1
|
Wahi K, Freidman N, Wang Q, Devadason M, Quek LE, Pang A, Lloyd L, Larance M, Zanini F, Harvey K, O'Toole S, Guan YF, Holst J. Macropinocytosis mediates resistance to loss of glutamine transport in triple-negative breast cancer. EMBO J 2024; 43:5857-5882. [PMID: 39420093 PMCID: PMC11611898 DOI: 10.1038/s44318-024-00271-6] [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: 02/26/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Triple-negative breast cancer (TNBC) metabolism and cell growth uniquely rely on glutamine uptake by the transporter ASCT2. Despite previous data reporting cell growth inhibition after ASCT2 knockdown, we here show that ASCT2 CRISPR knockout is tolerated by TNBC cell lines. Despite the loss of a glutamine transporter and low rate of glutamine uptake, intracellular glutamine steady-state levels were increased in ASCT2 knockout compared to control cells. Proteomics analysis revealed upregulation of macropinocytosis, reduction in glutamine efflux and increased glutamine synthesis in ASCT2 knockout cells. Deletion of ASCT2 in the TNBC cell line HCC1806 induced a strong increase in macropinocytosis across five ASCT2 knockout clones, compared to a modest increase in ASCT2 knockdown. In contrast, ASCT2 knockout impaired cell proliferation in the non-macropinocytic HCC1569 breast cancer cells. These data identify macropinocytosis as a critical secondary glutamine acquisition pathway in TNBC and a novel resistance mechanism to strategies targeting glutamine uptake alone. Despite this adaptation, TNBC cells continue to rely on glutamine metabolism for their growth, providing a rationale for targeting of more downstream glutamine metabolism components.
Collapse
Affiliation(s)
- Kanu Wahi
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia.
| | - Natasha Freidman
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qian Wang
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
| | - Michelle Devadason
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Lake-Ee Quek
- School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Angel Pang
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia
| | - Larissa Lloyd
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia
| | - Mark Larance
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Fabio Zanini
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Kate Harvey
- Cancer Ecosystems Program, Garvan Institute of Medical Research, UNSW Sydney, Kensington, NSW, Australia
| | - Sandra O'Toole
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Yi Fang Guan
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia
| | - Jeff Holst
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
2
|
Small L, Nguyen TV, Larance M, Saunders DN, Hoy AJ, Schmitz-Peiffer C, Cooney GJ, Brandon AE. Liver proteomics identifies a disconnect between proteins associated with de novo lipogenesis and triglyceride storage. J Lipid Res 2024; 65:100687. [PMID: 39490929 PMCID: PMC11626007 DOI: 10.1016/j.jlr.2024.100687] [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: 08/19/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
De novo lipogenesis (DNL) has been implicated in the development and progression of liver steatosis. Hepatic DNL is strongly influenced by dietary macronutrient composition with diets high in carbohydrate increasing DNL while diets high in fat decrease DNL. The enzymes in the core DNL pathway have been well characterized; however, less is known about other liver proteins that play accessory or regulatory roles. In the current study, we associate measured rates of hepatic DNL and fat content with liver proteomic analysis in mice to identify known and unknown proteins that may have a role in DNL. Male mice were fed either a standard chow diet, a semipurified high starch or high-fat diet. Both semipurified diets resulted in increased body weight, fat mass, and liver triglyceride content compared to chow controls, and hepatic DNL was increased in the high starch and decreased in high fat-fed mice. Proteomic analysis identified novel proteins associated with DNL that are involved in taurine metabolism, suggesting a link between these pathways. There was no relationship between proteins that associated with DNL and those associated with liver triglyceride content. Further analysis identified proteins that are differentially regulated when comparing a nonpurified chow diet to either of the semipurified diets, which provide a set of proteins that are influenced by dietary complexity. Finally, we compared the liver proteome between 4- and 30-week diet-fed mice and found remarkable similarity suggesting that metabolic remodeling of the liver occurs rapidly in response to differing dietary components. Together, these findings highlight novel proteins associated with hepatic DNL independently of liver fat content and suggest rapid liver metabolic remodeling in response to dietary composition changes.
Collapse
Affiliation(s)
- Lewin Small
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney, Sydney, NSW, Australia; Garvan Institute, Sydney, NSW, Australia.
| | | | - Mark Larance
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Darren N Saunders
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrew J Hoy
- School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Carsten Schmitz-Peiffer
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney, Sydney, NSW, Australia; Garvan Institute, Sydney, NSW, Australia
| | - Gregory J Cooney
- Garvan Institute, Sydney, NSW, Australia; School of Medical Sciences, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Amanda E Brandon
- School of Life and Environmental Sciences, Charles Perkins Centre, Faculty of Science, The University of Sydney, Sydney, NSW, Australia; Garvan Institute, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Russo DA, Oliinyk D, Pohnert G, Meier F, Zedler JAZ. EXCRETE workflow enables deep proteomics of the microbial extracellular environment. Commun Biol 2024; 7:1189. [PMID: 39322645 PMCID: PMC11424642 DOI: 10.1038/s42003-024-06910-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
Extracellular proteins play a significant role in shaping microbial communities which, in turn, can impact ecosystem function, human health, and biotechnological processes. Yet, for many ubiquitous microbes, there is limited knowledge regarding the identity and function of secreted proteins. Here, we introduce EXCRETE (enhanced exoproteome characterization by mass spectrometry), a workflow that enables comprehensive description of microbial exoproteomes from minimal starting material. Using cyanobacteria as a case study, we benchmark EXCRETE and show a significant increase over current methods in the identification of extracellular proteins. Subsequently, we show that EXCRETE can be miniaturized and adapted to a 96-well high-throughput format. Application of EXCRETE to cyanobacteria from different habitats (Synechocystis sp. PCC 6803, Synechococcus sp. PCC 11901, and Nostoc punctiforme PCC 73102), and in different cultivation conditions, identified up to 85% of all potentially secreted proteins. Finally, functional analysis reveals that cell envelope maintenance and nutrient acquisition are central functions of the predicted cyanobacterial secretome. Collectively, these findings challenge the general belief that cyanobacteria lack secretory proteins and suggest that multiple functions of the secretome are conserved across freshwater, marine, and terrestrial species.
Collapse
Affiliation(s)
- David A Russo
- Bioorganic Analytics, Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Jena, Germany.
| | - Denys Oliinyk
- Functional Proteomics, Jena University Hospital, Jena, Germany
| | - Georg Pohnert
- Bioorganic Analytics, Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Jena, Germany
| | - Florian Meier
- Functional Proteomics, Jena University Hospital, Jena, Germany
| | - Julie A Z Zedler
- Synthetic Biology of Photosynthetic Organisms, Matthias Schleiden Institute for Genetics, Bioinformatics and Molecular Botany, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
4
|
Reed BL, Tavoian D, Bailey EF, Funk JL, Coletta DK. Inspiratory muscle strength training to improve cardiometabolic health in patients with type 2 diabetes mellitus: protocol for the diabetes inspiratory training clinical trial. Front Endocrinol (Lausanne) 2024; 15:1383131. [PMID: 39345888 PMCID: PMC11427269 DOI: 10.3389/fendo.2024.1383131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/28/2024] [Indexed: 10/01/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex, chronic metabolic disease that carries with it a high prevalence of comorbid conditions, making T2DM one of the leading causes of death in the U.S. Traditional lifestyle interventions (e.g., diet, exercise) can counter some adverse effects of T2DM, however, participation in these activities is low with reasons ranging from physical discomfort to lack of time. Thus, there is a critical need to develop novel management strategies that effectively reduce cardiometabolic disease risk and address barriers to adherence. High-resistance inspiratory muscle strength training (IMST) is a time-efficient and simple breathing exercise that significantly reduces systolic and diastolic BP and improves vascular endothelial function in adults with above-normal blood pressure. Herein we describe the study protocol for a randomized clinical trial to determine the effects of a 6-week IMST regimen on glycemic control and insulin sensitivity in adults with T2DM. Our primary outcome measures include fasting plasma glucose, fasting serum insulin, and insulin resistance utilizing homeostatic model assessment for insulin resistance (HOMA-IR). Secondary outcome measures include resting systolic BP and endothelium-dependent dilation. Further, we will collect plasma for exploratory proteomic analyses. This trial seeks to establish the cardiometabolic effects of 6 weeks of high-resistance IMST in patients with T2DM.
Collapse
Affiliation(s)
- Baylee L Reed
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Dallin Tavoian
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - E Fiona Bailey
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Janet L Funk
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, United States
| | - Dawn K Coletta
- Department of Physiology, University of Arizona, Tucson, AZ, United States
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, United States
- Center for Disparities in Diabetes, Obesity and Metabolism, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
5
|
Itchins M, Liang S, Brown C, Barnes T, Marx G, Chin V, Kao S, Yip PY, Mersiades AJ, Nagrial A, Bray V, Peters G, Parakh S, Garg K, Li BT, McKay M, O'Byrne K, John T, Gill AJ, Molloy MP, Solomon BJ, Pavlakis N. ALKTERNATE: A Pilot Study Alternating Lorlatinib With Crizotinib in ALK-Positive NSCLC With Prior ALK Inhibitor Resistance. JTO Clin Res Rep 2024; 5:100703. [PMID: 39309618 PMCID: PMC11416292 DOI: 10.1016/j.jtocrr.2024.100703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/09/2024] [Accepted: 06/29/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction ALK-positive lung cancers represent a molecularly diverse disease. With drug exposure, driving selection pressure, and resistance pathways, disease relapse will emerge. There is compelling rationale to investigate novel treatment strategies, informed by dynamic circulating tumor DNA (ctDNA) monitoring. Methods The single-arm, pilot study ALKTERNATE investigated fixed alternating cycles of lorlatinib intercalated with crizotinib in individuals resistant to second-generation ALK inhibitors. Dynamic ctDNA explored the correlation with disease response and disease recurrence and defined disease resistance. The primary outcome was time-to-treatment failure, a composite of tolerability, feasibility, and efficacy. Secondary outcomes included standard survival measures, toxicity, pharmacokinetic analysis, and patient-reported outcomes. Tertiary outcomes were proteogenomic analyses of tissue and plasma. Results A total of 15 individuals were enrolled; three encountered primary resistance to lorlatinib induction. There were 12 participants who received alternating therapy, and this approach revealed safety, feasibility, and effectiveness. Patient-reported outcomes were maintained or improved on therapy, and toxicity was consistent with previous reports. The pharmacokinetic measures were similar to the single-arm drug experience. Median time-to-treatment failure was 10 months; overall survival was 23 months. ctDNA profiles indicated inferior survival in those with preexistent TP53 mutations and those without clear or cleared ctDNA at trial induction. The study defined a vastly heterogeneous population with an abundance of ALK coexisting with non-ALK resistance variants. Conclusions ALKTERNATE revealed feasibility with a novel alternating ALK inhibitor strategy in ALK-positive NSCLC. Results support progressing inquiry into this approach and propose a flexible design with drug(s) selected and alternating time frames, informed by real-time plasma profiling. Moving this concept to treatment naive may also optimize impact.
Collapse
Affiliation(s)
- Malinda Itchins
- Royal North Shore Hospital, St Leonards, Australia
- Northern Clinical School, University of Sydney, St Leonards, Australia
- Chris O'Brien Lifehouse, Camperdown, Australia
| | | | - Chris Brown
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | | | - Gavin Marx
- Sydney Adventist Hospital, Wahroonga, Australia
- Australian National University, Sydney, Australia
| | - Venessa Chin
- The Kinghorn Cancer Centre, St Vincent’s Hospital Sydney, Darlinghurst, Australia
- The Garvan Institute of Medical Research, Darlinghurst, Australia
- University of New South Wales, Darlinghurst, Australia
| | - Steven Kao
- Chris O'Brien Lifehouse, Camperdown, Australia
- Sydney Medical School, University of Sydney, Camperdown, Australia
| | - Po Yee Yip
- Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, Australia
- School of Medicine, Western Sydney University, Campbelltown, Australia
| | - Antony J. Mersiades
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, Australia
- Northern Beaches Hospital, Frenchs Forest, Australia
| | - Adnan Nagrial
- Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, Australia
- Blacktown Hospital, Blacktown, Australia
- Westmead Clinical School, University of Sydney, Westmead, Australia
| | | | - Geoffrey Peters
- Canberra Hospital, Canberra, Australia
- Australian National University, Canberra, Australia
| | - Sagun Parakh
- Olivia Newton-John Cancer Research Institute, Austin Hospital, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Australia
| | | | - Bob T. Li
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew McKay
- Kolling Institute, University of Sydney, St Leonards, Australia
| | | | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, Australia
- University of Melbourne, Melbourne, Australia
| | - Anthony J. Gill
- Royal North Shore Hospital, St Leonards, Australia
- Northern Clinical School, University of Sydney, St Leonards, Australia
| | - Mark P. Molloy
- Northern Clinical School, University of Sydney, St Leonards, Australia
- Kolling Institute, University of Sydney, St Leonards, Australia
| | - Benjamin J. Solomon
- Peter MacCallum Cancer Centre, Melbourne, Australia
- University of Melbourne, Melbourne, Australia
| | - Nick Pavlakis
- Royal North Shore Hospital, St Leonards, Australia
- Northern Clinical School, University of Sydney, St Leonards, Australia
| |
Collapse
|
6
|
Jia X, Crawford JC, Gebregzabher D, Monson EA, Mettelman RC, Wan Y, Ren Y, Chou J, Novak T, McQuilten HA, Clarke M, Bachem A, Foo IJ, Fritzlar S, Carrera Montoya J, Trenerry AM, Nie S, Leeming MG, Nguyen THO, Kedzierski L, Littler DR, Kueh A, Cardamone T, Wong CY, Hensen L, Cabug A, Laguna JG, Agrawal M, Flerlage T, Boyd DF, Van de Velde LA, Habel JR, Loh L, Koay HF, van de Sandt CE, Konstantinov IE, Berzins SP, Flanagan KL, Wakim LM, Herold MJ, Green AM, Smallwood HS, Rossjohn J, Thwaites RS, Chiu C, Scott NE, Mackenzie JM, Bedoui S, Reading PC, Londrigan SL, Helbig KJ, Randolph AG, Thomas PG, Xu J, Wang Z, Chua BY, Kedzierska K. High expression of oleoyl-ACP hydrolase underpins life-threatening respiratory viral diseases. Cell 2024; 187:4586-4604.e20. [PMID: 39137778 DOI: 10.1016/j.cell.2024.07.026] [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: 03/07/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024]
Abstract
Respiratory infections cause significant morbidity and mortality, yet it is unclear why some individuals succumb to severe disease. In patients hospitalized with avian A(H7N9) influenza, we investigated early drivers underpinning fatal disease. Transcriptomics strongly linked oleoyl-acyl-carrier-protein (ACP) hydrolase (OLAH), an enzyme mediating fatty acid production, with fatal A(H7N9) early after hospital admission, persisting until death. Recovered patients had low OLAH expression throughout hospitalization. High OLAH levels were also detected in patients hospitalized with life-threatening seasonal influenza, COVID-19, respiratory syncytial virus (RSV), and multisystem inflammatory syndrome in children (MIS-C) but not during mild disease. In olah-/- mice, lethal influenza infection led to survival and mild disease as well as reduced lung viral loads, tissue damage, infection-driven pulmonary cell infiltration, and inflammation. This was underpinned by differential lipid droplet dynamics as well as reduced viral replication and virus-induced inflammation in macrophages. Supplementation of oleic acid, the main product of OLAH, increased influenza replication in macrophages and their inflammatory potential. Our findings define how the expression of OLAH drives life-threatening viral disease.
Collapse
Affiliation(s)
- Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jeremy Chase Crawford
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Deborah Gebregzabher
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ebony A Monson
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robert C Mettelman
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yanmin Wan
- Shanghai Public Health Clinical Centre and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Yanqin Ren
- Shanghai Public Health Clinical Centre, Fudan University, Shanghai 201508, China
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tanya Novak
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital and Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Michele Clarke
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Isabelle J Foo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Julio Carrera Montoya
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Alice M Trenerry
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC 3052, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Dene R Littler
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Andrew Kueh
- Walter Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Tina Cardamone
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Chinn Yi Wong
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Aira Cabug
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jaime Gómez Laguna
- Department of Anatomy and Comparative Pathology and Toxicology, Pathology and Immunology Group, University of Córdoba, International Excellence Agrifood Campus "CeiA3", 14014 Córdoba, Spain
| | - Mona Agrawal
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David F Boyd
- Department of Molecular, Cell & Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Lee-Ann Van de Velde
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Carolien E van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Igor E Konstantinov
- Department of Cardiothoracic Surgery, Royal Children's Hospital, University of Melbourne, Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, Parkville, VIC 3052, Australia
| | - Stuart P Berzins
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3353, Australia
| | - Katie L Flanagan
- School of Health Sciences and School of Medicine, University of Tasmania, Launceston, TAS 7248, Australia; School of Health and Biomedical Science, RMIT University, Bundoora, VIC 3083, Australia; Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Marco J Herold
- Walter Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Amanda M Green
- Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Institute of Infection and Immunity, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Karla J Helbig
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
| | - Adrienne G Randolph
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital and Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Infectious Diseases Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA
| | - Jianqing Xu
- Shanghai Public Health Clinical Centre and Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Fudan University, Shanghai 201508, China
| | - Zhongfang Wang
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China.
| | - Brendon Y Chua
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Center for Influenza Disease and Emergence Response (CIDER), Athens, GA, USA.
| |
Collapse
|
7
|
Diaz-Vegas A, Cooke KC, Cutler HB, Yau B, Masson SWC, Harney D, Fuller OK, Potter M, Madsen S, Craw NR, Zhang Y, Moreno CL, Kebede MA, Neely GG, Stöckli J, Burchfield JG, James DE. Deletion of miPEP in adipocytes protects against obesity and insulin resistance by boosting muscle metabolism. Mol Metab 2024; 86:101983. [PMID: 38960128 PMCID: PMC11292358 DOI: 10.1016/j.molmet.2024.101983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
Mitochondria facilitate thousands of biochemical reactions, covering a broad spectrum of anabolic and catabolic processes. Here we demonstrate that the adipocyte mitochondrial proteome is markedly altered across multiple models of insulin resistance and reveal a consistent decrease in the level of the mitochondrial processing peptidase miPEP. OBJECTIVE To determine the role of miPEP in insulin resistance. METHODS To experimentally test this observation, we generated adipocyte-specific miPEP knockout mice to interrogate its role in the aetiology of insulin resistance. RESULTS We observed a strong phenotype characterised by enhanced insulin sensitivity and reduced adiposity, despite normal food intake and physical activity. Strikingly, these phenotypes vanished when mice were housed at thermoneutrality, suggesting that metabolic protection conferred by miPEP deletion hinges upon a thermoregulatory process. Tissue specific analysis of miPEP deficient mice revealed an increment in muscle metabolism, and upregulation of the protein FBP2 that is involved in ATP hydrolysis in the gluconeogenic pathway. CONCLUSION These findings suggest that miPEP deletion initiates a compensatory increase in skeletal muscle metabolism acting as a protective mechanism against diet-induced obesity and insulin resistance.
Collapse
Affiliation(s)
- Alexis Diaz-Vegas
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.
| | - Kristen C Cooke
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Harry B Cutler
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Belinda Yau
- School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Stewart W C Masson
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Dylan Harney
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Oliver K Fuller
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Meg Potter
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Søren Madsen
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Niamh R Craw
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Yiju Zhang
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Cesar L Moreno
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Melkam A Kebede
- School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - G Gregory Neely
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Jacqueline Stöckli
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - James G Burchfield
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|
8
|
Byeon S, McKay MJ, Molloy MP, Gill AJ, Samra JS, Mittal A, Sahni S. Novel serum protein biomarker panel for early diagnosis of pancreatic cancer. Int J Cancer 2024; 155:365-371. [PMID: 38519999 DOI: 10.1002/ijc.34928] [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: 10/27/2023] [Revised: 01/29/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers. Late presentation of disease at the time of diagnosis is one of the major reasons for dismal prognostic outcomes for PDAC patients. Currently, there is a lack of clinical biomarkers, which can be used to diagnose PDAC patients at an early resectable stage. This study performed proteomic mass spectrometry to identify novel blood-based biomarkers for early diagnosis of PDAC. Serum specimens from 88 PDAC patients and 88 healthy controls (60 discovery cohort and 28 validation cohort) were analyzed using data independent acquisition high resolution mass spectrometry to identify candidate biomarker proteins. A total of 249 proteins were identified and quantified by the mass spectrometric analysis. Six proteins were markedly (>1.5 fold) and significantly (p < .05; q < 0.1) increased in PDAC patients compared to healthy controls in discovery cohort. Notably, four of these six proteins were significantly upregulated in an independent validation cohort. The top three upregulated proteins (i.e., Polymeric Immunoglobulin Receptor [PIGR], von Willebrand Factor [vWF], and Fibrinogen) were validated using enzyme linked immunosorbent assay, which led to selection of PIGR and vWF as a diagnostic biomarker panel for PDAC. The panel showed high ability to diagnose early stage (stage I and II) PDAC patients (area under the curve [AUC]: 0.8926), which was further improved after the addition of clinically used prognostic biomarker (Ca 19-9) to the panel (AUC: 0.9798). In conclusion, a novel serum protein biomarker panel for early diagnosis of PDAC was identified.
Collapse
Affiliation(s)
- Sooin Byeon
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Matthew J McKay
- Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Mark P Molloy
- Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
- Bowel Cancer and Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Anthony J Gill
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Jaswinder S Samra
- Australian Pancreatic Centre, St Leonards, Sydney, New South Wales, Australia
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, New South Wales, Australia
| | - Anubhav Mittal
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, New South Wales, Australia
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, New South Wales, Australia
- Department of Surgery, The University of Notre Dame Australia, Sydney, New South Wales, Australia
| | - Sumit Sahni
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, New South Wales, Australia
| |
Collapse
|
9
|
Nazari H, Cho AN, Goss D, Thiery JP, Ebrahimi Warkiani M. Impact of brain organoid-derived sEVs on metastatic adaptation and invasion of breast carcinoma cells through a microphysiological system. LAB ON A CHIP 2024; 24:3434-3455. [PMID: 38888211 DOI: 10.1039/d4lc00296b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Brain metastases are common in triple-negative breast cancer (TNBC), suggesting a complex process of cancer spread. The mechanisms enabling TNBC cell adaptation and proliferation in the brain remain unclear. Small extracellular vesicles (sEVs) play a crucial role in communication between breast carcinoma cells and the brain. However, the lack of relevant models hinders understanding of sEV-mediated communication. The present study assesses the impact of brain organoid-derived sEVs (BO-sEVs) on various behaviours of the MDA-MB-231 cell line, chosen as a representative of TNBC in a 3D microfluidic model. Our results demonstrate that 150-200 nm sEVs expressing CD63, CD9, and CD81 from brain organoid media decrease MDA-MB-231 cell proliferation, enhance their wound-healing capacity, alter their morphology into more mesenchymal mode, and increase their stemness. BO-sEVs led to heightened PD-L1, CD49f, and vimentin levels of expression in MDA-MB-231 cells, suggesting an amplified immunosuppressive, stem-like, and mesenchymal phenotype. Furthermore, these sEVs also induced the expression of neural markers such as GFAP in carcinoma cells. The cytokine antibody profiling array also showed that BO-sEVs enhanced the secretion of MCP-1, IL-6, and IL-8 by MDA-MB-231 cells. Moreover, sEVs significantly enhance the migration and invasion of carcinoma cells toward brain organoids in a 3D organoid-on-a-chip system. Our findings emphasize the potential significance of metastatic site-derived sEVs as pivotal mediators in carcinoma progression and adaptation to the brain microenvironment, thereby unveiling novel therapeutic avenues.
Collapse
Affiliation(s)
- Hojjatollah Nazari
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, Australia.
| | - Ann-Na Cho
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Dale Goss
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, Australia.
| | - Jean Paul Thiery
- UMR 7057 CNRS Matter and Complex Systems, Université Paris Cité, Paris, France
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
10
|
Lewis JM, Jebeli L, Coulon PML, Lay CE, Scott NE. Glycoproteomic and proteomic analysis of Burkholderia cenocepacia reveals glycosylation events within FliF and MotB are dispensable for motility. Microbiol Spectr 2024; 12:e0034624. [PMID: 38709084 PMCID: PMC11237607 DOI: 10.1128/spectrum.00346-24] [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: 02/10/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Across the Burkholderia genus O-linked protein glycosylation is highly conserved. While the inhibition of glycosylation has been shown to be detrimental for virulence in Burkholderia cepacia complex species, such as Burkholderia cenocepacia, little is known about how specific glycosylation sites impact protein functionality. Within this study, we sought to improve our understanding of the breadth, dynamics, and requirement for glycosylation across the B. cenocepacia O-glycoproteome. Assessing the B. cenocepacia glycoproteome across different culture media using complementary glycoproteomic approaches, we increase the known glycoproteome to 141 glycoproteins. Leveraging this repertoire of glycoproteins, we quantitively assessed the glycoproteome of B. cenocepacia using Data-Independent Acquisition (DIA) revealing the B. cenocepacia glycoproteome is largely stable across conditions with most glycoproteins constitutively expressed. Examination of how the absence of glycosylation impacts the glycoproteome reveals that the protein abundance of only five glycoproteins (BCAL1086, BCAL2974, BCAL0525, BCAM0505, and BCAL0127) are altered by the loss of glycosylation. Assessing ΔfliF (ΔBCAL0525), ΔmotB (ΔBCAL0127), and ΔBCAM0505 strains, we demonstrate the loss of FliF, and to a lesser extent MotB, mirror the proteomic effects observed in the absence of glycosylation in ΔpglL. While both MotB and FliF are essential for motility, we find loss of glycosylation sites in MotB or FliF does not impact motility supporting these sites are dispensable for function. Combined this work broadens our understanding of the B. cenocepacia glycoproteome supporting that the loss of glycoproteins in the absence of glycosylation is not an indicator of the requirement for glycosylation for protein function. IMPORTANCE Burkholderia cenocepacia is an opportunistic pathogen of concern within the Cystic Fibrosis community. Despite a greater appreciation of the unique physiology of B. cenocepacia gained over the last 20 years a complete understanding of the proteome and especially the O-glycoproteome, is lacking. In this study, we utilize systems biology approaches to expand the known B. cenocepacia glycoproteome as well as track the dynamics of glycoproteins across growth phases, culturing media and in response to the loss of glycosylation. We show that the glycoproteome of B. cenocepacia is largely stable across conditions and that the loss of glycosylation only impacts five glycoproteins including the motility associated proteins FliF and MotB. Examination of MotB and FliF shows, while these proteins are essential for motility, glycosylation is dispensable. Combined this work supports that B. cenocepacia glycosylation can be dispensable for protein function and may influence protein properties beyond stability.
Collapse
Affiliation(s)
- Jessica M Lewis
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Leila Jebeli
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Pauline M L Coulon
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Catrina E Lay
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
11
|
Koay YC, Liu RP, McIntosh B, Vigder N, Lauren S, Bai AY, Tomita S, Li D, Harney D, Hunter B, Zhang Y, Yang J, Bannon P, Philp A, Philp A, Kaye DM, Larance M, Lal S, O’Sullivan JF. The Efficacy of Risk Factor Modification Compared to NAD + Repletion in Diastolic Heart Failure. JACC Basic Transl Sci 2024; 9:733-750. [PMID: 39070276 PMCID: PMC11282886 DOI: 10.1016/j.jacbts.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 07/30/2024]
Abstract
Heart failure (HF) with left ventricular diastolic dysfunction is a growing global concern. This study evaluated myocardial oxidized nicotinamide adenine dinucleotide (NAD+) levels in human systolic and diastolic HF and in a murine model of HF with preserved ejection fraction, exploring NAD+ repletion as therapy. We quantified myocardial NAD+ and nicotinamide phosphoribosyltransferase levels, assessing restoration with nicotinamide riboside (NR). Findings show significant NAD+ and nicotinamide phosphoribosyltransferase depletion in human diastolic HF myocardium, but NR successfully restored NAD+ levels. In murine HF with preserved ejection fraction, NR as preventive and therapeutic intervention improved metabolic and antioxidant profiles. This study underscores NAD+ repletion's potential in diastolic HF management.
Collapse
Affiliation(s)
- Yen Chin Koay
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Ren Ping Liu
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Bailey McIntosh
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Niv Vigder
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Serlin Lauren
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Angela Yu Bai
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Saki Tomita
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Desmond Li
- BCAL Diagnostics, National Innovation Centre, Eveleigh, New South Wales, Australia
| | - Dylan Harney
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Hunter
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory, The University of Sydney, New South Wales, Australia
| | - Yunwei Zhang
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Jean Yang
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul Bannon
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiothoracic Surgery, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Ashleigh Philp
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, New South Wales, Australia
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Andrew Philp
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Healthy Aging, Centenary Institute, Sydney, New South Wales, Australia
- School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - David M. Kaye
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia
- Faculty of Medicine, Monash University, Melbourne, Australia
| | - Mark Larance
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Sean Lal
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Precision Cardiovascular Laboratory, The University of Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F. O’Sullivan
- Cardiometabolic Medicine Group, The University of Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
12
|
Van Woerkom A, Harney DJ, Nagarajan SR, Hakeem-Sanni MF, Lin J, Hooke M, Pulpitel T, Cooney GJ, Larance M, Saunders DN, Brandon AE, Hoy AJ. Hepatic lipid droplet-associated proteome changes distinguish dietary-induced fatty liver from glucose tolerance in male mice. Am J Physiol Endocrinol Metab 2024; 326:E842-E855. [PMID: 38656127 PMCID: PMC11376491 DOI: 10.1152/ajpendo.00013.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
Fatty liver is characterized by the expansion of lipid droplets (LDs) and is associated with the development of many metabolic diseases. We assessed the morphology of hepatic LDs and performed quantitative proteomics in lean, glucose-tolerant mice compared with high-fat diet (HFD) fed mice that displayed hepatic steatosis and glucose intolerance as well as high-starch diet (HStD) fed mice who exhibited similar levels of hepatic steatosis but remained glucose tolerant. Both HFD- and HStD-fed mice had more and larger LDs than Chow-fed animals. We observed striking differences in liver LD proteomes of HFD- and HStD-fed mice compared with Chow-fed mice, with fewer differences between HFD and HStD. Taking advantage of our diet strategy, we identified a fatty liver LD proteome consisting of proteins common in HFD- and HStD-fed mice, as well as a proteome associated with glucose tolerance that included proteins shared in Chow and HStD but not HFD-fed mice. Notably, glucose intolerance was associated with changes in the ratio of adipose triglyceride lipase to perilipin 5 in the LD proteome, suggesting dysregulation of neutral lipid homeostasis in glucose-intolerant fatty liver. We conclude that our novel dietary approach uncouples ectopic lipid burden from insulin resistance-associated changes in the hepatic lipid droplet proteome.NEW & NOTEWORTHY This study identified a fatty liver lipid droplet proteome and one associated with glucose tolerance. Notably, glucose intolerance was linked with changes in the ratio of adipose triglyceride lipase to perilipin 5 that is indicative of dysregulation of neutral lipid homeostasis.
Collapse
Affiliation(s)
- Andries Van Woerkom
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Dylan J Harney
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Science, School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Shilpa R Nagarajan
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Mariam F Hakeem-Sanni
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Jinfeng Lin
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Matthew Hooke
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Tamara Pulpitel
- Faculty of Science, School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Gregory J Cooney
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Mark Larance
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Darren N Saunders
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Amanda E Brandon
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- Faculty of Science, School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew J Hoy
- Faculty of Medicine and Health, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
13
|
Pinello N, Song R, Lee Q, Calonne E, Larance M, Fuks F, Wong JJL. A multiomics dataset for the study of RNA modifications in human macrophage differentiation and polarisation. Sci Data 2024; 11:252. [PMID: 38418823 PMCID: PMC10902381 DOI: 10.1038/s41597-024-03076-8] [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: 09/26/2023] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
RNA modifications have emerged as central regulators of gene expression programs. Amongst RNA modifications are N6-methyladenosine (m6A) and RNA 5-hydroxymethylcytosine (5hmC). While m6A is established as a versatile regulator of RNA metabolism, the functions of RNA 5hmC are unclear. Despite some evidence linking RNA modifications to immunity, their implications in gene expression control in macrophage development and functions remain unclear. Here we present a multi-omics dataset capturing different layers of the gene expression programs driving macrophage differentiation and polarisation. We obtained mRNA-Seq, m6A-IP-Seq, 5hmC-IP-Seq, Polyribo-Seq and LC-MS/MS data from monocytes and resting-, pro- and anti-inflammatory-like macrophages. We present technical validation showing high quality and correlation between samples for all datasets, and evidence of biological consistency of modelled macrophages at the transcriptomic, epitranscriptomic, translational and proteomic levels. This multi-omics dataset provides a resource for the study of RNA m6A and 5hmC in the context of macrophage biology and spans the gene expression process from transcripts to proteins.
Collapse
Affiliation(s)
- Natalia Pinello
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
| | - Renhua Song
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
| | - Quintin Lee
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia
| | - Emilie Calonne
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB Cancer Research Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, The University of Sydney, Camperdown, 2050, New South Wales, Australia
| | - François Fuks
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB Cancer Research Center (U-CRC), Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Justin J-L Wong
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown, 2050, Australia.
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050, Australia.
| |
Collapse
|
14
|
Khan N, Sengupta P. Technological Advancement and Trend in Selective Bioanalytical Sample Extraction through State of the Art 3-D Printing Techniques Aiming 'Sorbent Customization as per need'. Crit Rev Anal Chem 2024:1-21. [PMID: 38319592 DOI: 10.1080/10408347.2024.2305275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The inherent complexity of biological matrices and presence of several interfering substances in biological samples make them unsuitable for direct analysis. An effective sample preparation technique assists in analyte enrichment, improving selectivity and sensitivity of bioanalytical method. Because of several key benefits of employing 3D printed sorbent in sample extraction, it has recently gained popularity across a variety of industries. Applications for 3D printing in the field of bioanalytical research have grown recently, particularly in the areas of miniaturization, (bio)sensing, sample preparation, and separation sciences. Due to the high expense of the solid phase microextraction cartridge, researcher approaches in-lab production of sorbent material for the extraction of analyte from biological samples. Owing to its distinct advantages such as low costs, automation capabilities, capacity to produce products in a variety of shapes, and reduction of tedious steps of sample preparation, 3D printed sorbents are gaining increased attention in the field of bioanalysis. It is also reported to offer high selectivity and assist in achieving a much lower limit of detection. In this review, we have discussed current advancements in different types of 3D printed sorbents, production methods, and their applications in the field of bioanalytical sample preparation.
Collapse
Affiliation(s)
- Nasir Khan
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Government of India, Gandhinagar, Gujarat, India
| | - Pinaki Sengupta
- National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Government of India, Gandhinagar, Gujarat, India
| |
Collapse
|
15
|
Dieters-Castator DZ, Manzanillo P, Yang HY, Modak RV, Rardin MJ, Gibson BW. Magnetic Bead-Based Workflow for Sensitive and Streamlined Cell Surface Proteomics. J Proteome Res 2024; 23:618-632. [PMID: 38226771 DOI: 10.1021/acs.jproteome.3c00432] [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] [Indexed: 01/17/2024]
Abstract
Cell surface proteins represent an important class of molecules for therapeutic targeting and cellular phenotyping. However, their enrichment and detection via mass spectrometry-based proteomics remains challenging due to low abundance, post-translational modifications, hydrophobic regions, and processing requirements. To improve their identification, we optimized a Cell-Surface Capture (CSC) workflow that incorporates magnetic bead-based processing. Using this approach, we evaluated labeling conditions (biotin tags and catalysts), enrichment specificity (streptavidin beads), missed cleavages (lysis buffers), nonenzymatic deamidation (digestion and deglycosylation buffers), and data acquisition methods (DDA, DIA, and TMT). Our findings support the use of alkoxyamine-PEG4-biotin plus 5-methoxy-anthranilic acid, SDS/urea-based lysis buffers, single-pot solid-phased-enhanced sample-preparation (SP3), and streptavidin magnetic beads for maximal surfaceome coverage. Notably, with semiautomated processing, sample handling was simplified and between ∼600 and 900 cell surface N-glycoproteins were identified from only 25-200 μg of HeLa protein. CSC also revealed significant differences between in vitro monolayer cultures and in vivo tumor xenografts of murine CT26 colon adenocarcinoma samples that may aid in target identification for drug development. Overall, the improved efficiency of the magnetic-based CSC workflow identified both previously reported and novel N-glycosites with less material and high reproducibility that should help advance the field of surfaceomics by providing insight in cellular phenotypes not previously documented.
Collapse
Affiliation(s)
| | - Paolo Manzanillo
- Inflammation, Amgen Research, South San Francisco, California 94080, United States
| | - Han-Yin Yang
- Discovery Proteomics, Amgen Research, South San Francisco, California 94080, United States
| | - Rucha V Modak
- Inflammation, Amgen Research, South San Francisco, California 94080, United States
| | - Matthew J Rardin
- Discovery Proteomics, Amgen Research, South San Francisco, California 94080, United States
| | - Bradford W Gibson
- Discovery Proteomics, Amgen Research, South San Francisco, California 94080, United States
| |
Collapse
|
16
|
Houlahan CB, Kong Y, Johnston B, Cielesh M, Chau TH, Fenwick J, Coleman PR, Hao H, Haltiwanger RS, Thaysen-Andersen M, Passam FH, Larance M. Analysis of the Healthy Platelet Proteome Identifies a New Form of Domain-Specific O-Fucosylation. Mol Cell Proteomics 2024; 23:100717. [PMID: 38237698 PMCID: PMC10879016 DOI: 10.1016/j.mcpro.2024.100717] [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: 07/14/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Platelet activation induces the secretion of proteins that promote platelet aggregation and inflammation. However, detailed analysis of the released platelet proteome is hampered by platelets' tendency to preactivate during their isolation and a lack of sensitive protocols for low abundance releasate analysis. Here, we detail the most sensitive analysis to date of the platelet releasate proteome with the detection of >1300 proteins. Unbiased scanning for posttranslational modifications within releasate proteins highlighted O-glycosylation as being a major component. For the first time, we detected O-fucosylation on previously uncharacterized sites including multimerin-1 (MMRN1), a major alpha granule protein that supports platelet adhesion to collagen and is a carrier for platelet factor V. The N-terminal elastin microfibril interface (EMI) domain of MMRN1, a key site for protein-protein interaction, was O-fucosylated at a conserved threonine within a new domain context. Our data suggest that either protein O-fucosyltransferase 1, or a novel protein O-fucosyltransferase, may be responsible for this modification. Mutating this O-fucose site on the EMI domain led to a >50% reduction of MMRN1 secretion, supporting a key role of EMI O-fucosylation in MMRN1 secretion. By comparing releasates from resting and thrombin-treated platelets, 202 proteins were found to be significantly released after high-dose thrombin stimulation. Complementary quantification of the platelet lysates identified >3800 proteins, which confirmed the platelet origin of releasate proteins by anticorrelation analysis. Low-dose thrombin treatment yielded a smaller subset of significantly regulated proteins with fewer secretory pathway enzymes. The extensive platelet proteome resource provided here (larancelab.com/platelet-proteome) allows identification of novel regulatory mechanisms for drug targeting to address platelet dysfunction and thrombosis.
Collapse
Affiliation(s)
- Callum B Houlahan
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Yvonne Kong
- Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Bede Johnston
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Michelle Cielesh
- Charles Perkins Centre, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - The Huong Chau
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales, Australia
| | - Jemma Fenwick
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia; Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul R Coleman
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Huilin Hao
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Robert S Haltiwanger
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Morten Thaysen-Andersen
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales, Australia; Institute for Glyco-Core Research, Nagoya University, Nagoya, Aichi, Japan
| | - Freda H Passam
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia; Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
17
|
O'Rourke MB, Hansbro PP, Molloy MP. Reusing Peptide Spectral Reference Libraries to Discover Putative Plasma Biomarkers of Response to Cancer Chemotherapy. Methods Mol Biol 2024; 2823:241-251. [PMID: 39052224 DOI: 10.1007/978-1-0716-3922-1_15] [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] [Indexed: 07/27/2024]
Abstract
Biofluids such as blood plasma are rich reservoirs of potential biomarkers for disease diagnosis, prognosis, and prediction of treatment response. However, mass spectrometry analysis of circulating plasma proteins remains challenging. The introduction of data-independent acquisition mass spectrometry (DIA-MS) is an important step toward addressing detection of less abundant plasma proteins. Numerous plasma peptide MS/MS spectral library datasets produced from extensive plasma fractionation are accessible from public archives, and these can be repurposed as spectral reference libraries to increase the depth of proteomic analysis when DIA-MS is used. Here we describe the workflow that relies on reusing the existing spectral reference libraries by populating them with locally obtained peptide MS/MS data acquired by DIA-MS. This approach was demonstrated effectively to identify putative plasma biomarkers of response to neoadjuvant chemotherapy in the setting of pancreatic ductal adenocarcinoma (PDAC) (O'Rourke et al., J Proteomics 231:103998, 2021).
Collapse
Affiliation(s)
- Matthew B O'Rourke
- Bowel Cancer & Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Philip P Hansbro
- Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Mark P Molloy
- Bowel Cancer & Biomarker Laboratory, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia.
| |
Collapse
|
18
|
Lim Kam Sian TCC, Sun C, Cain JE, Steele JR, Hanchapola I, Stoychev S, Schittenhelm RB, Faridi P. A Semiautomated Proteomics and Phosphoproteomics Protocol for the Identification of Novel Therapeutic Targets and Predictive Biomarkers in In Vivo Xenograft Models of Pediatric Cancers. Methods Mol Biol 2024; 2806:229-242. [PMID: 38676807 DOI: 10.1007/978-1-0716-3858-3_17] [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] [Indexed: 04/29/2024]
Abstract
Genomic profiling has identified therapeutic targets for precision treatment of certain cancers, but many patients lack actionable mutations. Additional omics approaches, like proteomics and phosphoproteomics, are essential for comprehensive mapping of cancer-associated molecular phenotypes. In vivo models, such as cell line and patient-derived xenografts (PDX), offer valuable insights into cancer biology and treatment strategies.This chapter presents a semiautomated high-throughput workflow for integrated proteomics and phosphoproteomics analysis on the Kingfish platform coupled with MagReSyn® Zr-IMAC HP. It enhances protein extraction from in vivo xenograft samples and provides better insights into cancers with poor prognosis. The approach successfully identified over 11,000 unique phosphosites and ~6000 proteins in SJSA-1 pediatric osteosarcoma xenografts, demonstrating its efficacy. This workflow is a valuable tool for studying tumor biology and developing precision oncology strategies.
Collapse
Affiliation(s)
- Terry C C Lim Kam Sian
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Medicine, Sub-Faculty of Clinical and Molecular Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Christie Sun
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Jason E Cain
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Joel R Steele
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Iresha Hanchapola
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| | - Pouya Faridi
- Monash Proteomics and Metabolomics Platform, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
- Department of Medicine, Sub-Faculty of Clinical and Molecular Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University, Clayton, VIC, Australia.
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
19
|
Wilken L, Lasswitz L, Scaturro P, Gerold G. Identification of RVFV Host Factors Using Quantitative Interaction Proteomics. Methods Mol Biol 2024; 2824:189-202. [PMID: 39039414 DOI: 10.1007/978-1-0716-3926-9_13] [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] [Indexed: 07/24/2024]
Abstract
Affinity enrichment coupled with liquid chromatography-tandem mass spectrometry (AE-LC-MS/MS) enables a comprehensive study of virus-host protein-protein interactions in cells and tissues infected with Rift Valley fever virus (RVFV) or ectopically expressing RVFV proteins. Depending on the research question, different experimental setups with carefully chosen controls are needed. Here, we describe the detailed workflow of sample preparation, processing, and cleanup, while also outlining critical points to consider when designing and performing AE-LC-MS/MS experiments.
Collapse
Affiliation(s)
- Lucas Wilken
- Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Lisa Lasswitz
- Institute of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Gisa Gerold
- Institute of Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany.
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden.
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden.
| |
Collapse
|
20
|
Morales-Suárez-Varela M, Llopis-Morales J. Intermittent fasting diet and health. Med Clin (Barc) 2023; 161:297-299. [PMID: 37474393 DOI: 10.1016/j.medcli.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/22/2023]
Affiliation(s)
- María Morales-Suárez-Varela
- Grupo de investigación en Epidemiología Social y Nutricional, Farmacoepidemiología y Salud Pública. Departamento de Medicina Preventiva y Salud Pública, Ciencias de la Alimentación, Toxicología y Medicina Legal. Facultad de Farmacia. Universitat de València, Burjassot (Valencia), España; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, España.
| | | |
Collapse
|
21
|
Hermesdorf M, Esselmann H, Morgado B, Jahn-Brodmann A, Herrera-Rivero M, Wiltfang J, Berger K. The association of body mass index and body composition with plasma amyloid beta levels. Brain Commun 2023; 5:fcad263. [PMID: 37901043 PMCID: PMC10608109 DOI: 10.1093/braincomms/fcad263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/04/2023] [Accepted: 10/08/2023] [Indexed: 10/31/2023] Open
Abstract
Blood-based analysis of amyloid-β is increasingly applied to incrementally establish diagnostic tests for Alzheimer's disease. To this aim, it is necessary to determine factors that can alter blood-based concentrations of amyloid-β. We cross-sectionally analysed amyloid-β-40 and amyloid-β-42 concentrations and the 40/42 ratio in 440 community-dwelling adults and associations with body mass index, waist-to-height ratio and body composition assessed using bioelectrical impedance analysis. Body mass index and waist-to-height ratio were inversely associated with plasma amyloid-β-42 concentrations. Body fat mass, but not body cell mass and extracellular mass, was inversely associated with amyloid-β-42 levels. The results indicate that plasma concentrations of amyloid-β-42 are lower in those with increased body mass index and body fat, and associations with amyloid-β-40 did not reach significance after controlling for multiple testing. The findings support the use of body mass index as an easy-to-measure factor that should be accounted for in diagnostic models for plasma amyloid-β.
Collapse
Affiliation(s)
- Marco Hermesdorf
- Institute of Epidemiology and Social Medicine, University of Münster, Münster 48149, Germany
| | - Hermann Esselmann
- Department of Psychiatry, University Medical Center Göttingen, Goettingen 37075, Germany
| | - Barbara Morgado
- Department of Psychiatry, University Medical Center Göttingen, Goettingen 37075, Germany
| | - Anke Jahn-Brodmann
- Department of Psychiatry, University Medical Center Göttingen, Goettingen 37075, Germany
| | - Marisol Herrera-Rivero
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Münster 48149, Germany
- Department of Psychiatry, University of Münster, Münster 48149, Germany
| | - Jens Wiltfang
- Department of Psychiatry, University Medical Center Göttingen, Goettingen 37075, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen 37075, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro 3810-29992, Portugal
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster 48149, Germany
| |
Collapse
|
22
|
Lee JY, Harney DJ, Teo JD, Kwok JB, Sutherland GT, Larance M, Don AS. The major TMEM106B dementia risk allele affects TMEM106B protein levels, fibril formation, and myelin lipid homeostasis in the ageing human hippocampus. Mol Neurodegener 2023; 18:63. [PMID: 37726834 PMCID: PMC10510131 DOI: 10.1186/s13024-023-00650-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND The risk for dementia increases exponentially from the seventh decade of life. Identifying and understanding the biochemical changes that sensitize the ageing brain to neurodegeneration will provide new opportunities for dementia prevention and treatment. This study aimed to determine how ageing and major genetic risk factors for dementia affect the hippocampal proteome and lipidome of neurologically-normal humans over the age of 65. The hippocampus was chosen as it is highly susceptible to atrophy with ageing and in several neurodegenerative diseases. METHODS Mass spectrometry-based proteomic and lipidomic analysis of CA1 hippocampus samples from 74 neurologically normal human donors, aged 66-104, was used in combination with multiple regression models and gene set enrichment analysis to identify age-dependent changes in the proteome and lipidome. ANOVA was used to test the effect of major dementia risk alleles in the TMEM106B and APOE genes on the hippocampal proteome and lipidome, adjusting for age, gender, and post-mortem interval. Fibrillar C-terminal TMEM106B fragments were isolated using sarkosyl fractionation and quantified by immunoblotting. RESULTS Forty proteins were associated with age at false discovery rate-corrected P < 0.05, including proteins that regulate cell adhesion, the cytoskeleton, amino acid and lipid metabolism, and ribosomal subunits. TMEM106B, a regulator of lysosomal and oligodendrocyte function, was regulated with greatest effect size. The increase in TMEM106B levels with ageing was specific to carriers of the rs1990622-A allele in the TMEM106B gene that increases risk for frontotemporal dementia, Alzheimer's disease, Parkinson's disease, and hippocampal sclerosis with ageing. Rs1990622-A was also associated with higher TMEM106B fibril content. Hippocampal lipids were not significantly affected by APOE genotype, however levels of myelin-enriched sulfatides and hexosylceramides were significantly lower, and polyunsaturated phospholipids were higher, in rs1990622-A carriers after controlling for APOE genotype. CONCLUSIONS Our study demonstrates that TMEM106B protein abundance is increased with brain ageing in humans, establishes that dementia risk allele rs1990622-A predisposes to TMEM106B fibril formation in the hippocampus, and provides the first evidence that rs1990622-A affects brain lipid homeostasis, particularly myelin lipids. Our data suggests that TMEM106B is one of a growing list of major dementia risk genes that affect glial lipid metabolism.
Collapse
Affiliation(s)
- Jun Yup Lee
- Charles Perkins Centre, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, Camperdown, NSW, 2006, Australia
| | - Dylan J Harney
- Charles Perkins Centre, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, Camperdown, NSW, 2006, Australia
| | - Jonathan D Teo
- Charles Perkins Centre, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, Camperdown, NSW, 2006, Australia
| | - John B Kwok
- School of Medical Sciences, Camperdown, NSW, 2006, Australia
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Greg T Sutherland
- Charles Perkins Centre, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, Camperdown, NSW, 2006, Australia
| | - Mark Larance
- Charles Perkins Centre, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, Camperdown, NSW, 2006, Australia
| | - Anthony S Don
- Charles Perkins Centre, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
23
|
Abstract
Overweight and obesity are an important public health problem that affects a significant part of the world population and increases the risk of many metabolic diseases. Weight loss is the primary goal in obesity treatment, and many different dietary interventions are tried for this purpose. Intermittent fasting is a diet that has become popular in recent years with the weight loss it provides and includes periods of fasting and feeding. In addition to providing weight loss, intermittent fasting also has positive effects on important risk factors such as glucoregulatory parameters, blood lipids, and oxidative stress. Intermittent fasting appears to be an effective and safe way to achieve weight loss in obesity. It could also have therapeutic effects on obesity-related diseases. The aim of this review was to bring together up-to-date information on the effects of intermittent fasting on obesity and various obesity-related diseases, mechanisms of action, possible benefits and harms, and potential uses.
Collapse
Affiliation(s)
- Caner Özyildirim
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara University, Ankara, Türkiye -
| | - Asli Uçar
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ankara University, Ankara, Türkiye
| |
Collapse
|
24
|
Vernardis SI, Demichev V, Lemke O, Grüning NM, Messner C, White M, Pietzner M, Peluso A, Collet TH, Henning E, Gille C, Campbell A, Hayward C, Porteous DJ, Marioni RE, Mülleder M, Zelezniak A, Wareham NJ, Langenberg C, Farooqi IS, Ralser M. The Impact of Acute Nutritional Interventions on the Plasma Proteome. J Clin Endocrinol Metab 2023; 108:2087-2098. [PMID: 36658456 PMCID: PMC10348471 DOI: 10.1210/clinem/dgad031] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
CONTEXT Humans respond profoundly to changes in diet, while nutrition and environment have a great impact on population health. It is therefore important to deeply characterize the human nutritional responses. OBJECTIVE Endocrine parameters and the metabolome of human plasma are rapidly responding to acute nutritional interventions such as caloric restriction or a glucose challenge. It is less well understood whether the plasma proteome would be equally dynamic, and whether it could be a source of corresponding biomarkers. METHODS We used high-throughput mass spectrometry to determine changes in the plasma proteome of i) 10 healthy, young, male individuals in response to 2 days of acute caloric restriction followed by refeeding; ii) 200 individuals of the Ely epidemiological study before and after a glucose tolerance test at 4 time points (0, 30, 60, 120 minutes); and iii) 200 random individuals from the Generation Scotland study. We compared the proteomic changes detected with metabolome data and endocrine parameters. RESULTS Both caloric restriction and the glucose challenge substantially impacted the plasma proteome. Proteins responded across individuals or in an individual-specific manner. We identified nutrient-responsive plasma proteins that correlate with changes in the metabolome, as well as with endocrine parameters. In particular, our study highlights the role of apolipoprotein C1 (APOC1), a small, understudied apolipoprotein that was affected by caloric restriction and dominated the response to glucose consumption and differed in abundance between individuals with and without type 2 diabetes. CONCLUSION Our study identifies APOC1 as a dominant nutritional responder in humans and highlights the interdependency of acute nutritional response proteins and the endocrine system.
Collapse
Affiliation(s)
- Spyros I Vernardis
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Vadim Demichev
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Oliver Lemke
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Nana-Maria Grüning
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Christoph Messner
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Matt White
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, CB2 0SL, UK
- Computational Medicine, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Alina Peluso
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
| | - Tinh-Hai Collet
- Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
- Service of Endocrinology, Diabetology, Nutrition and Therapeutic Education, Department of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Elana Henning
- Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Christoph Gille
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Michael Mülleder
- Core Facility High Throughput Mass Spectrometry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Aleksej Zelezniak
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius SE-412 96, Lithuania
- Randall Centre for Cell & Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, SE1 1UL London, UK
| | | | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, CB2 0SL, UK
- Computational Medicine, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, E1 1HH, UK
| | - I Sadaf Farooqi
- Metabolic Research Laboratories and National Institute for Health Research Cambridge Biomedical Research Centre, Wellcome-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Markus Ralser
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW1 1HT, UK
- Department of Biochemistry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
25
|
Harney DJ, Cielesh M, Roberts GE, Vila IK, Viengkhou B, Hofer MJ, Laguette N, Larance M. Dietary restriction induces a sexually dimorphic type I interferon response in mice with gene-environment interactions. Cell Rep 2023; 42:112559. [PMID: 37243595 DOI: 10.1016/j.celrep.2023.112559] [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: 07/12/2022] [Revised: 03/20/2023] [Accepted: 05/08/2023] [Indexed: 05/29/2023] Open
Abstract
Intermittent fasting (IF) is an established intervention to treat the growing obesity epidemic. However, the interaction between dietary interventions and sex remains a significant knowledge gap. In this study, we use unbiased proteome analysis to identify diet-sex interactions. We report sexual dimorphism in response to intermittent fasting within lipid and cholesterol metabolism and, unexpectedly, in type I interferon signaling, which was strongly induced in females. We verify that secretion of type I interferon is required for the IF response in females. Gonadectomy differentially alters the every-other-day fasting (EODF) response and demonstrates that sex hormone signaling can either suppress or enhance the interferon response to IF. IF fails to potentiate a stronger innate immune response when IF-treated animals were challenged with a viral mimetic. Lastly, the IF response changes with genotype and environment. These data reveal an interesting interaction between diet, sex, and the innate immune system.
Collapse
Affiliation(s)
- Dylan J Harney
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, 2006 NSW, Australia
| | - Michelle Cielesh
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Sydney, 2006 NSW, Australia
| | - Georgia E Roberts
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, 2006 NSW, Australia
| | | | - Barney Viengkhou
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, 2006 NSW, Australia
| | - Markus J Hofer
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, 2006 NSW, Australia
| | | | - Mark Larance
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Sydney, 2006 NSW, Australia.
| |
Collapse
|
26
|
Nigos LR, Scott NE, Brooks AG, Ait-Goughoulte M, Londrigan SL, Reading PC, Farrukee R. TRIM16 Overexpression in HEK293T Cells Results in Cell Line-Specific Antiviral Activity. Pathogens 2023; 12:852. [PMID: 37375542 DOI: 10.3390/pathogens12060852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
Host cell restriction factors are intracellular proteins that can inhibit virus replication. Characterisation of novel host cell restriction factors can provide potential targets for host-directed therapies. In this study, we aimed to assess a member of the Tripartite-motif family protein (TRIM) family, TRIM16, as a putative host cell restriction factor. To this end, we utilized constitutive or doxycycline-inducible systems to overexpress TRIM16 in HEK293T epithelial cells and then tested for its ability to inhibit growth by a range of RNA and DNA viruses. In HEK293T cells, overexpression of TRIM16 resulted in potent inhibition of multiple viruses, however, when TRIM16 was overexpressed in other epithelial cell lines (A549, Hela, or Hep2), virus inhibition was not observed. When investigating the antiviral activity of endogenous TRIM16, we report that siRNA-mediated knockdown of TRIM16 in A549 cells also modulated the mRNA expression of other TRIM proteins, complicating the interpretation of results using this method. Therefore, we used CRISPR/Cas9 editing to knockout TRIM16 in A549 cells and demonstrate that endogenous TRIM16 did not mediate antiviral activity against the viruses tested. Thus, while initial overexpression in HEK293T cells suggested that TRIM16 was a host cell restriction factor, alternative approaches did not validate these findings. These studies highlight the importance of multiple complementary experimental approaches, including overexpression analysis in multiple cell lines and investigation of the endogenous protein, when defining host cell restriction factors with novel antiviral activity.
Collapse
Affiliation(s)
- Lance R Nigos
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne, VIC 3000, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne, VIC 3000, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne, VIC 3000, Australia
| | - Malika Ait-Goughoulte
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne, VIC 3000, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne, VIC 3000, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Disease Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne, VIC 3000, Australia
| | - Rubaiyea Farrukee
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St., Melbourne, VIC 3000, Australia
| |
Collapse
|
27
|
Lewis J, Scott NE. CRISPRi-Mediated Silencing of Burkholderia O-Linked Glycosylation Systems Enables the Depletion of Glycosylation Yet Results in Modest Proteome Impacts. J Proteome Res 2023; 22:1762-1778. [PMID: 36995114 PMCID: PMC10243306 DOI: 10.1021/acs.jproteome.2c00790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Indexed: 03/31/2023]
Abstract
The process of O-linked protein glycosylation is highly conserved across the Burkholderia genus and mediated by the oligosaccharyltransferase PglL. While our understanding of Burkholderia glycoproteomes has increased in recent years, little is known about how Burkholderia species respond to modulations in glycosylation. Utilizing CRISPR interference (CRISPRi), we explored the impact of silencing of O-linked glycosylation across four species of Burkholderia; Burkholderia cenocepacia K56-2, Burkholderia diffusa MSMB375, Burkholderia multivorans ATCC17616, and Burkholderia thailandensis E264. Proteomic and glycoproteomic analyses revealed that while CRISPRi enabled inducible silencing of PglL, this did not abolish glycosylation, nor recapitulate phenotypes such as proteome changes or alterations in motility that are associated with glycosylation null strains, despite inhibition of glycosylation by nearly 90%. Importantly, this work also demonstrated that CRISPRi induction with high levels of rhamnose leads to extensive impacts on the Burkholderia proteomes, which without appropriate controls mask the impacts specifically driven by CRISPRi guides. Combined, this work revealed that while CRISPRi allows the modulation of O-linked glycosylation with reductions up to 90% at a phenotypic and proteome levels, Burkholderia appears to demonstrate a robust tolerance to fluctuations in glycosylation capacity.
Collapse
Affiliation(s)
- Jessica
M. Lewis
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute
for Infection and Immunity, Melbourne 3000, Australia
| | - Nichollas E. Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute
for Infection and Immunity, Melbourne 3000, Australia
| |
Collapse
|
28
|
O'Rourke MB, Januszewski AS, Sullivan DR, Lengyel I, Stewart AJ, Arya S, Ma RC, Galande S, Hardikar AA, Joglekar MV, Keech AC, Jenkins AJ, Molloy MP. Optimised plasma sample preparation and LC-MS analysis to support large-scale proteomic analysis of clinical trial specimens: Application to the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) trial. Proteomics Clin Appl 2023; 17:e2200106. [PMID: 36891577 PMCID: PMC10909541 DOI: 10.1002/prca.202200106] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/10/2023] [Accepted: 02/28/2023] [Indexed: 03/10/2023]
Abstract
PURPOSE Robust, affordable plasma proteomic biomarker workflows are needed for large-scale clinical studies. We evaluated aspects of sample preparation to allow liquid chromatography-mass spectrometry (LC-MS) analysis of more than 1500 samples from the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) trial of adults with type 2 diabetes. METHODS Using LC-MS with data-independent acquisition we evaluated four variables: plasma protein depletion, EDTA or citrated anti-coagulant blood collection tubes, plasma lipid depletion strategies and plasma freeze-thaw cycles. Optimised methods were applied in a pilot study of FIELD participants. RESULTS LC-MS of undepleted plasma conducted over a 45 min gradient yielded 172 proteins after excluding immunoglobulin isoforms. Cibachrome-blue-based depletion yielded additional proteins but with cost and time expenses, while immunodepleting albumin and IgG provided few additional identifications. Only minor variations were associated with blood collection tube type, delipidation methods and freeze-thaw cycles. From 65 batches involving over 1500 injections, the median intra-batch quantitative differences in the top 100 proteins of the plasma external standard were less than 2%. Fenofibrate altered seven plasma proteins. CONCLUSIONS AND CLINICAL RELEVANCE A robust plasma handling and LC-MS proteomics workflow for abundant plasma proteins has been developed for large-scale biomarker studies that balance proteomic depth with time and resource costs.
Collapse
Affiliation(s)
- Matthew B. O'Rourke
- Bowel Cancer & Biomarker LabSchool of Medical SciencesFaculty of Medicine and HealthThe University of SydneySydneyAustralia
- Centre for InflammationCentenary InstituteSydneyAustralia
- School of Life SciencesFaculty of ScienceUniversity of Technology SydneySydneyAustralia
| | - Andrzej S. Januszewski
- NHMRC Clinical Trials CentreFaculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - David R. Sullivan
- NHMRC Clinical Trials CentreFaculty of Medicine and HealthThe University of SydneySydneyAustralia
- Department of Chemical PathologyRoyal Prince Alfred HospitalNSW Health PathologyAustralia
| | - Imre Lengyel
- Wellcome‐Wolfson Institute for Experimental MedicineSchool of MedicineDentistry and Biomedical ScienceQueen's University BelfastBelfastBelfastUK
| | | | - Swati Arya
- School of MedicineUniversity of St AndrewsSt AndrewsFifeUK
| | - Ronald C. Ma
- Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong KongChina
| | | | - Anandwardhan A. Hardikar
- NHMRC Clinical Trials CentreFaculty of Medicine and HealthThe University of SydneySydneyAustralia
- Present address:
Diabetes and Islet Biology groupSchool of MedicineWestern Sydney UniversityCampbelltownAustralia
| | - Mugdha V. Joglekar
- NHMRC Clinical Trials CentreFaculty of Medicine and HealthThe University of SydneySydneyAustralia
- Present address:
Diabetes and Islet Biology groupSchool of MedicineWestern Sydney UniversityCampbelltownAustralia
| | - Anthony C. Keech
- NHMRC Clinical Trials CentreFaculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Alicia J. Jenkins
- NHMRC Clinical Trials CentreFaculty of Medicine and HealthThe University of SydneySydneyAustralia
- Baker Heart and Diabetes InstituteMelbourneAustralia
| | - Mark P. Molloy
- Bowel Cancer & Biomarker LabSchool of Medical SciencesFaculty of Medicine and HealthThe University of SydneySydneyAustralia
| |
Collapse
|
29
|
Wong SL, Kardia E, Vijayan A, Umashankar B, Pandzic E, Zhong L, Jaffe A, Waters SA. Molecular and Functional Characteristics of Airway Epithelium under Chronic Hypoxia. Int J Mol Sci 2023; 24:ijms24076475. [PMID: 37047450 PMCID: PMC10095024 DOI: 10.3390/ijms24076475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
Localized and chronic hypoxia of airway mucosa is a common feature of progressive respiratory diseases, including cystic fibrosis (CF). However, the impact of prolonged hypoxia on airway stem cell function and differentiated epithelium is not well elucidated. Acute hypoxia alters the transcription and translation of many genes, including the CF transmembrane conductance regulator (CFTR). CFTR-targeted therapies (modulators) have not been investigated in vitro under chronic hypoxic conditions found in CF airways in vivo. Nasal epithelial cells (hNECs) derived from eight CF and three non-CF participants were expanded and differentiated at the air-liquid interface (26-30 days) at ambient and 2% oxygen tension (hypoxia). Morphology, global proteomics (LC-MS/MS) and function (barrier integrity, cilia motility and ion transport) of basal stem cells and differentiated cultures were assessed. hNECs expanded at chronic hypoxia, demonstrating epithelial cobblestone morphology and a similar proliferation rate to hNECs expanded at normoxia. Hypoxia-inducible proteins and pathways in stem cells and differentiated cultures were identified. Despite the stem cells' plasticity and adaptation to chronic hypoxia, the differentiated epithelium was significantly thinner with reduced barrier integrity. Stem cell lineage commitment shifted to a more secretory epithelial phenotype. Motile cilia abundance, length, beat frequency and coordination were significantly negatively modulated. Chronic hypoxia reduces the activity of epithelial sodium and CFTR ion channels. CFTR modulator drug response was diminished. Our findings shed light on the molecular pathophysiology of hypoxia and its implications in CF. Targeting hypoxia can be a strategy to augment mucosal function and may provide a means to enhance the efficacy of CFTR modulators.
Collapse
Affiliation(s)
- Sharon L Wong
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Egi Kardia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Abhishek Vijayan
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Bala Umashankar
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Elvis Pandzic
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW 2052, Australia
| | - Adam Jaffe
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW 2052, Australia
| | - Shafagh A Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW 2052, Australia
| |
Collapse
|
30
|
Kreft IC, Hoogendijk AJ, van der Zwaan C, van Alphen FPJ, Boon-Spijker M, Prinsze F, Meijer AB, de Korte D, van den Hurk K, van den Biggelaar M. Mass spectrometry-based analysis on the impact of whole blood donation on the global plasma proteome. Transfusion 2023; 63:564-573. [PMID: 36722460 DOI: 10.1111/trf.17254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Biomonitoring may provide important insights into the impact of a whole blood donation for individual blood donors. STUDY DESIGN AND METHODS Here, we used unbiased mass spectrometry (MS)-based proteomics to assess longitudinal changes in the global plasma proteome, after a single blood donation for new and regular donors. Subsequently, we compared plasma proteomes of 76 male and female whole blood donors, that were grouped based on their ferritin and hemoglobin (Hb) levels. RESULTS The longitudinal analysis showed limited changes in the plasma proteomes of new and regular donors after a whole blood donation during a 180-day follow-up period, apart from a significant short-term decrease in fibronectin. No differences were observed in the plasma proteomes of donors with high versus low Hb and/or ferritin levels. Plasma proteins with the highest variation between and within donors included pregnancy zone protein, which was associated with sex, Alfa 1-antitrypsin which was associated with the allelic variation, and Immunoglobulin D. Coexpression analysis revealed clustering of proteins that are associated with platelet, red cell, and neutrophil signatures as well as with the complement system and immune responses, including a prominent correlating cluster of immunoglobulin M (IgM), immunoglobulin J chain (JCHAIN), and CD5 antigen-like (CD5L). DISCUSSION Overall, our proteomic approach shows that whole blood donation has a limited impact on the plasma proteins measured. Our findings suggest that plasma profiling can be successfully employed to consistently detect proteins and protein complexes that reflect the functionality and integrity of platelets, red blood cells, and immune cells in blood donors and thus highlights its potential use for donor health monitoring.
Collapse
Affiliation(s)
- Iris C Kreft
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Arie J Hoogendijk
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Carmen van der Zwaan
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Floris P J van Alphen
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Mariette Boon-Spijker
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Femmeke Prinsze
- Donor Studies, Department of Donor Medicine Research, Sanquin Research, Amsterdam, The Netherlands
| | - Alexander B Meijer
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, The Netherlands
| | - Dirk de Korte
- Department of Blood Cell Research, Sanquin Research, Amsterdam, The Netherlands
- Department of Product and Process Development, Sanquin Blood Bank, Amsterdam, The Netherlands
| | - Katja van den Hurk
- Donor Studies, Department of Donor Medicine Research, Sanquin Research, Amsterdam, The Netherlands
| | - Maartje van den Biggelaar
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Murray HC, Miller K, Brzozowski JS, Kahl RGS, Smith ND, Humphrey SJ, Dun MD, Verrills NM. Synergistic Targeting of DNA-PK and KIT Signaling Pathways in KIT Mutant Acute Myeloid Leukemia. Mol Cell Proteomics 2023; 22:100503. [PMID: 36682716 PMCID: PMC9986649 DOI: 10.1016/j.mcpro.2023.100503] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 12/19/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common and aggressive form of acute leukemia, with a 5-year survival rate of just 24%. Over a third of all AML patients harbor activating mutations in kinases, such as the receptor tyrosine kinases FLT3 (receptor-type tyrosine-protein kinase FLT3) and KIT (mast/stem cell growth factor receptor kit). FLT3 and KIT mutations are associated with poor clinical outcomes and lower remission rates in response to standard-of-care chemotherapy. We have recently identified that the core kinase of the non-homologous end joining DNA repair pathway, DNA-PK (DNA-dependent protein kinase), is activated downstream of FLT3; and targeting DNA-PK sensitized FLT3-mutant AML cells to standard-of-care therapies. Herein, we investigated DNA-PK as a possible therapeutic vulnerability in KIT mutant AML, using isogenic FDC-P1 mouse myeloid progenitor cell lines transduced with oncogenic mutant KIT (V560G and D816V) or vector control. Targeted quantitative phosphoproteomic profiling identified phosphorylation of DNA-PK in the T2599/T2605/S2608/S2610 cluster in KIT mutant cells, indicative of DNA-PK activation. Accordingly, proliferation assays revealed that KIT mutant FDC-P1 cells were more sensitive to the DNA-PK inhibitors M3814 or NU7441, compared with empty vector controls. DNA-PK inhibition combined with inhibition of KIT signaling using the kinase inhibitors dasatinib or ibrutinib, or the protein phosphatase 2A activators FTY720 or AAL(S), led to synergistic cell death. Global phosphoproteomic analysis of KIT-D816V cells revealed that dasatinib and M3814 single-agent treatments inhibited extracellular signal-regulated kinase and AKT (RAC-alpha serine/threonine-protein kinase)/MTOR (serine/threonine-protein kinase mTOR) activity, with greater inhibition of both pathways when used in combination. Combined dasatinib and M3814 treatment also synergistically inhibited phosphorylation of the transcriptional regulators MYC and MYB. This study provides insight into the oncogenic pathways regulated by DNA-PK beyond its canonical role in DNA repair and demonstrates that DNA-PK is a promising therapeutic target for KIT mutant cancers.
Collapse
Affiliation(s)
- Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Kasey Miller
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Joshua S Brzozowski
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Richard G S Kahl
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Nathan D Smith
- Analytical and Biomolecular Research Facility, Advanced Mass Spectrometry Unit, University of Newcastle, Callaghan, New South Wales, Australia
| | - Sean J Humphrey
- School of Life and Environmental Sciences, and The Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia.
| |
Collapse
|
32
|
The Small-Protein Enrichment Assay (SPEA) for Analysis of Low Abundance Peptide Hormones in Plasma. Methods Mol Biol 2023; 2628:265-276. [PMID: 36781791 DOI: 10.1007/978-1-0716-2978-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
The analysis of low abundance peptide hormones such as insulin in blood plasma is difficult with unbiased mass spectrometry-based proteomics, as they are overshadowed by very abundant proteins such as albumin and IgG. The small-protein enrichment assay (SPEA) can greatly increase detection and discovery of these factors through specific enrichment, which enables fast and efficient analysis of many small-protein factors using a single untargeted LC-MS/MS acquisition. SPEA uses an alcohol-acid-based dissociation and precipitation step, prior to denaturing SEC to remove the large highly abundant plasma proteins leaving only a small-protein fraction. This is followed by an efficient sample preparation and cleanup before either data-dependent acquisition (DDA), or data-independent acquisition (DIA), LC-MS/MS analysis. Combining these workflows increases discovery of proteins, posttranslational modifications (PTMs), and cleavage sites using DDA, while DIA provides consistent analysis useful for large cohort analysis.
Collapse
|
33
|
Kavanagh K, Bashore AC, Davis MA, Jorgensen MJ, McClouth CJ, Beavers DA, Parks JS. Early time-restricted feeding improves high-density lipoprotein amount and function in nonhuman primates, without effects on body composition. Obesity (Silver Spring) 2023; 31 Suppl 1:75-84. [PMID: 36229981 PMCID: PMC9877107 DOI: 10.1002/oby.23564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 08/01/2022] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Time-restricted feeding (TRF), whereby caloric intake is limited to a <12-hour window, is a potential regimen to ameliorate metabolic syndrome and cardiovascular disease (CVD) risk co-occurring with aging and with obesity. Early TRF (eTRF; early morning feeding followed by overnight fasting) times calorie consumption with hepatic circadian gene expression rhythms. Brief TRF trials demonstrate that high-density lipoprotein (HDL) cholesterol increases similar to diet/exercise interventions, which may impart beneficial CVD effects. Using a nonhuman primate (NHP) model, the efficacy of eTRF to raise HDL and increase plasma cholesterol efflux capacity (CEC) (primarily mediated by cholesterol efflux to HDL particles, a process that is inversely associated with CVD risk) was examined. METHODS Adult (8-16 years old, n = 25) and geriatric (≥17 years old) NHPs were randomized to ad libitum feeding or eTRF for 12 months, and relevant body composition, glycemic control, and plasma HDL cholesterol levels and CEC were measured. RESULTS Impaired CEC was found in geriatric NHPs. eTRF induced larger-sized HDL particles, increased HDL apolipoprotein A-1 content, lowered triglyceride concentrations, and increased plasma CEC (primarily to HDL particles) in both adult and geriatric NHPs without changes in glycemic control or body composition. CONCLUSIONS A beneficial effect of eTRF on increasing HDL CEC in NHPs was demonstrated.
Collapse
Affiliation(s)
- Kylie Kavanagh
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- College of Health and MedicineUniversity of TasmaniaHobartAustralia
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Alexander C. Bashore
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Matthew A. Davis
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Matthew J. Jorgensen
- Department of PathologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Christopher J. McClouth
- Department of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Daniel A. Beavers
- Department of Public Health SciencesWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - John S. Parks
- Department of Internal Medicine‐Section on Molecular MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
34
|
Lee JY, Harney D, Kwok J, Larance M, Don AS. The major TMEM106B dementia risk allele affects TMEM106B protein levels and myelin lipid homeostasis in the ageing human hippocampus. RESEARCH SQUARE 2023:rs.3.rs-2392941. [PMID: 36711721 PMCID: PMC9882607 DOI: 10.21203/rs.3.rs-2392941/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background The risk for dementia increases exponentially from the seventh decade of life. Identifying and understanding the biochemical changes that sensitize the ageing brain to neurodegeneration will provide new opportunities for dementia prevention and treatment. This study aimed to determine how ageing and major genetic risk factors for dementia affect the hippocampal proteome and lipidome of neurologically-normal humans over the age of 65. The hippocampus was chosen as it is highly susceptible to atrophy with ageing and in several neurodegenerative diseases. Methods Mass spectrometry-based proteomic and lipidomic analysis of CA1 hippocampus samples from 74 neurologically normal human donors, aged 66-104, was used in combination with multiple regression models and gene set enrichment analysis to identify age-dependent changes in the proteome and lipidome. ANOVA was used to test the effect of major dementia risk alleles in the TMEM106B and APOE genes on the hippocampal proteome and lipidome, adjusting for age, gender, and post-mortem interval. Results Forty proteins were associated with age at false discovery rate-corrected P < 0.05, including proteins that regulate cell adhesion, the cytoskeleton, amino acid and lipid metabolism, and ribosomal subunits. Transmembrane protein 106B (TMEM106B), a regulator of lysosomal and oligodendrocyte function, was regulated with greatest effect size. The increase in TMEM106B levels with age was specific to carriers of the rs1990622-A allele in the TMEM106B gene that is associated with increased risk for frontotemporal dementia, Alzheimer's disease, Parkinson's disease, and hippocampal sclerosis with ageing. Hippocampal lipids were not significantly affected by APOE genotype, however levels of myelin-enriched sulfatides and hexosylceramides were significantly lower, and polyunsaturated phospholipids were higher, in rs1990622-A carriers after controlling for APOE genotype. Conclusions Our study provides the first evidence that TMEM106B protein abundance is increased with brain ageing in humans, and the first evidence that the major TMEM106B dementia risk allele affects brain lipid homeostasis, with a clear effect on myelin lipid content. Our data implies that TMEM106B is one of a growing list of major dementia risk genes that affect glial lipid metabolism.
Collapse
Affiliation(s)
- Jun Yup Lee
- The University of Sydney SMS: The University of Sydney School of Medical Sciences
| | | | - John Kwok
- The University of Sydney SMS: The University of Sydney School of Medical Sciences
| | - Mark Larance
- The University of Sydney SMS: The University of Sydney School of Medical Sciences
| | | |
Collapse
|
35
|
Chen H, Aneman I, Nikolic V, Karadzov Orlic N, Mikovic Z, Stefanovic M, Cakic Z, Jovanovic H, Town SEL, Padula MP, McClements L. Maternal plasma proteome profiling of biomarkers and pathogenic mechanisms of early-onset and late-onset preeclampsia. Sci Rep 2022; 12:19099. [PMID: 36351970 PMCID: PMC9646706 DOI: 10.1038/s41598-022-20658-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 09/16/2022] [Indexed: 11/10/2022] Open
Abstract
Preeclampsia is still the leading cause of morbidity and mortality in pregnancy without a cure. There are two phenotypes of preeclampsia, early-onset (EOPE) and late-onset (LOPE) with poorly defined pathogenic differences. This study aimed to facilitate better understanding of the mechanisms of pathophysiology of EOPE and LOPE, and identify specific biomarkers or therapeutic targets. In this study, we conducted an untargeted, label-free quantitative proteomic analyses of plasma samples from pregnant women with EOPE (n = 17) and LOPE (n = 11), and age, BMI-matched normotensive controls (n = 18). Targeted proteomics approach was also employed to validate a subset of proteins (n = 17). In total, there were 26 and 20 differentially abundant proteins between EOPE or LOPE, and normotensive controls, respectively. A series of angiogenic and inflammatory proteins, including insulin-like growth factor-binding protein 4 (IGFBP4; EOPE: FDR = 0.0030 and LOPE: FDR = 0.00396) and inter-alpha-trypsin inhibitor heavy chain H2-4 (ITIH2-4), were significantly altered in abundance in both phenotypes. Through validation we confirmed that ITIH2 was perturbed only in LOPE (p = 0.005) whereas ITIH3 and ITIH4 were perturbed in both phenotypes (p < 0.05). Overall, lipid metabolism/transport proteins associated with atherosclerosis were highly abundant in LOPE, however, ECM proteins had a more pronounced role in EOPE. The complement cascade and binding and uptake of ligands by scavenger receptors, pathways, were associated with both EOPE and LOPE.
Collapse
Affiliation(s)
- Hao Chen
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Ingrid Aneman
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Valentina Nikolic
- grid.11374.300000 0001 0942 1176Department of Pharmacology and Toxicology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Natasa Karadzov Orlic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia ,grid.7149.b0000 0001 2166 9385Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Zeljko Mikovic
- Department of Gynaecology and Obstetrics, Narodni Front, Belgrade, Serbia ,grid.7149.b0000 0001 2166 9385Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Stefanovic
- grid.11374.300000 0001 0942 1176Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Nis, Nis, Serbia ,grid.418653.d0000 0004 0517 2741Gynaecology and Obstetrics Clinic, Clinical Centre Nis, Nis, Serbia
| | - Zoran Cakic
- Department of Gynaecology and Obstetrics, General Hospital of Leskovac, Leskovac, Serbia
| | - Hristina Jovanovic
- grid.11374.300000 0001 0942 1176Department of Pharmacology and Toxicology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Stephanie E. L. Town
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Matthew P. Padula
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| | - Lana McClements
- grid.117476.20000 0004 1936 7611School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia ,grid.117476.20000 0004 1936 7611Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Ultimo, NSW Australia
| |
Collapse
|
36
|
Knoot CJ, Wantuch PL, Robinson LS, Rosen DA, Scott NE, Harding CM. Discovery and characterization of a new class of O-linking oligosaccharyltransferases from the Moraxellaceae family. Glycobiology 2022; 33:57-74. [PMID: 36239418 PMCID: PMC9829042 DOI: 10.1093/glycob/cwac070] [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: 09/07/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 01/12/2023] Open
Abstract
Bacterial protein glycosylation is commonly mediated by oligosaccharyltransferases (OTases) that transfer oligosaccharides en bloc from preassembled lipid-linked precursors to acceptor proteins. Natively, O-linking OTases usually transfer a single repeat unit of the O-antigen or capsular polysaccharide to the side chains of serine or threonine on acceptor proteins. Three major families of bacterial O-linking OTases have been described: PglL, PglS, and TfpO. TfpO is limited to transferring short oligosaccharides both in its native context and when heterologously expressed in glycoengineered Escherichia coli. On the other hand, PglL and PglS can transfer long-chain polysaccharides when expressed in glycoengineered E. coli. Herein, we describe the discovery and functional characterization of a novel family of bacterial O-linking OTases termed TfpM from Moraxellaceae bacteria. TfpM proteins are similar in size and sequence to TfpO enzymes but can transfer long-chain polysaccharides to acceptor proteins. Phylogenetic analyses demonstrate that TfpM proteins cluster in distinct clades from known bacterial OTases. Using a representative TfpM enzyme from Moraxella osloensis, we determined that TfpM glycosylates a C-terminal threonine of its cognate pilin-like protein and identified the minimal sequon required for glycosylation. We further demonstrated that TfpM has broad substrate tolerance and can transfer diverse glycans including those with glucose, galactose, or 2-N-acetyl sugars at the reducing end. Last, we find that a TfpM-derived bioconjugate is immunogenic and elicits serotype-specific polysaccharide IgG responses in mice. The glycan substrate promiscuity of TfpM and identification of the minimal TfpM sequon renders this enzyme a valuable additional tool for expanding the glycoengineering toolbox.
Collapse
Affiliation(s)
- Cory J Knoot
- Omniose, 4340 Duncan Ave, Suite 202, St. Louis, MO 63110, USA
| | - Paeton L Wantuch
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, 4990 Children’s Place, St. Louis, MO 63110, USA
| | | | - David A Rosen
- Department of Pediatrics, Division of Infectious Diseases, Washington University School of Medicine, 4990 Children’s Place, St. Louis, MO 63110, USA,Department of Molecular Microbiology, Washington University School of Medicine, 660 Euclid Ave, St. Louis, MO 63110, USA
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3010, Australia
| | | |
Collapse
|
37
|
Witjaksono F, Prafiantini E, Rahmawati A. Effect of intermittent fasting 5:2 on body composition and nutritional intake among employees with obesity in Jakarta: a randomized clinical trial. BMC Res Notes 2022; 15:323. [PMID: 36224641 PMCID: PMC9559012 DOI: 10.1186/s13104-022-06209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/10/2022] [Accepted: 09/19/2022] [Indexed: 12/02/2022] Open
Abstract
Objective This study aimed to determine the effect of intermittent fasting 5:2 on body composition in employees with obesity in Jakarta. Results Fifty participants were included; 25 were allocated to the fasting group and 25 to the control group. There was no significant change in fat mass, fat-free mass, skeletal muscle, and BMI (p > 0.05). Significant in-group changes were observed in body weight (p = 0.023) and BMI (p = 0.018) in the fasting group. Dietary intake was similar before and during the intervention. The reduction in macronutrient intake resulted in a statistically significant difference in carbohydrate, protein, and fat intake in the two groups (p < 0.05). Intermittent fasting 5:2 results in weight loss but does not affect fat mass and fat-free mass reductions. None of the between-group differences were clinically relevant. Trial registration: ClinicalTrials.gov with ID: NCT04319133 registered on 24 March 2020. Supplementary information The online version contains supplementary material available at 10.1186/s13104-022-06209-7.
Collapse
Affiliation(s)
- Fiastuti Witjaksono
- Departement of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia.
| | - Erfi Prafiantini
- Departement of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Anni Rahmawati
- Departement of Nutrition, Faculty of Medicine, Universitas Indonesia - Dr. Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|
38
|
Systemic Biomarkers and Unique Pathways in Different Phenotypes of Heart Failure with Preserved Ejection Fraction. Biomolecules 2022; 12:biom12101419. [PMID: 36291628 PMCID: PMC9599828 DOI: 10.3390/biom12101419] [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: 09/07/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for around 50% of all heart failure cases. It is a heterogeneous condition with poorly understood pathogenesis. Here, we aimed to identify unique pathogenic mechanisms in acute and chronic HFpEF and hypertrophic cardiomyopathy (HCM). We performed unbiased, comprehensive proteomic analyses of plasma samples from gender- and BMI-matched patients with acute HFpEF (n = 8), chronic HFpEF (n = 9) and HCM (n = 14) using liquid chromatography–mass spectrometry. Distinct molecular signatures were observed in different HFpEF forms. Clusters of biomarkers differentially abundant between HFpEF forms were predominantly associated with microvascular inflammation. New candidate protein markers were also identified, including leucine-rich alpha-2-glycoprotein 1 (LRG1), serum amyloid A1 (SAA1) and inter-alpha-trypsin inhibitor heavy chain 3 (ITIH3). Our study is the first to apply systematic, quantitative proteomic screening of plasma samples from patients with different subtypes of HFpEF and identify candidate biomarkers for improved management of acute and chronic HFpEF and HCM.
Collapse
|
39
|
Feng J, Tang X, Song L, Zhou Z, Jiang Y, Huang Y. Potential biomarkers and immune characteristics of small bowel adenocarcinoma. Sci Rep 2022; 12:16204. [PMID: 36171259 PMCID: PMC9519963 DOI: 10.1038/s41598-022-20599-5] [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: 04/05/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022] Open
Abstract
Small bowel adenocarcinoma (SBA) is a gastrointestinal malignancy with low incidence but poor prognosis, and its pathogenesis is still unclear. This study aimed to explore potential disease-causing biomarkers of SBA. The gene expression datasets of SBA and normal samples were downloaded from the Gene Expression Omnibus database. First, differential gene expression analysis and weighted gene coexpression network analysis (WGCNA) were performed. Common genes (CGs) were obtained by intersection of differentially expressed genes (DEGs) and optimal modal genes of WGCNA. Subsequently, a protein‒protein interaction network was established to screen hub genes, and target genes were obtained by Lasso regression analysis of hub genes. An SBA risk prediction model was established based on target genes. The prediction accuracy of the model was evaluated by the area under the receiver operating characteristic curve (AUC). The levels of immune cell infiltration and activation of immune pathways were compared between SBA and normal samples using the "ggpubr" and "reshape2" packages. A total of 1058 DEGs were identified. WGCNA showed that the signature gene in the brown module was significantly associated with SBA (p = 7E−17), and 469 CGs were obtained. Four target genes (APOA4, APOB, COL1A2, FN1) were identified and showed excellent prediction of SBA risk (AUC = 0.965). In addition, active dendritic cells and macrophages showed higher infiltration levels in SBA. Meanwhile, the APC_co_stimulation pathway and parainflammation pathway were strongly active in SBA. Four target genes (APOA4, APOB, COL1A2, FN1) may be involved in the pathogenesis of small bowel adenocarcinoma.
Collapse
Affiliation(s)
- Jinggao Feng
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan, China.
| | - Xiayu Tang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan, China
| | - Liusong Song
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan, China
| | - Zhipeng Zhou
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan, China
| | - Yuan Jiang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan, China
| | - Yao Huang
- Department of Gastrointestinal and Anorectal Surgery, The Central Hospital of Yongzhou, No. 151, Xiaoshui West Road, Lingling District, Yongzhou, 425100, Hunan, China
| |
Collapse
|
40
|
Cominetti O, Núñez Galindo A, Corthésy J, Carayol J, Germain N, Galusca B, Estour B, Hager J, Gheldof N, Dayon L. Proteomics reveals unique plasma signatures in constitutional thinness. Proteomics Clin Appl 2022; 16:e2100114. [PMID: 35579096 PMCID: PMC9787820 DOI: 10.1002/prca.202100114] [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: 11/11/2021] [Revised: 04/14/2022] [Accepted: 05/13/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE Studying the plasma proteome of control versus constitutionally thin (CT) individuals, exposed to overfeeding, may give insights into weight-gain management, providing relevant information to the clinical entity of weight-gain resistant CT, and discovering new markers for the condition. EXPERIMENTAL DESIGN Untargeted protein relative quantification of 63 CT and normal-weight individuals was obtained in blood plasma at baseline, during and after an overfeeding challenge using mass spectrometry-based proteomics. RESULTS The plasma proteome of CT subjects presented limited specificity with respect to controls at baseline. Yet, CT showed lower levels of inflammatory C-reactive protein and larger levels of protective insulin-like growth factor-binding protein 2. Differences were more marked during and after overfeeding. CT plasma proteome showed larger magnitude and significance in response, suggesting enhanced "resilience" and more rapid adaptation to changes. Four proteins behaved similarly between CT and controls, while five were regulated in opposite fashion. Ten proteins were differential during overfeeding in CT only (including increased fatty acid-binding protein and glyceraldehyde-3-phosphate dehydrogenase, and decreased apolipoprotein C-II and transferrin receptor protein 1). CONCLUSIONS AND CLINICAL RELEVANCE This first proteomic profiling of a CT cohort reveals different plasma proteomes between CT subjects and controls in a longitudinal clinical trial. Our molecular observations further support that the resistance to weight gain in CT subjects appears predominantly biological. CLINICALTRIALS gov Identifier: NCT02004821.
Collapse
Affiliation(s)
- Ornella Cominetti
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland
| | - Antonio Núñez Galindo
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland
| | - John Corthésy
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland
| | - Jérôme Carayol
- Nestlé Institute of Health SciencesNestlé ResearchLausanneSwitzerland,Present address:
Playtika Switzerland SARue du Port‐Franc 2ALausanne1003Switzerland
| | - Natacha Germain
- Division of EndocrinologyDiabetes, Metabolism and Eating Disorders, CHU St‐EtienneFrance
| | - Bogdan Galusca
- Division of EndocrinologyDiabetes, Metabolism and Eating Disorders, CHU St‐EtienneFrance
| | - Bruno Estour
- Division of EndocrinologyDiabetes, Metabolism and Eating Disorders, CHU St‐EtienneFrance
| | - Jörg Hager
- Nestlé Institute of Health SciencesNestlé ResearchLausanneSwitzerland
| | - Nele Gheldof
- Nestlé Institute of Health SciencesNestlé ResearchLausanneSwitzerland,Present address:
VPA ‐ AVP‐R‐Administration, EPFLBI A2 483, Station 7Lausanne1015Switzerland
| | - Loïc Dayon
- Nestlé Institute of Food Safety & Analytical SciencesNestlé ResearchLausanneSwitzerland,Institut des Sciences et Ingénierie ChimiquesÉcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| |
Collapse
|
41
|
Obaideen K, Abu Shihab KH, Madkour MI, Faris ME. Seven decades of Ramadan intermittent fasting research: Bibliometrics analysis, global trends, and future directions. Diabetes Metab Syndr 2022; 16:102566. [PMID: 35872466 DOI: 10.1016/j.dsx.2022.102566] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/28/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND AND AIMS There is a large body of research focused on various aspects related to Ramadan intermittent fasting (RIF) and human health and disease. This study aimed to quantify the bibliometric data of RIF medical research over the past seven decades and explore these variables qualitatively via text mining analysis. METHODS We used the Scopus search engine to identify published articles related to RIF from inception to December 31, 2021. All types of research articles were included. Scientometric and bibliometric measures were determined using Excel, Biblioshiny, and VOSviewer. This study proposed a bibliometric and text mining method to qualitatively and quantitatively recognize the RIF research trend. RESULTS The Scopus search returned 1915 relevant articles. Most citations pertained to publications from the last two decades, and most publications were original research articles. These publications had received around 27,000 citations, and the 20 most prolific publishing journals had an average h-index of 112.25. More than one-third of all medical publications were in open-access journals. There was a 13-fold increase in medical research on RIF over the past few decades. We identified the 10 most prolific publishing countries, institutes, journals, and authors. We also identified five scientific hotspots of RIF scientific literature, which were: diabetes, metabolic health, public health, physiology, and maternity. CONCLUSION This is the first comprehensive bibliometric analysis of medical research related to RIF. The research gaps identified will shape future research directions and foster collaborative research activities toward enhanced medical nutrition research revolving around RIF.
Collapse
Affiliation(s)
- Khaled Obaideen
- Sustainable Energy & Power Systems Research Centre, RISE, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Katia H Abu Shihab
- Department of Clinical Nutrition and Dietetics, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohamed I Madkour
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - MoezAlIslam E Faris
- Department of Clinical Nutrition and Dietetics, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
42
|
10,12-conjugated linoleic acid supplementation improves HDL composition and function in mice. J Lipid Res 2022; 63:100241. [PMID: 35714730 PMCID: PMC9283942 DOI: 10.1016/j.jlr.2022.100241] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/12/2022] [Accepted: 06/06/2022] [Indexed: 12/31/2022] Open
Abstract
Obesity is associated with inflammation, insulin resistance, and type 2 diabetes, which are major risk factors for CVD. One dietary component of ruminant animal foods, 10,12-conjugated linoleic acid (10,12 CLA), has been shown to promote weight loss in humans. Previous work has shown that 10,12 CLA is atheroprotective in mice by a mechanism that may be distinct from its weight loss effects, but this exact mechanism is unclear. To investigate this, we evaluated HDL composition and function in obese LDL receptor (Ldlr−/−) mice that were losing weight because of 10,12 CLA supplementation or caloric restriction (CR; weight-matched control group) and in an obese control group consuming a high-fat high-sucrose diet. We show that 10,12 CLA-HDL exerted a stronger anti-inflammatory effect than CR- or high-fat high-sucrose-HDL in cultured adipocytes. Furthermore, the 10,12 CLA-HDL particle (HDL-P) concentration was higher, attributed to more medium- and large-sized HDL-Ps. Passive cholesterol efflux capacity of 10,12 CLA-HDL was elevated, as was expression of HDL receptor scavenger receptor class B type 1 in the aortic arch. Murine macrophages treated with 10,12 CLA in vitro exhibited increased expression of cholesterol transporters Abca1 and Abcg1, suggesting increased cholesterol efflux potential of these cells. Finally, proteomics analysis revealed elevated Apoa1 content in 10,12 CLA-HDL-Ps, consistent with a higher particle concentration, and particles were also enriched with alpha-1-antitrypsin, an emerging anti-inflammatory and antiatherosclerotic HDL-associated protein. We conclude that 10,12 CLA may therefore exert its atheroprotective effects by increasing HDL-P concentration, HDL anti-inflammatory potential, and promoting beneficial effects on cholesterol efflux.
Collapse
|
43
|
Harney DJ, Larance M. Annotated Protein Database Using Known Cleavage Sites for Rapid Detection of Secreted Proteins. J Proteome Res 2022; 21:965-974. [DOI: 10.1021/acs.jproteome.1c00806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Dylan J. Harney
- Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, 2006 Sydney, Australia
| | - Mark Larance
- Charles Perkins Centre and School of Life and Environmental Sciences, University of Sydney, 2006 Sydney, Australia
- Charles Perkins Centre and School of Medical Sciences, University of Sydney, 2006 Sydney, Australia
| |
Collapse
|
44
|
Smyth SP, Nixon B, Anderson AL, Murray HC, Martin JH, MacDougall LA, Robertson SA, Skerrett-Byrne DA, Schjenken JE. Elucidation of the protein composition of mouse seminal vesicle fluid. Proteomics 2022; 22:e2100227. [PMID: 35014747 DOI: 10.1002/pmic.202100227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/16/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023]
Abstract
The seminal vesicles are male accessory sex glands that contribute the major portion of the seminal plasma in which mammalian spermatozoa are bathed during ejaculation. In addition to conveying sperm through the ejaculatory duct, seminal vesicle secretions support sperm survival after ejaculation, and influence the female reproductive tract to promote receptivity to pregnancy. Analysis of seminal vesicle fluid (SVF) composition by proteomics has proven challenging, due to its highly biased protein signature with a small subset of dominant proteins and the difficulty of solubilizing this viscous fluid. As such, publicly available proteomic datasets identify only 85 SVF proteins in total. To address this limitation, we report a new preparative methodology involving sequential solubilization of mouse SVF in guanidine hydrochloride, acetone precipitation, and analysis by label-free mass spectrometry. Using this strategy, we identified 126 SVF proteins, including 83 previously undetected in SVF. Members of the seminal vesicle secretory protein family were the most abundant, accounting for 79% of all peptide spectrum matches. Functional analysis identified inflammation and formation of the vaginal plug as the two most prominent biological processes. Other notable processes included modulation of sperm function and regulation of the female reproductive tract immune environment. Together, these findings provide a robust methodological framework for future SVF studies and identify novel proteins with potential to influence both male and female reproductive physiology.
Collapse
Affiliation(s)
- Shannon P Smyth
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Brett Nixon
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Amanda L Anderson
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, NSW, Australia.,Precision Medicine Research Program, Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Jacinta H Martin
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Lily A MacDougall
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - David A Skerrett-Byrne
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| | - John E Schjenken
- School of Environmental and Life Sciences, College of Engineering, Science and Environment, The University of Newcastle, NSW, Australia.,Infertility and Reproduction Research Program, Hunter Medical Research Institute, NSW, Australia
| |
Collapse
|
45
|
Scott NE. Glycopeptide-Centric Approaches for the Characterization of Microbial Glycoproteomes. Methods Mol Biol 2022; 2456:153-171. [PMID: 35612741 DOI: 10.1007/978-1-0716-2124-0_11] [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] [Indexed: 10/18/2022]
Abstract
Protein glycosylation is increasingly recognized as a common class of modifications within microbial species that can shape protein functions and the proteome at large. Due to this, there is an increasing need for robust analytical methods, which allow for the identification and characterization of microbial glycopeptides from proteome samples in a high-throughput manner. Using affinity-based enrichment (either hydrophilicity or antibody-based approaches) glycopeptides can easily be separated from non-glycosylated peptides and analyzed using mass spectrometry. By utilizing multiple mass spectrometry fragmentation approaches and open searching-based bioinformatic techniques, novel glycopeptides can be identified and characterized without prior knowledge of the glycans used for glycosylation. Using these approaches, glycopeptides within samples can rapidly be identified as well as quantified to understand how glycosylation changes in response to stimuli or how changes in glycosylation systems impact the glycoproteome. This chapter outlines a set of robust protocols for the initial preparation, enrichment, and analysis of microbial glycopeptides for both qualitative and quantitative glycoproteomic studies. Using these approaches, glycosylation events can be easily identified by researchers without the need for extensive manual analysis of proteomic datasets.
Collapse
Affiliation(s)
- Nichollas E Scott
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
46
|
Bhatti SI, Mindikoglu AL. The impact of dawn to sunset fasting on immune system and its clinical significance in Covid-19 pandemic. Metabol Open 2021; 13:100162. [PMID: 34977523 PMCID: PMC8713419 DOI: 10.1016/j.metop.2021.100162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
Dawn to sunset fasting, a type of intermittent fasting commonly practiced in the month of Ramadan, requires fasting from dawn to sunset without food or liquid intake. Dawn and dusk are two transition time zones of the day that play a critical role in the human circadian rhythm. Practicing dawn to sunset fasting requires the alignment of mealtimes and wake-sleep times with the human biological dawn and dusk. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) impairs immune cell responses at multiple levels and leads to severe Coronavirus Disease 2019 (COVID-19). It generates high levels of pro-inflammatory cytokines and chemokines, also known as a cytokine storm, leads to mitochondrial dysfunction and generation of excessive amounts of mitochondrial reactive oxygen species, downregulates autophagy to escape detection for unchecked replication, and alters gut microbiome composition. Severe cases of COVID-19 have been associated with several comorbidities that impair immune responses (e.g., obesity, diabetes, malignancy) and blood laboratory abnormalities (e.g., elevated procalcitonin, C-reactive protein, interleukin-6 (IL-6), leukocytosis, lymphopenia). Several studies of dawn to sunset fasting showed anti-inflammatory effect by suppressing several pro-inflammatory cytokines, reducing oxidative stress, inducing a proteome response associated with increased autophagy, remodeling the gut microbiome, and improving the components of metabolic syndrome (e.g., obesity, blood glucose levels, blood pressure, lipids). In conclusion, dawn to sunset fasting has the potential to optimize the immune system function against SARS-CoV-2 during the COVID-19 pandemic as it suppresses chronic inflammation and oxidative stress, improves metabolic profile, and remodels the gut microbiome. This review presents scientific literature related to the effects of dawn to sunset fasting on the immune system. Studies are needed to assess and confirm the potential benefits of dawn to sunset fasting against SARS-CoV-2.
Collapse
Affiliation(s)
- Sundus I Bhatti
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| | - Ayse L Mindikoglu
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA.,Michael E. DeBakey Department of Surgery, Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
47
|
Yau B, Naghiloo S, Diaz-Vegas A, Carr AV, Van Gerwen J, Needham EJ, Jevon D, Chen SY, Hoehn KL, Brandon AE, Macia L, Cooney GJ, Shortreed MR, Smith LM, Keller MP, Thorn P, Larance M, James DE, Humphrey SJ, Kebede MA. Proteomic pathways to metabolic disease and type 2 diabetes in the pancreatic islet. iScience 2021; 24:103099. [PMID: 34622154 PMCID: PMC8479695 DOI: 10.1016/j.isci.2021.103099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/09/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic islets are essential for maintaining physiological blood glucose levels, and declining islet function is a hallmark of type 2 diabetes. We employ mass spectrometry-based proteomics to systematically analyze islets from 9 genetic or diet-induced mouse models representing a broad cross-section of metabolic health. Quantifying the islet proteome to a depth of >11,500 proteins, this study represents the most detailed analysis of mouse islet proteins to date. Our data highlight that the majority of islet proteins are expressed in all strains and diets, but more than half of the proteins vary in expression levels, principally due to genetics. Associating these varied protein expression levels on an individual animal basis with individual phenotypic measures reveals islet mitochondrial function as a major positive indicator of metabolic health regardless of strain. This compendium of strain-specific and dietary changes to mouse islet proteomes represents a comprehensive resource for basic and translational islet cell biology.
Collapse
Affiliation(s)
- Belinda Yau
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Sheyda Naghiloo
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Alexis Diaz-Vegas
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Austin V. Carr
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Julian Van Gerwen
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Elise J. Needham
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Dillon Jevon
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Sing-Young Chen
- Department of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2052, Australia
| | - Kyle L. Hoehn
- Department of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2052, Australia
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Amanda E. Brandon
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Laurance Macia
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Gregory J. Cooney
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | | | - Lloyd M. Smith
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter Thorn
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| | - Mark Larance
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - David E. James
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Sean J. Humphrey
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, NSW, Australia
| | - Melkam A. Kebede
- Discipline of Physiology, School of Medical Sciences, Charles Perkins Centre, University of Sydney, Camperdown 2006, Australia
| |
Collapse
|
48
|
Intermittent Fasting and the Possible Benefits in Obesity, Diabetes, and Multiple Sclerosis: A Systematic Review of Randomized Clinical Trials. Nutrients 2021; 13:nu13093179. [PMID: 34579056 PMCID: PMC8469355 DOI: 10.3390/nu13093179] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
Intermittent fasting has become popular in recent years and is controversially presented as a possible therapeutic adjunct. A bibliographic review of the literature on intermittent fasting and obesity, diabetes, and multiple sclerosis was carried out. The scientific quality of the methodology and the results obtained were evaluated in pairs. Intermittent fasting has beneficial effects on the lipid profile, and it is associated with weight loss and a modification of the distribution of abdominal fat in people with obesity and type 2 diabetes as well as an improvement in the control of glycemic levels. In patients with multiple sclerosis, the data available are too scarce to draw any firm conclusions, but it does appear that intermittent fasting may be a safe and feasible intervention. However, it is necessary to continue investigating its long-term effects since so far, the studies carried out are small and of short duration.
Collapse
|
49
|
Saraswat M, Garapati K, Mun DG, Pandey A. Extensive heterogeneity of glycopeptides in plasma revealed by deep glycoproteomic analysis using size-exclusion chromatography. Mol Omics 2021; 17:939-947. [PMID: 34368825 PMCID: PMC8664156 DOI: 10.1039/d1mo00132a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Several plasma glycoproteins are clinically useful as biomarkers in a variety of diseases. Although thousands of proteins are present in plasma, >95% of the plasma proteome by mass is represented by only 22 proteins. This necessitates strategies to deplete the abundant proteins and enrich other subsets of proteins. Although glycoproteins are abundant in plasma, in routine proteomic analyses, glycopeptides are not often investigated. Traditional methods such as lectin-based enrichment of glycopeptides followed by deglycosylation have helped understand the glycoproteome, but they lack any information about the attached glycans. Here, we apply size-exclusion chromatography (SEC) as a simple strategy to enrich intact N-glycopeptides based on their larger size which achieves broad selectivity regardless of the nature of attached glycans. Using this approach, we identified 1317 N-glycopeptides derived from 266 glycosylation sites on 154 plasma glycoproteins. The deep coverage achieved by this approach was evidenced by extensive heterogeneity that was observed. For instance, 20-100 glycopeptides were observed per protein for the 15 most-glycosylated glycoproteins. Notably, we discovered 615 novel glycopeptides of which 39 glycosylation sites (from 38 glycoproteins) were not included in protein databases such as Uniprot and GlyConnectDB. Finally, we also identified 12 novel glycopeptides containing di-sialic acid, which is a rare glycan epitope. Our results demonstrate the utility of SEC for efficient LC-MS/MS-based deep glycoproteomics analysis of human plasma. Overall, the SEC-based method described here is a simple, rapid and high-throughput strategy for characterization of any glycoproteome.
Collapse
Affiliation(s)
- Mayank Saraswat
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA. and Institute of Bioinformatics, International Technology Park, Bangalore 560066, Karnataka, India and Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Kishore Garapati
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA. and Institute of Bioinformatics, International Technology Park, Bangalore 560066, Karnataka, India and Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India and Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore 560029, India
| | - Dong-Gi Mun
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA. and Institute of Bioinformatics, International Technology Park, Bangalore 560066, Karnataka, India and Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India and Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Hosur Road, Bangalore 560029, India and Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
50
|
Kearney AL, Norris DM, Ghomlaghi M, Kin Lok Wong M, Humphrey SJ, Carroll L, Yang G, Cooke KC, Yang P, Geddes TA, Shin S, Fazakerley DJ, Nguyen LK, James DE, Burchfield JG. Akt phosphorylates insulin receptor substrate to limit PI3K-mediated PIP3 synthesis. eLife 2021; 10:e66942. [PMID: 34253290 PMCID: PMC8277355 DOI: 10.7554/elife.66942] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/30/2021] [Indexed: 01/16/2023] Open
Abstract
The phosphoinositide 3-kinase (PI3K)-Akt network is tightly controlled by feedback mechanisms that regulate signal flow and ensure signal fidelity. A rapid overshoot in insulin-stimulated recruitment of Akt to the plasma membrane has previously been reported, which is indicative of negative feedback operating on acute timescales. Here, we show that Akt itself engages this negative feedback by phosphorylating insulin receptor substrate (IRS) 1 and 2 on a number of residues. Phosphorylation results in the depletion of plasma membrane-localised IRS1/2, reducing the pool available for interaction with the insulin receptor. Together these events limit plasma membrane-associated PI3K and phosphatidylinositol (3,4,5)-trisphosphate (PIP3) synthesis. We identified two Akt-dependent phosphorylation sites in IRS2 at S306 (S303 in mouse) and S577 (S573 in mouse) that are key drivers of this negative feedback. These findings establish a novel mechanism by which the kinase Akt acutely controls PIP3 abundance, through post-translational modification of the IRS scaffold.
Collapse
Affiliation(s)
- Alison L Kearney
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Dougall M Norris
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Milad Ghomlaghi
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash UniversityClaytonAustralia
- Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Martin Kin Lok Wong
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Luke Carroll
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Guang Yang
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Kristen C Cooke
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Pengyi Yang
- Charles Perkins Centre, School of Mathematics and Statistics, University of SydneySydneyAustralia
- Computational Systems Biology Group, Children's Medical Research Institute, University of SydneyWestmeadAustralia
| | - Thomas A Geddes
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
- Computational Systems Biology Group, Children's Medical Research Institute, University of SydneyWestmeadAustralia
| | - Sungyoung Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash UniversityClaytonAustralia
- Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Daniel J Fazakerley
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of CambridgeCambridgeUnited Kingdom
| | - Lan K Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash UniversityClaytonAustralia
- Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
- School of Medical Sciences, University of SydneySydneyAustralia
| | - James G Burchfield
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| |
Collapse
|