
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Wendy E Heywood
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Kevin Mills
- Great Ormond Street UCL Institute of Child Health, London, UK
| | - Robert E Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, Great Ormond Street UCL Institute of Child Health, University College London, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Cambridge, UK.
- Theory of Condensed Matter, Cavendish Laboratory/Dept Physics, University of Cambridge, Cambridge, UK.
| | | | - Marko Z Nikolić
- UCL Respiratory, Division of Medicine, University College London, London, UK.
- University College London Hospitals NHS Foundation Trust, London, UK.
| | - Claire M Smith
- Great Ormond Street UCL Institute of Child Health, London, UK.
| |
Collapse
|
5
|
Traks T, Reemann P, Eskla KL, Ottas A, Jagomäe T, Liira R, Ilves L, Jaks V, Raam L, Abram K, Kingo K. High-throughput proteomic analysis of chronic inflammatory skin diseases: Psoriasis and atopic dermatitis. Exp Dermatol 2024; 33:e15079. [PMID: 38654506 DOI: 10.1111/exd.15079] [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: 08/14/2023] [Revised: 03/13/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Common characteristics in the pathogenesis of psoriasis (PS) and atopic dermatitis (AD) have been presumed, but only a few studies have clearly supported this. The current aim was to find possible similarities and differences in protein expression patterns between these two major chronic inflammatory skin diseases. High-throughput tandem mass spectrometry proteomic analysis was performed using full thickness skin samples from adult PS patients, AD patients and healthy subjects. We detected a combined total of 3045 proteins in the three study groups. According to principal component analysis, there was significant overlap between the proteomic profiles of PS and AD, and both clearly differed from that of healthy skin. The following validation of selected proteins with western blot analysis showed similar tendencies in expression levels and produced statistically significant results. The expression of periostin (POSTN) was consistently high in AD and very low or undetectable in PS (5% FDR corrected p < 0.001), suggesting POSTN as a potential biomarker to distinguish these diseases. Immunohistochemistry further confirmed higher POSTN expression in AD compared to PS skin. Overall, our findings support the concept that these two chronic skin diseases might share considerably more common mechanisms in pathogenesis than has been suspected thus far.
Collapse
Affiliation(s)
- Tanel Traks
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Clinical Research Centre, Tartu University Hospital, University of Tartu, Tartu, Estonia
| | - Paula Reemann
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Kattri-Liis Eskla
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Aigar Ottas
- Clinical Research Centre, Tartu University Hospital, University of Tartu, Tartu, Estonia
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Toomas Jagomäe
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Rasmus Liira
- Institute of Physics, University of Tartu, Tartu, Estonia
| | - Liis Ilves
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Viljar Jaks
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Liisi Raam
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Kristi Abram
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Külli Kingo
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| |
Collapse
|
6
|
Ellis S, Way R, Nel M, Burleigh A, Doykov I, Kembou-Ringert J, Woodall M, Masonou T, Case KM, Ortez AT, McHugh TD, Casal A, McCoy LE, Murdan S, Hynds RE, Gilmour KC, Grandjean L, Cortina-Borja M, Heywood WE, Mills K, Smith CM. Salivary IgA and vimentin differentiate in vitro SARS-CoV-2 infection: A study of 290 convalescent COVID-19 patients. Mucosal Immunol 2024; 17:124-136. [PMID: 38007005 PMCID: PMC11139657 DOI: 10.1016/j.mucimm.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
SARS-CoV-2 initially infects cells in the nasopharynx and oral cavity. The immune system at these mucosal sites plays a crucial role in minimizing viral transmission and infection. To develop new strategies for preventing SARS-CoV-2 infection, this study aimed to identify proteins that protect against viral infection in saliva. We collected 551 saliva samples from 290 healthcare workers who had tested positive for COVID-19, before vaccination, between June and December 2020. The samples were categorized based on their ability to block or enhance infection using in vitro assays. Mass spectrometry and enzyme-linked immunosorbent assay experiments were used to identify and measure the abundance of proteins that specifically bind to SARS-CoV-2 antigens. Immunoglobulin (Ig)A specific to SARS-CoV-2 antigens was detectable in over 83% of the convalescent saliva samples. We found that concentrations of anti-receptor-binding domain IgA >500 pg/µg total protein in saliva correlate with reduced viral infectivity in vitro. However, there is a dissociation between the salivary IgA response to SARS-CoV-2, and systemic IgG titers in convalescent COVID-19 patients. Then, using an innovative technique known as spike-baited mass spectrometry, we identified novel spike-binding proteins in saliva, most notably vimentin, which correlated with increased viral infectivity in vitro and could serve as a therapeutic target against COVID-19.
Collapse
Affiliation(s)
- Samuel Ellis
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rosie Way
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Miranda Nel
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alice Burleigh
- UCL Great Ormond Street Institute of Child Health, London, UK; Centre for Adolescent Rheumatology, University College London, London, UK
| | - Ivan Doykov
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Tereza Masonou
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Timothy D McHugh
- UCL Centre for Clinical Microbiology, Royal Free Hospital, London, UK
| | - Antonio Casal
- Department of Pharmaceutics, UCL School of Pharmacy, London, UK
| | - Laura E McCoy
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | | | - Robert E Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kimberly C Gilmour
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Louis Grandjean
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Wendy E Heywood
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kevin Mills
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire M Smith
- UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
7
|
Gurung S, Timmermand OV, Perocheau D, Gil-Martinez AL, Minnion M, Touramanidou L, Fang S, Messina M, Khalil Y, Spiewak J, Barber AR, Edwards RS, Pinto PL, Finn PF, Cavedon A, Siddiqui S, Rice L, Martini PGV, Ridout D, Heywood W, Hargreaves I, Heales S, Mills PB, Waddington SN, Gissen P, Eaton S, Ryten M, Feelisch M, Frassetto A, Witney TH, Baruteau J. mRNA therapy corrects defective glutathione metabolism and restores ureagenesis in preclinical argininosuccinic aciduria. Sci Transl Med 2024; 16:eadh1334. [PMID: 38198573 PMCID: PMC7615535 DOI: 10.1126/scitranslmed.adh1334] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 10/06/2023] [Indexed: 01/12/2024]
Abstract
The urea cycle enzyme argininosuccinate lyase (ASL) enables the clearance of neurotoxic ammonia and the biosynthesis of arginine. Patients with ASL deficiency present with argininosuccinic aciduria, an inherited metabolic disease with hyperammonemia and a systemic phenotype coinciding with neurocognitive impairment and chronic liver disease. Here, we describe the dysregulation of glutathione biosynthesis and upstream cysteine utilization in ASL-deficient patients and mice using targeted metabolomics and in vivo positron emission tomography (PET) imaging using (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG). Up-regulation of cysteine metabolism contrasted with glutathione depletion and down-regulated antioxidant pathways. To assess hepatic glutathione dysregulation and liver disease, we present [18F]FSPG PET as a noninvasive diagnostic tool to monitor therapeutic response in argininosuccinic aciduria. Human hASL mRNA encapsulated in lipid nanoparticles improved glutathione metabolism and chronic liver disease. In addition, hASL mRNA therapy corrected and rescued the neonatal and adult Asl-deficient mouse phenotypes, respectively, enhancing ureagenesis. These findings provide mechanistic insights in liver glutathione metabolism and support clinical translation of mRNA therapy for argininosuccinic aciduria.
Collapse
Affiliation(s)
- Sonam Gurung
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | | | - Dany Perocheau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ana Luisa Gil-Martinez
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Loukia Touramanidou
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sherry Fang
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Martina Messina
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Youssef Khalil
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Justyna Spiewak
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Abigail R Barber
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Richard S Edwards
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Patricia Lipari Pinto
- Santa Maria's Hospital, Lisbon North University Hospital Center, 1649-028 Lisbon, Portugal
| | | | | | | | - Lisa Rice
- Moderna Inc., Cambridge, MA 02139, USA
| | | | - Deborah Ridout
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Wendy Heywood
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ian Hargreaves
- Pharmacy and Biomolecular Sciences, Liverpool John Moore University, Liverpool L3 5UG, UK
| | - Simon Heales
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Philippa B Mills
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Simon N Waddington
- EGA Institute for Women's Health, University College London, London WC1E 6HX, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of Witswatersrand, Braamfontein, 2000 Johannesburg, South Africa
| | - Paul Gissen
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Mina Ryten
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | | | - Timothy H Witney
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Julien Baruteau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| |
Collapse
|
8
|
Hällqvist J, Pinto RC, Heywood WE, Cordey J, Foulkes AJM, Slattery CF, Leckey CA, Murphy EC, Zetterberg H, Schott JM, Mills K, Paterson RW. A Multiplexed Urinary Biomarker Panel Has Potential for Alzheimer's Disease Diagnosis Using Targeted Proteomics and Machine Learning. Int J Mol Sci 2023; 24:13758. [PMID: 37762058 PMCID: PMC10531486 DOI: 10.3390/ijms241813758] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
As disease-modifying therapies are now available for Alzheimer's disease (AD), accessible, accurate and affordable biomarkers to support diagnosis are urgently needed. We sought to develop a mass spectrometry-based urine test as a high-throughput screening tool for diagnosing AD. We collected urine from a discovery cohort (n = 11) of well-characterised individuals with AD (n = 6) and their asymptomatic, CSF biomarker-negative study partners (n = 5) and used untargeted proteomics for biomarker discovery. Protein biomarkers identified were taken forward to develop a high-throughput, multiplexed and targeted proteomic assay which was tested on an independent cohort (n = 21). The panel of proteins identified are known to be involved in AD pathogenesis. In comparing AD and controls, a panel of proteins including MIEN1, TNFB, VCAM1, REG1B and ABCA7 had a classification accuracy of 86%. These proteins have been previously implicated in AD pathogenesis. This suggests that urine-targeted mass spectrometry has potential utility as a diagnostic screening tool in AD.
Collapse
Affiliation(s)
- Jenny Hällqvist
- Translational Mass Spectrometry Research Group, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.H.); (K.M.)
| | - Rui C. Pinto
- Faculty of Medicine, School of Public Health, Imperial College London, London SW7 2BX, UK
| | - Wendy E. Heywood
- Translational Mass Spectrometry Research Group, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.H.); (K.M.)
| | - Jonjo Cordey
- Translational Mass Spectrometry Research Group, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.H.); (K.M.)
| | | | | | - Claire A. Leckey
- Translational Mass Spectrometry Research Group, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.H.); (K.M.)
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Eimear C. Murphy
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
- UK Dementia Research Institute, UCL, London WC1E 6BT, UK
| | - Jonathan M. Schott
- National Hospital for Neurology and Neurosurgery, Queen Square London, London WC1N 3BG, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; (J.H.); (K.M.)
| | - Ross W. Paterson
- National Hospital for Neurology and Neurosurgery, Queen Square London, London WC1N 3BG, UK
- Darent Valley Hospital, Dartford DA2 8DA, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
| |
Collapse
|
9
|
Sharman K, Patterson NH, Weiss A, Neumann EK, Guiberson ER, Ryan DJ, Gutierrez DB, Spraggins JM, Van de Plas R, Skaar EP, Caprioli RM. Rapid Multivariate Analysis Approach to Explore Differential Spatial Protein Profiles in Tissue. J Proteome Res 2023; 22:1394-1405. [PMID: 35849531 PMCID: PMC9845430 DOI: 10.1021/acs.jproteome.2c00206] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Spatially targeted proteomics analyzes the proteome of specific cell types and functional regions within tissue. While spatial context is often essential to understanding biological processes, interpreting sub-region-specific protein profiles can pose a challenge due to the high-dimensional nature of the data. Here, we develop a multivariate approach for rapid exploration of differential protein profiles acquired from distinct tissue regions and apply it to analyze a published spatially targeted proteomics data set collected from Staphylococcus aureus-infected murine kidney, 4 and 10 days postinfection. The data analysis process rapidly filters high-dimensional proteomic data to reveal relevant differentiating species among hundreds to thousands of measured molecules. We employ principal component analysis (PCA) for dimensionality reduction of protein profiles measured by microliquid extraction surface analysis mass spectrometry. Subsequently, k-means clustering of the PCA-processed data groups samples by chemical similarity. Cluster center interpretation revealed a subset of proteins that differentiate between spatial regions of infection over two time points. These proteins appear involved in tricarboxylic acid metabolomic pathways, calcium-dependent processes, and cytoskeletal organization. Gene ontology analysis further uncovered relationships to tissue damage/repair and calcium-related defense mechanisms. Applying our analysis in infectious disease highlighted differential proteomic changes across abscess regions over time, reflecting the dynamic nature of host-pathogen interactions.
Collapse
Affiliation(s)
- Kavya Sharman
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
- Program in Chemical & Physical Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Nathan Heath Patterson
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Andy Weiss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States
| | - Elizabeth K Neumann
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Emma R Guiberson
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Daniel J Ryan
- Pfizer Inc., Chesterfield, Missouri 63017, United States
| | - Danielle B Gutierrez
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Raf Van de Plas
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Delft Center for Systems and Control, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
10
|
Toma L, Vignali G, Maffioli E, Tambuzzi S, Giaccari R, Mattarozzi M, Nonnis S, Milioli M, Franceschetti L, Paredi G, Negri A, Riccardi B, Cattaneo C, Careri M, Tedeschi G, Bruno S. Mass spectrometry-based proteomic strategy for ecchymotic skin examination in forensic pathology. Sci Rep 2023; 13:6116. [PMID: 37059833 PMCID: PMC10104867 DOI: 10.1038/s41598-023-32520-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023] Open
Abstract
Mass spectrometry (MS)-based proteomics has recently attracted the attention from forensic pathologists. This work is the first report of the development of a shotgun bottom-up proteomic approach based on rapid protein extraction and nano-liquid chromatography/high-resolution mass spectrometry applied to full-thickness human skin for the differential analysis of normal and ecchymotic tissues to identify new biomarkers for bruise characterization and dating. We identified around 2000 proteins from each pooled extract. The method showed excellent precision on independent replicates, with Pearson correlation coefficients always higher than 95%. Glycophorin A, a known biomarker of vital wounds from immunochemical studies, was identified only in ecchymotic tissues, as confirmed by Western blotting analysis. This finding suggests that this protein can be used as a MS-detectable biomarker of wound vitality. By focusing on skin samples from individuals with known wound dating, besides Glycophorin A, other proteins differentially expressed in ecchymotic samples and dependant on wound age were identified, although further analysis on larger datasets are needed to validate these findings. This study paves the way for an in-depth investigation of the potential of MS-based techniques for wound examination in forensic pathology, overcoming the limitations of immunochemical assays.
Collapse
Affiliation(s)
- Lorenzo Toma
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Giulia Vignali
- Institute of Legal Medicine, Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Elisa Maffioli
- Department of Veterinary Medicine and Animal Science, University of Milan, 26900, Lodi, Italy
| | - Stefano Tambuzzi
- Institute of Legal Medicine, Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Roberta Giaccari
- Food and Drug Department, University of Parma, 43124, Parma, Italy
| | - Monica Mattarozzi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy.
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science, University of Milan, 26900, Lodi, Italy.
- CRC Innovation for Well-Being and Environment (I-WE), University of Milan, 20133, Milan, Italy.
| | - Marco Milioli
- Department of Pharmacokinetic, Biochemistry and Metabolism, Global Research and Preclinical Development, Chiesi Farmaceutici Spa, 43122, Parma, Italy
| | - Lorenzo Franceschetti
- Institute of Legal Medicine, Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Gianluca Paredi
- Food and Drug Department, University of Parma, 43124, Parma, Italy
| | - Armando Negri
- Department of Veterinary Medicine and Animal Science, University of Milan, 26900, Lodi, Italy
| | - Benedetta Riccardi
- Department of Pharmacokinetic, Biochemistry and Metabolism, Global Research and Preclinical Development, Chiesi Farmaceutici Spa, 43122, Parma, Italy
| | - Cristina Cattaneo
- Institute of Legal Medicine, Department of Biomedical Sciences for Health, University of Milan, 20133, Milan, Italy
| | - Maria Careri
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science, University of Milan, 26900, Lodi, Italy
- CRC Innovation for Well-Being and Environment (I-WE), University of Milan, 20133, Milan, Italy
| | - Stefano Bruno
- Food and Drug Department, University of Parma, 43124, Parma, Italy
| |
Collapse
|
11
|
Toomey CE, Heywood WE, Evans JR, Lachica J, Pressey SN, Foti SC, Al Shahrani M, D’Sa K, Hargreaves IP, Heales S, Orford M, Troakes C, Attems J, Gelpi E, Palkovits M, Lashley T, Gentleman SM, Revesz T, Mills K, Gandhi S. Mitochondrial dysfunction is a key pathological driver of early stage Parkinson's. Acta Neuropathol Commun 2022; 10:134. [PMID: 36076304 PMCID: PMC9461181 DOI: 10.1186/s40478-022-01424-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The molecular drivers of early sporadic Parkinson's disease (PD) remain unclear, and the presence of widespread end stage pathology in late disease masks the distinction between primary or causal disease-specific events and late secondary consequences in stressed or dying cells. However, early and mid-stage Parkinson's brains (Braak stages 3 and 4) exhibit alpha-synuclein inclusions and neuronal loss along a regional gradient of severity, from unaffected-mild-moderate-severe. Here, we exploited this spatial pathological gradient to investigate the molecular drivers of sporadic PD. METHODS We combined high precision tissue sampling with unbiased large-scale profiling of protein expression across 9 brain regions in Braak stage 3 and 4 PD brains, and controls, and verified these results using targeted proteomic and functional analyses. RESULTS We demonstrate that the spatio-temporal pathology gradient in early-mid PD brains is mirrored by a biochemical gradient of a changing proteome. Importantly, we identify two key events that occur early in the disease, prior to the occurrence of alpha-synuclein inclusions and neuronal loss: (i) a metabolic switch in the utilisation of energy substrates and energy production in the brain, and (ii) perturbation of the mitochondrial redox state. These changes may contribute to the regional vulnerability of developing alpha-synuclein pathology. Later in the disease, mitochondrial function is affected more severely, whilst mitochondrial metabolism, fatty acid oxidation, and mitochondrial respiration are affected across all brain regions. CONCLUSIONS Our study provides an in-depth regional profile of the proteome at different stages of PD, and highlights that mitochondrial dysfunction is detectable prior to neuronal loss, and alpha-synuclein fibril deposition, suggesting that mitochondrial dysfunction is one of the key drivers of early disease.
Collapse
Affiliation(s)
- Christina E. Toomey
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Wendy E. Heywood
- Translational Mass Spectrometry Research Group, Genetic & Genomic Medicine, Institute of Child Health, UCL, London, UK
| | - James R. Evans
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Joanne Lachica
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Sarah N. Pressey
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Sandrine C. Foti
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Mesfer Al Shahrani
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
- College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Karishma D’Sa
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Iain P. Hargreaves
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Simon Heales
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Michael Orford
- National Hospital for Neurology and Neurosurgery & Neurometabolic Unit, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire Troakes
- London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Johannes Attems
- Newcastle Brain Tissue Resource, Institute of Neuroscience and Newcastle University Institute for Ageing, Newcastle upon Tyne, UK
| | - Ellen Gelpi
- Neurological Tissue Bank, University of Barcelona, Barcelona, Spain
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Miklos Palkovits
- Human Brain Tissue Bank, Budapest, Semmelweis University, Budapest, Hungary
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, Genetic & Genomic Medicine, Institute of Child Health, UCL, London, UK
| | - Sonia Gandhi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
12
|
Khalil Y, Carrino S, Lin F, Ferlin A, Lad HV, Mazzacuva F, Falcone S, Rivers N, Banks G, Concas D, Aguilar C, Haynes AR, Blease A, Nicol T, Al-Shawi R, Heywood W, Potter P, Mills K, Gale DP, Clayton PT. Tissue Proteome of 2-Hydroxyacyl-CoA Lyase Deficient Mice Reveals Peroxisome Proliferation and Activation of ω-Oxidation. Int J Mol Sci 2022; 23:ijms23020987. [PMID: 35055171 PMCID: PMC8781152 DOI: 10.3390/ijms23020987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
Peroxisomal fatty acid α-oxidation is an essential pathway for the degradation of β-carbon methylated fatty acids such as phytanic acid. One enzyme in this pathway is 2-hydroxyacyl CoA lyase (HACL1), which is responsible for the cleavage of 2-hydroxyphytanoyl-CoA into pristanal and formyl-CoA. Hacl1 deficient mice do not present with a severe phenotype, unlike mice deficient in other α-oxidation enzymes such as phytanoyl-CoA hydroxylase deficiency (Refsum disease) in which neuropathy and ataxia are present. Tissues from wild-type and Hacl1−/− mice fed a high phytol diet were obtained for proteomic and lipidomic analysis. There was no phenotype observed in these mice. Liver, brain, and kidney tissues underwent trypsin digestion for untargeted proteomic liquid chromatography-mass spectrometry analysis, while liver tissues also underwent fatty acid hydrolysis, extraction, and derivatisation for fatty acid gas chromatography-mass spectrometry analysis. The liver fatty acid profile demonstrated an accumulation of phytanic and 2-hydroxyphytanic acid in the Hacl1−/− liver and significant decrease in heptadecanoic acid. The liver proteome showed a significant decrease in the abundance of Hacl1 and a significant increase in the abundance of proteins involved in PPAR signalling, peroxisome proliferation, and omega oxidation, particularly Cyp4a10 and Cyp4a14. In addition, the pathway associated with arachidonic acid metabolism was affected; Cyp2c55 was upregulated and Cyp4f14 and Cyp2b9 were downregulated. The kidney proteome revealed fewer significantly upregulated peroxisomal proteins and the brain proteome was not significantly different in Hacl1−/− mice. This study demonstrates the powerful insight brought by proteomic and metabolomic profiling of Hacl1−/− mice in better understanding disease mechanism in fatty acid α-oxidation disorders.
Collapse
Affiliation(s)
- Youssef Khalil
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
| | - Sara Carrino
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy
| | - Fujun Lin
- Department of Renal Medicine, University College London, London NW3 2PF, UK; (F.L.); (A.F.); (D.P.G.)
- Department of Nephrology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200082, China
| | - Anna Ferlin
- Department of Renal Medicine, University College London, London NW3 2PF, UK; (F.L.); (A.F.); (D.P.G.)
- Clinical Genetics and Genomics Laboratory, Royal Brompton Hospital, London SW3 6NP, UK
| | - Heena V. Lad
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Francesca Mazzacuva
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
- Department of Bioscience, University of East London, London E15 4LZ, UK
| | - Sara Falcone
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Natalie Rivers
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Gareth Banks
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Danilo Concas
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Carlos Aguilar
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Andrew R. Haynes
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Andy Blease
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Thomas Nicol
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Raya Al-Shawi
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, University College London, London NW3 2PF, UK;
| | - Wendy Heywood
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
| | - Paul Potter
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (H.V.L.); (S.F.); (N.R.); (G.B.); (D.C.); (C.A.); (A.R.H.); (A.B.); (T.N.); (P.P.)
| | - Kevin Mills
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
| | - Daniel P. Gale
- Department of Renal Medicine, University College London, London NW3 2PF, UK; (F.L.); (A.F.); (D.P.G.)
| | - Peter T. Clayton
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (Y.K.); (S.C.); (F.M.); (W.H.); (K.M.)
- Correspondence:
| |
Collapse
|
13
|
Heywood WE, Bliss E, Bahelil F, Cyrus T, Crescente M, Jones T, Iqbal S, Paredes LG, Toner AJ, Del Arroyo AG, O'Toole EA, Mills K, Ackland GL. Proteomic signatures for perioperative oxygen delivery in skin after major elective surgery: mechanistic sub-study of a randomised controlled trial. Br J Anaesth 2021; 127:511-520. [PMID: 34238546 DOI: 10.1016/j.bja.2021.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Maintaining adequate oxygen delivery (DO2) after major surgery is associated with minimising organ dysfunction. Skin is particularly vulnerable to reduced DO2. We tested the hypothesis that reduced perioperative DO2 fuels inflammation in metabolically compromised skin after major surgery. METHODS Participants undergoing elective oesophagectomy were randomised immediately after surgery to standard of care or haemodynamic therapy to achieve their individualised preoperative DO2. Abdominal punch skin biopsies were snap-frozen before and 48 h after surgery. On-line two-dimensional liquid chromatography and ultra-high-definition label-free mass spectrometry was used to characterise the skin proteome. The primary outcome was proteomic changes compared between normal (≥preoperative value before induction of anaesthesia) and low DO2 (<preoperative value before induction of anaesthesia) after surgery. Secondary outcomes were functional enrichment analysis of up/down-regulated proteins (Ingenuity pathway analysis; STRING Protein-Protein Interaction Networks). Immunohistochemistry and immunoblotting confirmed selected proteomic findings in skin biopsies obtained from patients after hepatic resection. RESULTS Paired punch skin biopsies were obtained from 35 participants (mean age: 68 yr; 31% female), of whom 17 underwent oesophagectomy. There were 14/2096 proteins associated with normal (n=10) vs low (n=7) DO2 after oesophagectomy. Failure to maintain preoperative DO2 was associated with upregulation of proteins counteracting oxidative stress. Normal DO2 after surgery was associated with pathways involving leucocyte recruitment and upregulation of an antimicrobial peptidoglycan recognition protein. Immunohistochemistry (n=6 patients) and immunoblots after liver resection (n=12 patients) supported the proteomic findings. CONCLUSIONS Proteomic profiles in serial skin biopsies identified organ-protective mechanisms associated with normal DO2 after major surgery. CLINICAL TRIAL REGISTRATION ISRCTN76894700.
Collapse
Affiliation(s)
- Wendy E Heywood
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
| | - Emily Bliss
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
| | - Fatima Bahelil
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
| | - Trinda Cyrus
- Translational Medicine & Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Marilena Crescente
- Department of Life Sciences, Manchester Metropolitan University Manchester, UK
| | - Timothy Jones
- Translational Medicine & Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Sadaf Iqbal
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, UK
| | - Laura G Paredes
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, UK
| | - Andrew J Toner
- University College London NHS Hospitals Trust, London, UK
| | - Ana G Del Arroyo
- Translational Medicine & Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Edel A O'Toole
- Department of Anesthesia, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, UCL Institute of Child Health, London, UK
| | - Gareth L Ackland
- Translational Medicine & Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
14
|
Nielsen MMK, Aryal E, Safari E, Mojsoska B, Jenssen H, Prabhala BK. Current State of SLC and ABC Transporters in the Skin and Their Relation to Sweat Metabolites and Skin Diseases. Proteomes 2021; 9:proteomes9020023. [PMID: 34065737 PMCID: PMC8163169 DOI: 10.3390/proteomes9020023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/02/2023] Open
Abstract
With a relatively large surface area (2 m2) and 15% of total body mass, the skin forms the largest organ of the human body. The main functions of the skin include regulation of body temperature by insulation or sweating, regulation of the nervous system, regulation of water content, and protection against external injury. To perform these critical functions, the skin encodes genes for transporters responsible for the cellular trafficking of essential nutrients and metabolites to maintain cellular hemostasis. However, the knowledge on the expression, regulation, and function of these transporters is very limited and needs more work to elucidate how these transporters play a role both in disease progression and in healing. Furthermore, SLC and ABC transporters are understudied, and even less studied in skin. There are sparse reports on relation between transporters in skin and sweat metabolites. This mini review focuses on the current state of SLC and ABC transporters in the skin and their relation to sweat metabolites and skin diseases.
Collapse
Affiliation(s)
- Marcus M. K. Nielsen
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
| | - Eva Aryal
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
| | - Elnaz Safari
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran;
| | - Biljana Mojsoska
- Institute of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark; (B.M.); (H.J.)
| | - Håvard Jenssen
- Institute of Science and Environment, Roskilde University, Universitetsvej 1, 4000 Roskilde, Denmark; (B.M.); (H.J.)
| | - Bala Krishna Prabhala
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; (M.M.K.N.); (E.A.)
- Correspondence:
| |
Collapse
|
15
|
Michaud SA, Pětrošová H, Jackson AM, McGuire JC, Sinclair NJ, Ganguly M, Flenniken AM, Nutter LMJ, McKerlie C, Schibli D, Smith D, Borchers CH. Process and Workflow for Preparation of Disparate Mouse Tissues for Proteomic Analysis. J Proteome Res 2020; 20:305-316. [PMID: 33151080 DOI: 10.1021/acs.jproteome.0c00399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigated the effect of homogenization strategy and protein precipitation on downstream protein quantitation using multiple reaction monitoring mass spectrometry (MRM-MS). Our objective was to develop a workflow capable of processing disparate tissue types with high throughput, minimal variability, and maximum purity. Similar abundances of endogenous proteins were measured in nine different mouse tissues regardless of the homogenization method used; however, protein precipitation had strong positive effects on several targets. The best throughput was achieved by lyophilizing tissues to dryness, followed by homogenization via bead-beating without sample buffer. Finally, the effect of tissue perfusion prior to dissection and collection was explored in 20 mouse tissues. MRM-MS showed decreased abundances of blood-related proteins in perfused tissues; however, complete removal was not achieved. Concentrations of nonblood proteins were largely unchanged, although significantly higher variances were observed for proteins from the perfused lung, indicating that perfusion may not be suitable for this organ. We present a simple yet effective tissue processing workflow consisting of harvest of fresh nonperfused tissue, novel lyophilization and homogenization by bead-beating, and protein precipitation. This workflow can be applied to a range of mouse tissues with the advantages of simplicity, minimal manual manipulation of samples, use of commonly available equipment, and high sample quality.
Collapse
Affiliation(s)
- Sarah A Michaud
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada
| | - Helena Pětrošová
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada
| | - Angela M Jackson
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada
| | - Jamie C McGuire
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada
| | - Nicholas J Sinclair
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada
| | - Milan Ganguly
- The Center for Phenogenomics, Toronto M5T 3H7, Ontario, Canada.,The Hospital for Sick Children, Toronto M5G 1X8, Ontario, Canada
| | - Ann M Flenniken
- The Center for Phenogenomics, Toronto M5T 3H7, Ontario, Canada.,Sinai Health Lunenfeld-Tanenbaum Research Institute, Toronto M5G 1X5, Ontario, Canada
| | - Lauryl M J Nutter
- The Center for Phenogenomics, Toronto M5T 3H7, Ontario, Canada.,The Hospital for Sick Children, Toronto M5G 1X8, Ontario, Canada
| | - Colin McKerlie
- The Center for Phenogenomics, Toronto M5T 3H7, Ontario, Canada.,The Hospital for Sick Children, Toronto M5G 1X8, Ontario, Canada
| | - David Schibli
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada
| | - Derek Smith
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada
| | - Christoph H Borchers
- University of Victoria-Genome British Columbia Proteomics Centre, Victoria V8Z 7X8, British Columbia, Canada.,Department of Data Intensive Science and Engineering, Skolkovo Innovation Center, Skolkovo Institute of Science and Technology, Nobel Street, Moscow 143026, Russia.,Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal H3T 1E2, Quebec, Canada.,Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal H3T 1E2, Quebec, Canada
| |
Collapse
|
16
|
Cable CJ, Kaplan N, Getsios S, Thomas PM, Perez White BE. Biotin Identification Proteomics in Three-Dimensional Organotypic Human Skin Cultures. Methods Mol Biol 2020; 2109:185-197. [PMID: 31123999 PMCID: PMC6874900 DOI: 10.1007/7651_2019_239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biotin identification (BioID) proteomics facilitates the unbiased detection of protein interaction neighborhoods in live cells. The BioID technique relies on the covalent biotin alteration of vicinal proteins by a modified bacterial biotin ligase. The biotin ligase is fused to a protein of interest to identify putative protein-protein interactions. Here, we describe the adaptation of this technique for use in three-dimensional epidermal cultures. Due to the covalent biotin modification of proteins, our protocol allows for the complete solubilization of the total cellular protein content in differentiated keratinocytes. Thus, a comprehensive network of potential interactors of a protein of interest can be mapped.
Collapse
Affiliation(s)
- Calvin J Cable
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Nihal Kaplan
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Spiro Getsios
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Paul M Thomas
- Department of Molecular Biosciences, Northwestern University, Chicago, IL, USA
- Northwestern Proteomics, Northwestern University, Chicago, IL, USA
| | - Bethany E Perez White
- Department of Dermatology, Northwestern University, Chicago, IL, USA.
- Skin Disease Research Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
17
|
Wong CW, LeGrand CF, Kinnear BF, Sobota RM, Ramalingam R, Dye DE, Raghunath M, Lane EB, Coombe DR. In Vitro Expansion of Keratinocytes on Human Dermal Fibroblast-Derived Matrix Retains Their Stem-Like Characteristics. Sci Rep 2019; 9:18561. [PMID: 31811191 PMCID: PMC6897920 DOI: 10.1038/s41598-019-54793-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/19/2019] [Indexed: 01/07/2023] Open
Abstract
The long-term expansion of keratinocytes under conditions that avoid xenogeneic components (i.e. animal serum- and feeder cell-free) generally causes diminished proliferation and increased terminal differentiation. Here we present a culture system free of xenogeneic components that retains the self-renewal capacity of primary human keratinocytes. In vivo the extracellular matrix (ECM) of the tissue microenvironment has a major influence on a cell's fate. We used ECM from human dermal fibroblasts, cultured under macromolecular crowding conditions to facilitate matrix deposition and organisation, in a xenogeneic-free keratinocyte expansion protocol. Phospholipase A2 decellularisation produced ECM whose components resembled the core matrix composition of natural dermis by proteome analyses. Keratinocytes proliferated rapidly on these matrices, retained their small size, expressed p63, lacked keratin 10 and rarely expressed keratin 16. The colony forming efficiency of these keratinocytes was enhanced over that of keratinocytes grown on collagen I, indicating that dermal fibroblast-derived matrices maintain the in vitro expansion of keratinocytes in a stem-like state. Keratinocyte sheets formed on such matrices were multi-layered with superior strength and stability compared to the single-layered sheets formed on collagen I. Thus, keratinocytes expanded using our xenogeneic-free protocol retained a stem-like state, but when triggered by confluence and calcium concentration, they stratified to produce epidermal sheets with a potential clinical use.
Collapse
Affiliation(s)
- Chee-Wai Wong
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, 6102, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Science, Curtin University, Bentley, WA, 6102, Australia
| | - Catherine F LeGrand
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, 6102, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Science, Curtin University, Bentley, WA, 6102, Australia
| | - Beverley F Kinnear
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, 6102, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Science, Curtin University, Bentley, WA, 6102, Australia
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, No. 07-48A Proteos, Singapore, 138673, Singapore
| | - Rajkumar Ramalingam
- Skin Research Institute of Singapore and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, 06-06 Immunos, Singapore, 138648, Singapore
| | - Danielle E Dye
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, 6102, Australia
- Curtin Health Innovation Research Institute, Faculty of Health Science, Curtin University, Bentley, WA, 6102, Australia
| | - Michael Raghunath
- Centre for Cell Biology and Tissue Engineering, Competence Centre for Tissue Engineering and Substance Testing (TEDD), Institute for Chemistry and Biotechnology, ZHAW School of Life Science and Facility Management, Zurich University of Applied Science, Winterthur, Switzerland
| | - E Birgitte Lane
- Skin Research Institute of Singapore and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, 06-06 Immunos, Singapore, 138648, Singapore
| | - Deirdre R Coombe
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Bentley, WA, 6102, Australia.
- Curtin Health Innovation Research Institute, Faculty of Health Science, Curtin University, Bentley, WA, 6102, Australia.
- Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia.
| |
Collapse
|
18
|
Manning JE, Cantaert T. Time to Micromanage the Pathogen-Host-Vector Interface: Considerations for Vaccine Development. Vaccines (Basel) 2019; 7:E10. [PMID: 30669682 PMCID: PMC6466432 DOI: 10.3390/vaccines7010010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
The current increase in vector-borne disease worldwide necessitates novel approaches to vaccine development targeted to pathogens delivered by blood-feeding arthropod vectors into the host skin. A concept that is gaining traction in recent years is the contribution of the vector or vector-derived components, like salivary proteins, to host-pathogen interactions. Indeed, the triad of vector-host-pathogen interactions in the skin microenvironment can influence host innate and adaptive responses alike, providing an advantage to the pathogen to establish infection. A better understanding of this "bite site" microenvironment, along with how host and vector local microbiomes immunomodulate responses to pathogens, is required for future vaccines for vector-borne diseases. Microneedle administration of such vaccines may more closely mimic vector deposition of pathogen and saliva into the skin with the added benefit of near painless vaccine delivery. Focusing on the 'micro'⁻from microenvironments to microbiomes to microneedles⁻may yield an improved generation of vector-borne disease vaccines in today's increasingly complex world.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh 12201, Cambodia.
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12201, Cambodia.
| |
Collapse
|
19
|
Ma J, Xu S, Wang X, Zhang J, Wang Y, Liu M, Jin L, Wu M, Qian D, Li X, Zhen Q, Guo H, Gao J, Yang S, Zhang X. Noninvasive analysis of skin proteins in healthy Chinese subjects using an Orbitrap Fusion Tribrid mass spectrometer. Skin Res Technol 2019; 25:424-433. [PMID: 30657212 DOI: 10.1111/srt.12668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/29/2018] [Accepted: 12/08/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Jie Ma
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Shuangjun Xu
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Xiaomeng Wang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Jing Zhang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Yaochi Wang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Mengting Liu
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Ling Jin
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Mingshun Wu
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Danfeng Qian
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Xueying Li
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Qi Zhen
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Huimin Guo
- Center for Biological TechnologyAnhui Agricultural University Hefei China
| | - Jinping Gao
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Sen Yang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| | - Xuejun Zhang
- Institute of Dermatology and Department of DermatologyThe First Affiliated Hospital, Anhui Medical University Hefei China
- The Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education Hefei China
| |
Collapse
|
20
|
Baud A, Little D, Wen TQ, Heywood WE, Gissen P, Mills K. An Optimized Method for the Proteomic Analysis of Low Volumes of Cell Culture Media and the Secretome: The Application and the Demonstration of Altered Protein Expression in iPSC-Derived Neuronal Cell Lines from Parkinson's Disease Patients. J Proteome Res 2019; 18:1198-1207. [PMID: 30562036 DOI: 10.1021/acs.jproteome.8b00831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Traditionally, cell culture medium in iPSC-derived cell work is not the main focus of the research and often is considered as just "food for cells". We demonstrate that by manipulation of the media and optimized methodology, it is possible to use this solution to study the proteins that the cell secretes (the "secretome"). This is particularly useful in the study of iPSC-derived neurons, which require long culture time. We demonstrate that media can be used to model diseases with optimized incubation and sampling times. The ability not to sacrifice cells allows significant cost and research benefits. In this manuscript we describe an optimized method for the analysis of the cell media from iPSC-derived neuronal lines from control and Parkinson's disease patients. We have evaluated the use of standard and supplement B27-free cell media as well as five different sample preparation techniques for proteomic analysis of the cell secretome. Mass spectral analysis of culture media allowed for the identification of >500 proteins, in 500 μL of media, which is less volume than reported previously (20-40 mL). Using shorter incubation times and our optimized methodology, we describe the use of this technique to study and describe potential disease mechanisms in Parkinson's disease.
Collapse
Affiliation(s)
- Anna Baud
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Daniel Little
- MRC Laboratory for Molecular Cell Biology , University College London , London , WC1E 6BT , U.K
| | - Teo Qi Wen
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Wendy E Heywood
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology , University College London , London , WC1E 6BT , U.K
| | - Kevin Mills
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| |
Collapse
|
21
|
Human Skin: Composition, Structure and Visualisation Methods. STUDIES IN MECHANOBIOLOGY, TISSUE ENGINEERING AND BIOMATERIALS 2019. [DOI: 10.1007/978-3-030-13279-8_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
22
|
Murray CE, Gami-Patel P, Gkanatsiou E, Brinkmalm G, Portelius E, Wirths O, Heywood W, Blennow K, Ghiso J, Holton JL, Mills K, Zetterberg H, Revesz T, Lashley T. The presubiculum is preserved from neurodegenerative changes in Alzheimer's disease. Acta Neuropathol Commun 2018; 6:62. [PMID: 30029687 PMCID: PMC6053705 DOI: 10.1186/s40478-018-0563-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022] Open
Abstract
In the majority of affected brain regions the pathological hallmarks of Alzheimer’s disease (AD) are β-amyloid (Aβ) deposits in the form of diffuse and neuritic plaques, tau pathology in the form of neurofibrillary tangles, neuropil threads and plaque-associated abnormal neurites in combination with an inflammatory response. However, the anatomical area of the presubiculum, is characterised by the presence of a single large evenly distributed ‘lake-like’ Aβ deposit with minimal tau deposition or accumulation of inflammatory markers. Post-mortem brain samples from sporadic AD (SAD) and familial AD (FAD) and two hereditary cerebral amyloid diseases, familial British dementia (FBD) and familial Danish dementia (FDD) were used to compare the morphology of the extracellular proteins deposited in the presubiculum compared to the entorhinal cortex. The level of tau pathology and the extent of microglial activation were quantitated in the two brain regions in SAD and FAD. Frozen tissue was used to investigate the Aβ species and proteomic differences between the two regions. Consistent with our previous investigations of FBD and FDD cases we were able to establish that the ‘lake-like’ pre-amyloid deposits of the presubiculum were not a unique feature of AD but they also found two non-Aβ amyloidosis. Comparing the presubiculum to the entorhinal cortex the number of neurofibrillary tangles and tau load were significantly reduced; there was a reduction in microglial activation; there were differences in the Aβ profiles and the investigation of the whole proteome showed significant changes in different protein pathways. In summary, understanding why the presubiculum has a different morphological appearance, biochemical and proteomic makeup compared to surrounding brain regions severely affected by neurodegeneration could lead us to understanding protective mechanisms in neurodegenerative diseases.
Collapse
|
23
|
Palkina NV, Komina AV, Aksenenko MB, Ruksha TG. Biometric Analysis of Melanoma Cells Due to Various Metastasis Origin. VESTNIK DERMATOLOGII I VENEROLOGII 2017. [DOI: 10.25208/0042-4609-2017-93-6-60-66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Objective: biometric analysis of melanoma cells derived from different types of primary or secondary tumors could be necessary for better understanding tumor heterogeneity as that phenomenon would affect significantly the anti-cancer therapy efficacy. Materials and Methods. A comparative analysis of melanoma cells that reflect different stages of tumor progression was accomplished with following parameters tested: intensity of apoptosis, proliferation/metabolic activity, the ratio of the cell cycle phases distribution, chromosomal constitution analysis, invasion, and migration capacity. Results. It was found that melanoma cells derived from visceral metastases characterized by a high proliferative/metabolic potential, migratory ability, and mitotic potential. Melanoma cells which represent earlier stages of carcinogenesis have higher invasive activity and percentage of polyploidy cells, indicating high mutational potential. Both cell lines have no differences in the expression of apoptosis. Conclusion. Melanoma cells derived from metastasis demonstrate various abilities for growth, migration, and invasion depending on metastasis origin. In that context, isolation of pathological cells and tissues, both native and fixed, followed by their individual testing for each patient will have a high demand for both fundamental and clinical medicine for more adequate therapy choice.
Collapse
|