1
|
Del Castillo Alferez J, Kooiker A, van Alphen FPJ, van der Zwaan C, Brinkman HJ, Meijers JCM, Meijer AB, van den Biggelaar M, van Duijl TT, SYMPHONY Consortium. Proteolytic signatures of coagulation identified by plasma peptidomics. J Thromb Haemost 2025:S1538-7836(25)00267-3. [PMID: 40286914 DOI: 10.1016/j.jtha.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/12/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Coagulation entails sequential proteolytic events in plasma, ultimately leading to fibrin clot formation. OBJECTIVES In this study, we employed a mass spectrometry-based peptidomics approach to characterize the molecular events of coagulation-induced limited proteolysis. METHODS Citrated plasma from healthy donors was in vitro-coagulated by recalcification combined with the addition of tissue factor (TF) in the absence or presence of hirudin. The formation of endogenous peptide products over time was monitored using a mass spectrometry approach with a de novo algorithm for peptide identification. RESULTS Plasma coagulation resulted in a distinct peptidome enriched with activation peptides of prothrombin and FXIIIA, fibrinopeptides A and B, reactive center loops of protease inhibitors, the bait region of α2-macroglobulin, and additional proteolytic hotspots outside the coagulation system. While thrombin inhibition blocked almost all TF-initiated limited proteolysis, most events were TF concentration-independent, with the exception of prothrombin, fibrinogen, FV, FXIIIA, α2-macroglobulin, protein C inhibitor, complement C3, and plexin domain-containing 2. The order of events of fibrinopeptide A and B formation-prothrombin conversion, FXIIIA activation, and protease inhibitor proteolysis-followed the kinetics of thrombin generation. CONCLUSION Plasma peptidomics of coagulation-initiated limited proteolysis captures peptide products derived from pro- and anticoagulant events and proteolytic signatures beyond the classical coagulation system. We envision that this peptidomics strategy enables the assessment of functional aspects of coagulation in bleeding and thrombotic disorders at the molecular level.
Collapse
Affiliation(s)
| | - Alette Kooiker
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | | | - Carmen van der Zwaan
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Herm-Jan Brinkman
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Joost C M Meijers
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, The Netherlands
| | - Alexander B Meijer
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | | | - Tirsa T van Duijl
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands.
| | | |
Collapse
Collaborators
Emile van den Akker, Wala Al Arashi, Ryanne Arisz, Lieke Baas, Ruben Bierings, Maartje van den Biggelaar, Johan Boender, Anske van der Bom, Mettine Bos, Martijn Brands, Annelien Bredenoord, Laura Bukkems, Lex Burdorf, Jessica Del Castillo Alferez, Michael Cloesmeijer, Marjon Cnossen, Mariëtte Driessens, Jeroen Eikenboom, Karin Fijnvandraat, Kathelijn Fischer, Geertje Goedhart, Tine Goedhart, Samantha Gouw, Rieke van der Graaf, Masja de Haas, Lotte Haverman, Jan Hazelzet, Shannon van Hoorn, Elise Huisman, Nathalie Jansen, Alexander Janssen, Sean de Jong, Sjoerd Koopman, Marieke Kruip, Sebastiaan Laan, Frank Leebeek, Nikki van Leeuwen, Hester Lingsma, Moniek de Maat, Ron Mathôt, Felix van der Meer, Karina Meijer, Sander Meijer, Stephan Meijer, Iris van Moort, Caroline Mussert, Hans Kristian Ploos van Amstel, Suzanne Polinder, Diaz Prameyllawati, Simone Reitsma, Eliza Roest, Lorenzo Romano, Saskia Schols, Roger Schutgens, Rolf Urbanus, Carin Uyl, Jan Voorberg, Huan Zhang, Minka Zivkovic,
Collapse
|
2
|
Kalogeropoulos K, Savickas S, Haack AM, Larsen CA, Mikosiński J, Schoof EM, Smola H, Bundgaard L, Auf dem Keller U. High-Throughput and High-Sensitivity Biomarker Monitoring in Body Fluid by Fast LC SureQuant IS-Targeted Quantitation. Mol Cell Proteomics 2024; 23:100868. [PMID: 39442693 PMCID: PMC11609441 DOI: 10.1016/j.mcpro.2024.100868] [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: 03/26/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024] Open
Abstract
Targeted proteomics methods have been greatly improved and refined over the last decade and are becoming increasingly the method of choice in protein and peptide quantitative assays. Despite the tremendous progress, targeted proteomics assays still suffer from inadequate sensitivity for lower abundant proteins and throughput, especially in complex biological samples. These attributes are essential for establishing targeted proteomics methods at the forefront of clinical use. Here, we report an assay utilizing the SureQuant internal standard-triggered targeted method on a latest generation mass spectrometer coupled with an EvoSep One liquid chromatography platform, which displays high sensitivity and a high throughput of 100 samples per day. We demonstrate the robustness of this method by quantifying proteins spanning six orders of magnitude in human wound fluid exudates, a biological fluid that exhibits sample complexity and composition similar to plasma. Among the targets quantified were low-abundance proteins such at tumor necrosis factor A and interleukin 1-β, highlighting the value of this method in the quantification of trace amounts of invaluable biomarkers that were until recently hardly accessible by targeted proteomics methods. Taken together, this method extends the toolkit of targeted proteomics assays and will help to drive forward mass spectrometry-based proteomics biomarker quantification.
Collapse
Affiliation(s)
| | - Simonas Savickas
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Aleksander M Haack
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Cathrine A Larsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Erwin M Schoof
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Louise Bundgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
3
|
Nanoff C, Yang Q, Hellinger R, Hermann M. Activation of the Calcium-Sensing Receptor by a Subfraction of Amino Acids Contained in Thyroid Drainage Fluid. ACS Pharmacol Transl Sci 2024; 7:1937-1950. [PMID: 39022353 PMCID: PMC11249632 DOI: 10.1021/acsptsci.3c00350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024]
Abstract
Hypoparathyroidism is a common sequela of thyroid surgery; in this study, we aimed at exploring the pathogenesis behind it. The following premises suggest that wound fluid might be a causative agent. (i) Parathyroid hormone secretion is under feedback control by the calcium-sensing receptor, which responds to a diverse array of activating ligands. (ii) Postoperative hypoparathyroidism arises from a secretory deficiency of the parathyroid glands. Even in patients later unaffected by hypoparathyroidism, parathyroid hormone levels drop within hours after surgery. (iii) Wound fluid is bound to enter the tissue around the thyroid bed, where the parathyroid glands are located. Its composition is shaped by a series of proteolytic reactions triggered by wounding. Using thyroid drainage as a surrogate, we addressed the possibility that wound fluid contains compounds activating the calcium-sensing receptor. Drainage fluid ultrafiltrate was found to be rich in amino acids, and on separation by HPLC, compounds activating the calcium-sensing receptor partitioned with hydrophilic matter that rendered buffer acidic. The data show that glutamate and aspartate at millimolar concentrations supported activation of the calcium-sensing receptor, an effect contingent on low pH. In the presence of glutamate/aspartate, protons activated the calcium-sensing receptor with a pH50 of 6.1, and at pH 5, produced maximal activation. This synergistic mode of action was exclusive; glutamine/asparagine did not substitute for the acidic amino acids, nor did Ca2+ substitute for protons. NPS-2143, a negative allosteric receptor modulator completely blocked receptor activation by glutamate/aspartate and by fractionated drainage fluid. Thus, wound fluid may be involved in suppressing parathyroid hormone secretion.
Collapse
Affiliation(s)
- Christian Nanoff
- Centre
for Physiology and Pharmacology, Gaston H. Glock Laboratories for
Exploratory Drug Research, Medizinische
Universität Wien, Währinger Straße 13A, Vienna 1090, Austria
| | - Qiong Yang
- Centre
for Physiology and Pharmacology, Gaston H. Glock Laboratories for
Exploratory Drug Research, Medizinische
Universität Wien, Währinger Straße 13A, Vienna 1090, Austria
| | - Roland Hellinger
- Centre
for Physiology and Pharmacology, Gaston H. Glock Laboratories for
Exploratory Drug Research, Medizinische
Universität Wien, Währinger Straße 13A, Vienna 1090, Austria
| | - Michael Hermann
- Department
of Surgery, Vienna Hospital Association,
Klinik Landstraße, Juchgasse 25, Vienna 1030, Austria
| |
Collapse
|
4
|
Keng JW, Lee SK, Sang SH, Liew KB, Teo SS, Mossadeq WMSM, Chow SC, Akowuah GA, Lee SK, Mai CW, Chew YL. Cassia alata and Its Phytochemicals: A Promising Natural Strategy in Wound Recovery. SCI 2024; 6:34. [DOI: 10.3390/sci6020034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2025] Open
Abstract
Cassia alata, a traditional herb with a global presence, is renowned for its anti-inflammatory, antibacterial, and antifungal properties, making it a go-to remedy for skin ailments. While it has demonstrated wound healing capabilities in both in vitro and in vivo studies, the precise mechanisms remain elusive. This review aims to highlight its key phytochemicals, their effects, and the mechanism of action. The compounds that have been reviewed and discussed include kaempferol, apigenin, quercetin, rhein, and rutin. These polyphenols play important roles in normal and impaired wound healing processes, encompassing hemostasis, inflammation, proliferation, and tissue remodeling.
Collapse
Affiliation(s)
- Jing-Wen Keng
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Sue-Kei Lee
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Sze-Huey Sang
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Kai-Bin Liew
- Faculty of Pharmacy, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya 63000, Malaysia
| | - Swee-Sen Teo
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | | | - Sek-Chuen Chow
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 46150, Malaysia
| | - Gabriel Akyirem Akowuah
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 46150, Malaysia
| | - Siew-Keah Lee
- M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Kajang 43000, Malaysia
| | - Chun-Wai Mai
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
5
|
Li T, Zhao J, Yue Y, Han B, Wang W, Zhang H, Liu Z, Chen Z, Tian X. Preparation of nano-silver containing black phosphorus based on quaternized chitosan hydrogel and evaluating its effect on skin wound healing. Int J Biol Macromol 2024; 268:131950. [PMID: 38685547 DOI: 10.1016/j.ijbiomac.2024.131950] [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: 01/17/2024] [Revised: 04/14/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Hydrogels with favorable biocompatibility and antibacterial properties are essential in postoperative wound hemorrhage care, facilitating rapid wound healing. The present investigation employed electrostatic adsorption of black phosphorus nanosheets (BPNPs) and nano‑silver (AgNPs) to cross-link the protonated amino group NH3+ of quaternized chitosan (QCS) with the hydroxyl group of hyaluronic acid (HA). The electrostatic interaction between the two groups resulted in the formation of a three-dimensional gel network structure. Additionally, the hydrogel containing AgNPs deposited onto BPNPs was assessed for its antibacterial properties and effects on wound healing. Hydrogel demonstrated an outstanding drug-loading capacity and could be employed for wound closure. AgNPs loaded on the BPNPs released silver ions and exhibited potent antibacterial properties when exposed to 808 nm near-infrared (NIR) radiation. The ability of the hydrogel to promote wound healing in an acute wound model was further evaluated. The BPNPs were combined with HA and QCS in the aforementioned hydrogel system to improve adhesion, combine the photothermal and antibacterial properties of the BPNPs, and promote wound healing. Therefore, the reported hydrogels displayed excellent biocompatibility and hold significant potential for application in the field of tissue engineering for skin wound treatment.
Collapse
Affiliation(s)
- Tianjiao Li
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Jiaqi Zhao
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Yajuan Yue
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Bo Han
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Wenjuan Wang
- Sinopharm Xinjiang Pharmaceutical Co. LTD, Urumqi 830032, China
| | - Han Zhang
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Zhiyong Liu
- College of Chemistry and Chemical Engineering, Shihezi University, Shihezi 832003, China.
| | - Zhenyang Chen
- Sinopharm Xinjiang Pharmaceutical Co. LTD, Urumqi 830032, China.
| | - Xing Tian
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China; Sinopharm Xinjiang Pharmaceutical Co. LTD, Urumqi 830032, China.
| |
Collapse
|
6
|
Kollet O, Das A, Karamanos N, Auf dem Keller U, Sagi I. Redefining metalloproteases specificity through network proteolysis. Trends Mol Med 2024; 30:147-163. [PMID: 38036391 PMCID: PMC11004056 DOI: 10.1016/j.molmed.2023.11.001] [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: 09/09/2023] [Revised: 10/18/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023]
Abstract
Proteolytic processes on cell surfaces and extracellular matrix (ECM) sustain cell behavior and tissue integrity in health and disease. Matrix metalloproteases (MMPs) and a disintegrin and metalloproteases (ADAMs) remodel cell microenvironments through irreversible proteolysis of ECM proteins and cell surface bioactive molecules. Pan-MMP inhibitors in inflammation and cancer clinical trials have encountered challenges due to promiscuous activities of MMPs. Systems biology advances revealed that MMPs initiate multifactorial proteolytic cascades, creating new substrates, activating or suppressing other MMPs, and generating signaling molecules. This review highlights the intricate network that underscores the role of MMPs beyond individual substrate-enzyme activities. Gaining insight into MMP function and tissue specificity is crucial for developing effective drug discovery strategies and novel therapeutics. This requires considering the dynamic cellular processes and consequences of network proteolysis.
Collapse
Affiliation(s)
- Orit Kollet
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Alakesh Das
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Nikos Karamanos
- University of Patras, Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, Patras, Greece
| | - Ulrich Auf dem Keller
- Technical University of Denmark, Department of Biotechnology and Biomedicine, Lyngby, Denmark
| | - Irit Sagi
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel.
| |
Collapse
|
7
|
Naba A. 10 years of extracellular matrix proteomics: Accomplishments, challenges, and future perspectives. Mol Cell Proteomics 2023; 22:100528. [PMID: 36918099 PMCID: PMC10152135 DOI: 10.1016/j.mcpro.2023.100528] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
The extracellular matrix (ECM) is a complex assembly of hundreds of proteins forming the architectural scaffold of multicellular organisms. In addition to its structural role, the ECM conveys signals orchestrating cellular phenotypes. Alterations of ECM composition, abundance, structure, or mechanics, have been linked to diseases and disorders affecting all physiological systems, including fibrosis and cancer. Deciphering the protein composition of the ECM and how it changes in pathophysiological contexts is thus the first step toward understanding the roles of the ECM in health and disease and toward the development of therapeutic strategies to correct disease-causing ECM alterations. Potentially, the ECM also represents a vast, yet untapped reservoir of disease biomarkers. ECM proteins are characterized by unique biochemical properties that have hindered their study: they are large, heavily and uniquely post-translationally modified, and highly insoluble. Overcoming these challenges, we and others have devised mass-spectrometry-based proteomic approaches to define the ECM composition, or "matrisome", of tissues. This review provides a historical overview of ECM proteomics research and presents the latest advances that now allow the profiling of the ECM of healthy and diseased tissues. The second part highlights recent examples illustrating how ECM proteomics has emerged as a powerful discovery pipeline to identify prognostic cancer biomarkers. The third part discusses remaining challenges limiting our ability to translate findings to clinical application and proposes approaches to overcome them. Last, the review introduces readers to resources available to facilitate the interpretation of ECM proteomics datasets. The ECM was once thought to be impenetrable. MS-based proteomics has proven to be a powerful tool to decode the ECM. In light of the progress made over the past decade, there are reasons to believe that the in-depth exploration of the matrisome is within reach and that we may soon witness the first translational application of ECM proteomics.
Collapse
Affiliation(s)
- Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA; University of Illinois Cancer Center, Chicago, IL 60612, USA.
| |
Collapse
|
8
|
Kalogeropoulos K, Bundgaard L, Auf dem Keller U. Sensitive and High-Throughput Exploration of Protein N-Termini by TMT-TAILS N-Terminomics. Methods Mol Biol 2023; 2718:111-135. [PMID: 37665457 DOI: 10.1007/978-1-0716-3457-8_7] [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: 09/05/2023]
Abstract
Terminal amine isotopic labeling of substrates (TAILS) is a sensitive and robust quantitative mass spectrometry (MS)-based proteomics method used for the characterization of physiological or proteolytically processed protein N-termini, as well as other N-terminal posttranslational modifications (PTMs). TAILS is a well-established, high-throughput, negative enrichment workflow that enables system-wide exploration of N-terminomes independent of sample complexity. TAILS makes use of amine reactivity of free N-termini and a highly efficient aldehyde-functionalized polymer to deplete internal peptides generated after proteolytic digestion during sample preparation. Thereby, it enriches for natural N-termini, allowing for unbiased and complete investigation of differential proteolysis, protease substrate discovery, and analysis of N-terminal PTMs. In this chapter, we provide a state-of-the-art protocol, with detailed steps in all parts of the TAILS sample preparation, MS analysis, and post-processing of acquired data.
Collapse
Affiliation(s)
| | - Louise Bundgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
9
|
Haack AM, Overall CM, Auf dem Keller U. Degradomics technologies in matrisome exploration. Matrix Biol 2022; 114:1-17. [PMID: 36280126 DOI: 10.1016/j.matbio.2022.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/05/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Consisting of a defined set of extracellular proteins secreted from resident cells and with minor contributions from serum proteins, the extracellular matrix (ECM) is an essential component of all tissues. Maintaining tissue homeostasis, structural support and cellular control through cell-ECM communication, the ECM has come to be viewed as not just a passive structural entity but rather as a dynamic signaling conduit between cells and the extracellular compartment. Proteins and their cleavage products mediate this communication, and aberrant signaling, either directly or indirectly distorting the ECM, results in pathological conditions including cancer, inflammation, fibrosis, and neurodegenerative diseases. Characterization of ECM components, the matrisome, the extracellular environment and their changes in disease is therefore of importance to understand and mitigate by developing novel therapeutics. Liquid chromatography-mass spectrometry (LC-MS) proteomics has been integral to protein and proteome research for decades and long superseded the obsolescent gel-based approaches. A continuous effort has ensured progress with increased sensitivity and throughput as more advanced equipment has been developed hand in hand with specialized enrichment, detection, and identification methods. Part of this effort lies in the field of degradomics, a branch of proteomics focused on discovering novel protease substrates by identification of protease-generated neo-N termini, the N-terminome, and characterizing the responsible protease networks. Various methods to do so have been developed, some specialized for specific tissue types, others for particular proteases, throughput, or ease of use. This review aims to provide an overview of the state-of-the-art proteomics techniques that have successfully been recently utilized to characterize proteolytic cleavages in the ECM and thereby guided new research and understanding of the ECM and matrisome biology.
Collapse
Affiliation(s)
- Aleksander M Haack
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, DK-2800 Kongens Lyngby, Denmark
| | - Christopher M Overall
- Department of Biochemistry and Molecular Biology, Department of Oral Biological and Medical Sciences, Centre for Blood Research, University of British Columbia, 4.401 Life Sciences Institute, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
10
|
Lan X, Guo J, Li J, Qiang W, Du L, Zhou T, Li X, Wu Z, Yang J. Xanthan gum/oil body-microgel emulsions with enhanced transdermal absorption for accelerating wound healing. Int J Biol Macromol 2022; 222:1376-1387. [PMID: 36126813 DOI: 10.1016/j.ijbiomac.2022.09.134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/21/2022]
Abstract
The oil body comprises lipid droplets surrounded by a surface embedded with oil body-related proteins. To form a drug delivery system, an oleosin can be fused with foreign proteins and bound to the oil body surface. Here, safflower oil bodies carrying oleosin-human epidermal growth factor (hEGF) were mixed with xanthan gum to form self-assembled polymers, referred as an oil body microgel emulsion (OBEME) without any chemical crosslinking agent. The physicochemical properties of OBEME were evaluated and compared with those of natural lipid droplets. The electrostatic interaction between xanthan gum and oil bodies prevents excessive cross-linking and forms a uniform network structure. The basic properties of OBEME were characterized by scanning electron microscopy, cryo-scanning electron microscopy, rheology, and thermogravimetric analysis. The OBEME is an interconnected network and presents a smooth surface without any pores; it remains stable at room temperature for 90 days, and is not affected by low-speed centrifugation and repeated freeze-thaw cycles as indicated by particle size, potential, and fluorescence microscopy analyses. The OBEME enlarges the skin tissue gap, enhances skin permeability, and shows a good slow-release effect in the transdermal absorption test in vivo. It demonstrates a wound healing effect; further, it regulates the inflammatory response of full-layer skin wounds in rats, as well as accelerate angiogenesis, and promote re-epithelialization and remodeling. The OBEME as a bioactive molecule-carbohydrate complex can effectively accelerate skin regeneration and has great translational potential to provide low-cost alternative wound care treatments.
Collapse
Affiliation(s)
- Xinxin Lan
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
| | - Jinnan Guo
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
| | - Jing Li
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
| | - Weidong Qiang
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
| | - Linna Du
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
| | - Tingting Zhou
- Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun 130000, China
| | - Xiaokun Li
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China
| | - Zhuofu Wu
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China.
| | - Jing Yang
- College of Life Science, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
11
|
Kirketerp-Møller K, Doerfler P, Schoefmann N, Wolff-Winiski B, Niazi O, Pless V, Karlsmark T, Ågren MS. Biomarkers of Skin Graft Healing in Venous Leg Ulcers. Acta Derm Venereol 2022; 102:adv00749. [PMID: 35604238 PMCID: PMC9574695 DOI: 10.2340/actadv.v102.201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
There is a need for biomarkers that predict the success of transplantation of venous leg ulcers (with autologous split-thickness skin grafts). The primary objective of this exploratory study was to investigate the association between split-thickness skin graft healing in venous leg ulcers and candidate wound fluid biomarkers representing inflammatory cell and endogenous proteinase activities, and bioactivity. A secondary objective was to compare biomarker levels of the 17 venous leg ulcers with sterile split-thickness skin graft donor-site wounds in another 10 patients with venous leg ulcers. Wound fluids were collected for 24 h using a validated method. The concentration of pre-operative matrix metalloproteinase-9 in wound fluid was higher in venous leg ulcers showing good healing (n = 10) than in venous leg ulcers showing poor healing (n = 7) 12 weeks after transplantation with meshed split-thickness skin grafts. The diagnostic value of matrix metalloproteinase-9 was good according to receiver-operating characteristic curve analysis. Matrix metalloproteinase activity in wound fluids from split-thickness skin graft donor-site wounds increased as a function of time and healing, but was still lower than matrix metalloproteinase activity in venous leg ulcer wound fluids, which showed increased levels of most biomarkers except for matrix metalloproteinase-9 and matrix metalloproteinase-2. In conclusion, wound fluid matrix metalloproteinase-9 concentration is a potential predictive biomarker of split-thickness skin graft healing in venous leg ulcers.
Collapse
Affiliation(s)
- Klaus Kirketerp-Møller
- Copenhagen Wound Healing Center and Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Kalogeropoulos K, Savickas S, Haack AM, Larsen CA, Mikosiński J, Schoof EM, Smola H, Bundgaard L, Auf dem Keller U. WITHDRAWN: High-throughput and high-sensitivity biomarker monitoring in body fluid by FAIMS-enhanced fast LC SureQuant™ IS targeted quantitation. Mol Cell Proteomics 2022:100251. [PMID: 35644345 DOI: 10.1016/j.mcpro.2022.100251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022] Open
Affiliation(s)
- Konstantinos Kalogeropoulos
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark
| | - Simonas Savickas
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark
| | - Aleksander M Haack
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark
| | - Cathrine A Larsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark
| | - Jacek Mikosiński
- Poradnia Chorób Naczyń Obwodowych "MIKOMED", Ul. Pługowa 51/53, 94-238 Łódź, Poland
| | - Erwin M Schoof
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark
| | - Hans Smola
- Paul Hartmann AG, Paul-Hartmann-Straße 12, 89522 Heidenheim an der Brenz, Germany
| | - Louise Bundgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark.
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, Kgs. Lyngby, 2800, Denmark.
| |
Collapse
|
13
|
van der Plas MJA, Cai J, Petrlova J, Saleh K, Kjellström S, Schmidtchen A. Method development and characterisation of the low-molecular-weight peptidome of human wound fluids. eLife 2021; 10:e66876. [PMID: 34227939 PMCID: PMC8260221 DOI: 10.7554/elife.66876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/03/2021] [Indexed: 01/13/2023] Open
Abstract
The normal wound healing process is characterised by proteolytic events, whereas infection results in dysfunctional activations by endogenous and bacterial proteases. Peptides, downstream reporters of these proteolytic actions, could therefore serve as a promising tool for diagnosis of wounds. Using mass-spectrometry analyses, we here for the first time characterise the peptidome of human wound fluids. Sterile post-surgical wound fluids were found to contain a high degree of peptides in comparison to human plasma. Analyses of the peptidome from uninfected healing wounds and Staphylococcus aureus -infected wounds identify unique peptide patterns of various proteins, including coagulation and complement factors, proteases, and antiproteinases. Together, the work defines a workflow for analysis of peptides derived from wound fluids and demonstrates a proof-of-concept that such fluids can be used for analysis of qualitative differences of peptide patterns from larger patient cohorts, providing potential biomarkers for wound healing and infection.
Collapse
Affiliation(s)
- Mariena JA van der Plas
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of CopenhagenCopenhagenDenmark
| | - Jun Cai
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of CopenhagenCopenhagenDenmark
| | - Jitka Petrlova
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
| | - Karim Saleh
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
- Dermatology, Skane University HospitalLundSweden
| | - Sven Kjellström
- Division of Mass Spectrometry, Department of Clinical Sciences, Lund UniversityLundSweden
| | - Artur Schmidtchen
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
- Dermatology, Skane University HospitalLundSweden
- Copenhagen Wound Healing Center, Bispebjerg Hospital, Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
14
|
Frey AM, Chaput D, Shaw LN. Insight into the human pathodegradome of the V8 protease from Staphylococcus aureus. Cell Rep 2021; 35:108930. [PMID: 33826899 PMCID: PMC8054439 DOI: 10.1016/j.celrep.2021.108930] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/03/2020] [Accepted: 03/11/2021] [Indexed: 12/03/2022] Open
Abstract
Staphylococcus aureus possesses ten extracellular proteases with mostly unknown targets in the human proteome. To assist with bacterial protease target discovery, we have applied and compared two N-terminomics methods to investigate cleavage of human serum proteins by S. aureus V8 protease, discovering 85 host-protein targets. Among these are virulence-relevant complement, iron sequestration, clotting cascade, and host protease inhibitor proteins. Protein cleavage sites have been identified, providing insight into the disruption of host protein function by V8. Complement proteins are cleaved within peptidase and sushi domains, and host protease inhibitors are cleaved outside their protease-trapping motifs. Our data highlight the potential for further application of N-terminomics in discovery of bacterial protease substrates in other host niches and provide omics-scale insight into the role of the V8 protease in S. aureus pathogenesis. S. aureus-secreted proteases are central to disease causation, but the discovery of their host substrates has been limited. Frey et al. use N-terminomic approaches to uncover human serum targets of the V8 protease that are from virulence-relevant processes such as the host inflammatory network and nutrient sequestration.
Collapse
Affiliation(s)
- Andrew Michael Frey
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Lindsey Neil Shaw
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
15
|
Ozols M, Eckersley A, Platt CI, Stewart-McGuinness C, Hibbert SA, Revote J, Li F, Griffiths CEM, Watson REB, Song J, Bell M, Sherratt MJ. Predicting Proteolysis in Complex Proteomes Using Deep Learning. Int J Mol Sci 2021; 22:3071. [PMID: 33803033 PMCID: PMC8002881 DOI: 10.3390/ijms22063071] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Both protease- and reactive oxygen species (ROS)-mediated proteolysis are thought to be key effectors of tissue remodeling. We have previously shown that comparison of amino acid composition can predict the differential susceptibilities of proteins to photo-oxidation. However, predicting protein susceptibility to endogenous proteases remains challenging. Here, we aim to develop bioinformatics tools to (i) predict cleavage site locations (and hence putative protein susceptibilities) and (ii) compare the predicted vulnerabilities of skin proteins to protease- and ROS-mediated proteolysis. The first goal of this study was to experimentally evaluate the ability of existing protease cleavage site prediction models (PROSPER and DeepCleave) to identify experimentally determined MMP9 cleavage sites in two purified proteins and in a complex human dermal fibroblast-derived extracellular matrix (ECM) proteome. We subsequently developed deep bidirectional recurrent neural network (BRNN) models to predict cleavage sites for 14 tissue proteases. The predictions of the new models were tested against experimental datasets and combined with amino acid composition analysis (to predict ultraviolet radiation (UVR)/ROS susceptibility) in a new web app: the Manchester proteome susceptibility calculator (MPSC). The BRNN models performed better in predicting cleavage sites in native dermal ECM proteins than existing models (DeepCleave and PROSPER), and application of MPSC to the skin proteome suggests that: compared with the elastic fiber network, fibrillar collagens may be susceptible primarily to protease-mediated proteolysis. We also identify additional putative targets of oxidative damage (dermatopontin, fibulins and defensins) and protease action (laminins and nidogen). MPSC has the potential to identify potential targets of proteolysis in disparate tissues and disease states.
Collapse
Affiliation(s)
- Matiss Ozols
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (A.E.); (C.I.P.); (C.S.-M.); (S.A.H.)
| | - Alexander Eckersley
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (A.E.); (C.I.P.); (C.S.-M.); (S.A.H.)
| | - Christopher I. Platt
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (A.E.); (C.I.P.); (C.S.-M.); (S.A.H.)
| | - Callum Stewart-McGuinness
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (A.E.); (C.I.P.); (C.S.-M.); (S.A.H.)
| | - Sarah A. Hibbert
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (A.E.); (C.I.P.); (C.S.-M.); (S.A.H.)
| | - Jerico Revote
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC 3800, Australia;
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia;
| | - Fuyi Li
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3800, Australia;
| | - Christopher E. M. Griffiths
- Centre for Dermatology Research, Faculty of Biology, Medicine and Health, and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (C.E.M.G.); (R.E.B.W.)
- NIHR Manchester Biomedical Research Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Rachel E. B. Watson
- Centre for Dermatology Research, Faculty of Biology, Medicine and Health, and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (C.E.M.G.); (R.E.B.W.)
- NIHR Manchester Biomedical Research Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Jiangning Song
- Infection and Immunity Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia;
- Monash Centre for Data Science, Faculty of Information Technology, Monash University, Melbourne, VIC 3800, Australia
| | - Mike Bell
- Research and Development, Walgreens Boots Alliance, Thane Road, Nottingham NG90 1BS, UK;
| | - Michael J. Sherratt
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (A.E.); (C.I.P.); (C.S.-M.); (S.A.H.)
| |
Collapse
|
16
|
Weiss SAI, Rehm SRT, Perera NC, Biniossek ML, Schilling O, Jenne DE. Origin and Expansion of the Serine Protease Repertoire in the Myelomonocyte Lineage. Int J Mol Sci 2021; 22:ijms22041658. [PMID: 33562184 PMCID: PMC7914634 DOI: 10.3390/ijms22041658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/26/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
The deepest evolutionary branches of the trypsin/chymotrypsin family of serine proteases are represented by the digestive enzymes of the gastrointestinal tract and the multi-domain proteases of the blood coagulation and complement system. Similar to the very old digestive system, highly diverse cleavage specificities emerged in various cell lineages of the immune defense system during vertebrate evolution. The four neutrophil serine proteases (NSPs) expressed in the myelomonocyte lineage, neutrophil elastase, proteinase 3, cathepsin G, and neutrophil serine protease 4, collectively display a broad repertoire of (S1) specificities. The origin of NSPs can be traced back to a circulating liver-derived trypsin-like protease, the complement factor D ancestor, whose activity is tightly controlled by substrate-induced activation and TNFα-induced locally upregulated protein secretion. However, the present-day descendants are produced and converted to mature enzymes in precursor cells of the bone marrow and are safely sequestered in granules of circulating neutrophils. The potential site and duration of action of these cell-associated serine proteases are tightly controlled by the recruitment and activation of neutrophils, by stimulus-dependent regulated secretion of the granules, and by various soluble inhibitors in plasma, interstitial fluids, and in the inflammatory exudate. An extraordinary dynamic range and acceleration of immediate defense responses have been achieved by exploiting the high structural plasticity of the trypsin fold.
Collapse
Affiliation(s)
- Stefanie A. I. Weiss
- Comprehensive Pneumology Center (CPC-M), Institute of Lung Biology and Disease (iLBD) Helmholtz Zentrum München and University Hospital of the Ludwig-Maximilians University (LMU), 81377 Munich, Germany; (S.A.I.W.); (S.R.T.R.)
| | - Salome R. T. Rehm
- Comprehensive Pneumology Center (CPC-M), Institute of Lung Biology and Disease (iLBD) Helmholtz Zentrum München and University Hospital of the Ludwig-Maximilians University (LMU), 81377 Munich, Germany; (S.A.I.W.); (S.R.T.R.)
| | | | - Martin L. Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany;
| | - Oliver Schilling
- Institute of Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany;
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dieter E. Jenne
- Comprehensive Pneumology Center (CPC-M), Institute of Lung Biology and Disease (iLBD) Helmholtz Zentrum München and University Hospital of the Ludwig-Maximilians University (LMU), 81377 Munich, Germany; (S.A.I.W.); (S.R.T.R.)
- Max Planck Institute of Neurobiology, 82152 Planegg-Martinsried, Germany
- Correspondence:
| |
Collapse
|
17
|
New strategies to identify protease substrates. Curr Opin Chem Biol 2020; 60:89-96. [PMID: 33220627 DOI: 10.1016/j.cbpa.2020.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/27/2020] [Indexed: 12/31/2022]
Abstract
Proteome dynamics is governed by transcription, translation, and post-translational modifications. Limited proteolysis is an irreversible post-translational modification that generates multiple but unique proteoforms from almost every native protein. Elucidating these proteoforms and understanding their dynamics at a system-wide level is of utmost importance because uncontrolled proteolytic cleavages correlate with many pathologies. Mass spectrometry-based degradomics has revolutionized protease research and invented workflows for global identification of protease substrates with resolution down to precise cleavage sites. In this review, we provide an overview of current strategies in protease substrate degradomics and introduce the concept of workflow, mass spectrometry-based and in silico enrichment of protein termini with the perspective of full deconvolution of digital proteome maps for precision medicine, and degradomics biomarker diagnostics.
Collapse
|
18
|
Hofsetz E, Demir F, Szczepanowska K, Kukat A, Kizhakkedathu JN, Trifunovic A, Huesgen PF. The Mouse Heart Mitochondria N Terminome Provides Insights into ClpXP-Mediated Proteolysis. Mol Cell Proteomics 2020; 19:1330-1345. [PMID: 32467259 PMCID: PMC8014998 DOI: 10.1074/mcp.ra120.002082] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/24/2020] [Indexed: 12/29/2022] Open
Abstract
The mammalian mitochondrial proteome consists of more than 1100 annotated proteins and their proteostasis is regulated by only a few ATP-dependent protease complexes. Technical advances in protein mass spectrometry allowed for detailed description of the mitoproteome from different species and tissues and their changes under specific conditions. However, protease-substrate relations within mitochondria are still poorly understood. Here, we combined Terminal Amine Isotope Labeling of Substrates (TAILS) N termini profiling of heart mitochondria proteomes isolated from wild type and Clpp-/- mice with a classical substrate-trapping screen using FLAG-tagged proteolytically active and inactive CLPP variants to identify new ClpXP substrates in mammalian mitochondria. Using TAILS, we identified N termini of more than 200 mitochondrial proteins. Expected N termini confirmed sequence determinants for mitochondrial targeting signal (MTS) cleavage and subsequent N-terminal processing after import, but the majority were protease-generated neo-N termini mapping to positions within the proteins. Quantitative comparison revealed widespread changes in protein processing patterns, including both strong increases or decreases in the abundance of specific neo-N termini, as well as an overall increase in the abundance of protease-generated neo-N termini in CLPP-deficient mitochondria that indicated altered mitochondrial proteostasis. Based on the combination of altered processing patterns, protein accumulation and stabilization in CLPP-deficient mice and interaction with CLPP, we identified OAT, HSPA9 and POLDIP2 and as novel bona fide ClpXP substrates. Finally, we propose that ClpXP participates in the cooperative degradation of UQCRC1. Together, our data provide the first landscape of the heart mitochondria N terminome and give further insights into regulatory and assisted proteolysis mediated by ClpXP.
Collapse
Affiliation(s)
- Eduard Hofsetz
- Institute for Mitochondrial Diseases and Aging at CECAD Research Centre, and Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany, Medical Faculty and University Hospital, University of Cologne, Cologne, Germany
| | - Fatih Demir
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Germany
| | - Karolina Szczepanowska
- Institute for Mitochondrial Diseases and Aging at CECAD Research Centre, and Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany, Medical Faculty and University Hospital, University of Cologne, Cologne, Germany
| | - Alexandra Kukat
- Institute for Mitochondrial Diseases and Aging at CECAD Research Centre, and Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany, Medical Faculty and University Hospital, University of Cologne, Cologne, Germany
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, School of Biomedical Engineering, Department of Pathology & Laboratory Medicine, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aleksandra Trifunovic
- Institute for Mitochondrial Diseases and Aging at CECAD Research Centre, and Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany, Medical Faculty and University Hospital, University of Cologne, Cologne, Germany.
| | - Pitter F Huesgen
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne, Germany, Medical Faculty and University Hospital, University of Cologne, Cologne, Germany; Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Germany; Institute for Biochemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
19
|
Dengjel J, Bruckner-Tuderman L, Nyström A. Skin proteomics - analysis of the extracellular matrix in health and disease. Expert Rev Proteomics 2020; 17:377-391. [PMID: 32552150 DOI: 10.1080/14789450.2020.1773261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The skin protects the human body from external insults and regulates water and temperature homeostasis. A highly developed extracellular matrix (ECM) supports the skin and instructs its cell functions. Reduced functionality of the ECM is often associated with skin diseases that cause physical impairment and also have implications on social interactions and quality of life of affected individuals. AREAS COVERED With a focus on the skin ECM we discuss how mass spectrometry (MS)-based proteomic approaches first contributed to establishing skin protein inventories and then facilitated elucidation of molecular functions and disease mechanisms. EXPERT OPINION MS-based proteomic approaches have significantly contributed to our understanding of skin pathophysiology, but also revealed the challenges in assessing the skin ECM. The numerous posttranslational modifications of ECM proteins, like glycosylation, crosslinking, oxidation, and proteolytic maturation in disease settings can be difficult to tackle and remain understudied. Increased ease of handling of LC-MS/MS systems and automated/streamlined data analysis pipelines together with the accompanying increased usage of LC-MS/MS approaches will ensure that in the coming years MS-based proteomic approaches will continue to play a vital part in skin disease research. They will facilitate the elucidation of molecular disease mechanisms and, ultimately, identification of new druggable targets.
Collapse
Affiliation(s)
- Jörn Dengjel
- Department of Biology, University of Fribourg , Fribourg, Switzerland
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg , Freiburg, University of Freiburg, Freiburg, Germany Germany
| | - Alexander Nyström
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg , Freiburg, University of Freiburg, Freiburg, Germany Germany
| |
Collapse
|
20
|
Savickas S, Kastl P, auf dem Keller U. Combinatorial degradomics: Precision tools to unveil proteolytic processes in biological systems. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140392. [DOI: 10.1016/j.bbapap.2020.140392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/28/2022]
|
21
|
Bundgaard L, Savickas S, Auf dem Keller U. Mapping the N-Terminome in Tissue Biopsies by PCT-TAILS. Methods Mol Biol 2020; 2043:285-296. [PMID: 31463921 DOI: 10.1007/978-1-4939-9698-8_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Proteases play pivotal roles in multiple biological processes in all living organisms and are tightly regulated under normal conditions, but alterations in the proteolytic system and uncontrolled protease activity result in multiple pathological conditions. A disease will most often be defined by an ensemble of cleavage events-a proteolytic signature, thus the system-wide study of protease substrates has gained significant attention and identification of disease specific clusters of protease substrates holds great promise as targets for diagnostics and therapy.In this chapter we describe a method that enables fast and reproducible analysis of protease substrates and proteolytic products in an amount of tissue less than the quantity obtained by a standard biopsy. The method combines tissue disruption and protein extraction by pressure cycling technology (PCT), N-terminal enrichment by tandem mass tag (TMT)-terminal amine isotopic labeling of substrates (TAILS), peptide analysis by mass spectrometry (MS), and a general pipeline for interpretation of the data.
Collapse
Affiliation(s)
- Louise Bundgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Simonas Savickas
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
22
|
Ma H, Li R, Di X, Jin X, Wang Y, Lai B, Shi C, Ji M, Zhu X, Wang K. ITRAQ-based proteomic analysis reveals possible target-related proteins in human adrenocortical adenomas. BMC Genomics 2019; 20:655. [PMID: 31419939 PMCID: PMC6697928 DOI: 10.1186/s12864-019-6030-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 08/12/2019] [Indexed: 01/22/2023] Open
Abstract
Background Adrenocortical adenomas (ACAs) can lead to the autonomous secretion of aldosterone responsible for primary aldosteronism (PA), which is the most common form of secondary arterial hypertension. However, the authentic fundamental mechanisms underlying ACAs remain unclear. Objective Isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomics and bioinformatics analyses from etiological studies of ACAs were performed to screen the differentially expressed proteins (DEPs) and investigate the relevant mechanisms of their occurrence and development. Results could help determine therapeutic targets of clinical significance. Methods In the present study, iTRAQ-based proteomics was applied to analyze ACA tissue samples from normal adrenal cortex tissues adjacent to the tumor. Using proteins extracted from a panel of four pairs of ACA samples, we identified some upregulated proteins and other downregulated proteins in all four pairs of ACA samples compared with adjacent normal tissue. Subsequently, we predicted protein–protein interaction networks of three DEPs to determine the authentic functional factors in ACA. Results A total of 753 DEPs were identified, including 347 upregulated and 406 downregulated proteins. The expression of three upregulated proteins (E2F3, KRT6A, and ALDH1A2) was validated by Western blot in 24 ACA samples. Our data suggested that some DEPs might be important hallmarks during the development of ACA. Conclusions This study is the first proteomic research to investigate alterations in protein levels and affected pathways in ACA using the iTRAQ technique. Thus, this study not only provides a comprehensive dataset on overall protein changes but also sheds light on its potential molecular mechanism in human ACAs. Electronic supplementary material The online version of this article (10.1186/s12864-019-6030-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- He Ma
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China.,Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Ranwei Li
- Department of Urinary Surgery, the Second Hospital of Jilin University, Changchun, China
| | - Xin Di
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Xin Jin
- Department of Hematology, the Second Hospital of Jilin University, Changchun, China
| | - Yan Wang
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China
| | - Bingjie Lai
- Department of Intensive Care Unit, the Second Hospital of Jilin University, Changchun, China
| | - Cailian Shi
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Mingxin Ji
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Xinran Zhu
- Department of Anesthesiology, the Second Hospital of Jilin University, Changchun, China
| | - Ke Wang
- Department of Respiratory Medicine, the Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
23
|
Grozdanić M, Vidmar R, Vizovišek M, Fonović M. Degradomics in Biomarker Discovery. Proteomics Clin Appl 2019; 13:e1800138. [PMID: 31291060 DOI: 10.1002/prca.201800138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/01/2019] [Indexed: 12/13/2022]
Abstract
The upregulation of protease expression and proteolytic activity is implicated in numerous pathological conditions such as neurodegeneration, cancer, cardiovascular and autoimmune diseases, and bone degeneration. During disease progression, various proteases form characteristic patterns of cleaved proteins and peptides, which can affect disease severity and course of progression. It has been shown that qualitative and quantitative monitoring of cleaved protease substrates can provide relevant prognostic, diagnostic, and therapeutic information. As proteolytic fragments and peptides generated in the affected tissue are commonly translocated to blood, urine, and other proximal fluids, their possible application as biomarkers is the subject of ongoing research. The field of degradomics has been established to enable the global identification of proteolytic events on the organism level, utilizing proteomic approaches and sample preparation techniques that facilitate the detection of proteolytic processing of protease substrates in complex biological samples. In this review, some of the latest developments in degradomic methodologies used for the identification and validation of biologically relevant proteolytic events and their application in the search for clinically relevant biomarker candidates are presented. The current state of degradomics in clinics is discussed and the future perspectives of the field are outlined.
Collapse
Affiliation(s)
- Marija Grozdanić
- Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, SI-1000, Ljubljana, Slovenia.,International Postgraduate School Jožef Stefan, SI-1000, Ljubljana, Slovenia
| | - Robert Vidmar
- Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, SI-1000, Ljubljana, Slovenia
| | - Matej Vizovišek
- Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, SI-1000, Ljubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, SI-1000, Ljubljana, Slovenia
| |
Collapse
|
24
|
Montoya A, López MC, Vélez ID, Robledo SM. Label-free quantitative proteomic analysis reveals potential biomarkers for early healing in cutaneous leishmaniasis. PeerJ 2019; 6:e6228. [PMID: 30648003 PMCID: PMC6330957 DOI: 10.7717/peerj.6228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/06/2018] [Indexed: 01/08/2023] Open
Abstract
Background Leishmaniasis is a parasitic disease caused by more than 20 species of the Leishmania genus. The disease is globally distributed and is endemic in 97 countries and three territories in the tropical and subtropical regions. The efficacy of the current treatments is becoming increasingly low either due to incomplete treatment or resistant parasites. Failure of treatment is frequent, and therefore, the search for early biomarkers of therapeutic response in cutaneous leishmaniasis (CL) is urgently needed. Objective The aim of this study was to compare the proteomic profiles in patients with CL before and after 7 days of treatment and identify early biomarkers of curative response. Methods Four patients with a parasitological diagnosis of leishmaniasis with confirmation of species by PCR-RFLP were recruited. All patients had a single lesion, and a protein from the middle of the ulcer was quantified by liquid chromatography and mass spectrometry. Results A total of 12 proteins showed differential expression in the comparative LC-electrospray ionization MS/MS (LC-ESI-MS/MS) triplicate analysis. Seven of them were up-regulated and five of them were down-regulated. Calcium binding proteins A2, A8, and A9 and hemoglobin subunits alpha-2 and delta showed high correlation with epidermis development and immune response. Conclusion We identified changes in the profiles of proteins that had a positive therapeutic response to the treatment. The proteins identified with differential expression are related to the reduction of inflammation and increased tissue repair. These proteins can be useful as biomarkers for early monitoring of therapeutic response in CL.
Collapse
Affiliation(s)
- Andrés Montoya
- PECET, Facultad de Medicina, Universidad de Antioquia, Medellin, Antioquia, Colombia
| | - Manuel Carlos López
- Molecular Biology Department Consejo Superior de Investigaciones Científicas, Instituto de Parasitología y Biomedicina "López Neyra", Granade, Spain
| | - Ivan D Vélez
- PECET, Facultad de Medicina, Universidad de Antioquia, Medellin, Antioquia, Colombia
| | - Sara M Robledo
- PECET, Facultad de Medicina, Universidad de Antioquia, Medellin, Antioquia, Colombia
| |
Collapse
|
25
|
Cole LM, Clench MR, Francese S. Sample Treatment for Tissue Proteomics in Cancer, Toxicology, and Forensics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1073:77-123. [PMID: 31236840 DOI: 10.1007/978-3-030-12298-0_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Since the birth of proteomics science in the 1990, the number of applications and of sample preparation methods has grown exponentially, making a huge contribution to the knowledge in life science disciplines. Continuous improvements in the sample treatment strategies unlock and reveal the fine details of disease mechanisms, drug potency, and toxicity as well as enable new disciplines to be investigated such as forensic science.This chapter will cover the most recent developments in sample preparation strategies for tissue proteomics in three areas, namely, cancer, toxicology, and forensics, thus also demonstrating breath of application within the domain of health and well-being, pharmaceuticals, and secure societies.In particular, in the area of cancer (human tumor biomarkers), the most efficient and multi-informative proteomic strategies will be covered in relation to the subsequent application of matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) and liquid extraction surface analysis (LESA), due to their ability to provide molecular localization of tumor biomarkers albeit with different spatial resolution.With respect to toxicology, methodologies applied in toxicoproteomics will be illustrated with examples from its use in two important areas: the study of drug-induced liver injury (DILI) and studies of effects of chemical and environmental insults on skin, i.e., the effects of irritants, sensitizers, and ionizing radiation. Within this chapter, mainly tissue proteomics sample preparation methods for LC-MS/MS analysis will be discussed as (i) the use of LC-MS/MS is majorly represented in the research efforts of the bioanalytical community in this area and (ii) LC-MS/MS still is the gold standard for quantification studies.Finally, the use of proteomics will also be discussed in forensic science with respect to the information that can be recovered from blood and fingerprint evidence which are commonly encountered at the scene of the crime. The application of proteomic strategies for the analysis of blood and fingerprints is novel and proteomic preparation methods will be reported in relation to the subsequent use of mass spectrometry without any hyphenation. While generally yielding more information, hyphenated methods are often more laborious and time-consuming; since forensic investigations need quick turnaround, without compromising validity of the information, the prospect to develop methods for the application of quick forensic mass spectrometry techniques such as MALDI-MS (in imaging or profiling mode) is of great interest.
Collapse
Affiliation(s)
- L M Cole
- Biomolecular Science Research Centre, Centre for Mass Spectrometry Imaging, Sheffield Hallam University, Sheffield, UK
| | - M R Clench
- Biomolecular Science Research Centre, Centre for Mass Spectrometry Imaging, Sheffield Hallam University, Sheffield, UK
| | - S Francese
- Biomolecular Science Research Centre, Centre for Mass Spectrometry Imaging, Sheffield Hallam University, Sheffield, UK.
| |
Collapse
|
26
|
Herrera C, Escalante T, Rucavado A, Fox JW, Gutiérrez JM. Metalloproteinases in disease: identification of biomarkers of tissue damage through proteomics. Expert Rev Proteomics 2018; 15:967-982. [DOI: 10.1080/14789450.2018.1538800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Cristina Herrera
- Facultad de Farmacia, Universidad de Costa Rica, San José, Costa Rica
| | - Teresa Escalante
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Alexandra Rucavado
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Jay W. Fox
- School of Medicine, University of Virginia, Charlottesville, VA22959, USA
| | - José María Gutiérrez
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
27
|
Lim CH, Adav SS, Sze SK, Choong YK, Saravanan R, Schmidtchen A. Thrombin and Plasmin Alter the Proteome of Neutrophil Extracellular Traps. Front Immunol 2018; 9:1554. [PMID: 30038618 PMCID: PMC6046383 DOI: 10.3389/fimmu.2018.01554] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/22/2018] [Indexed: 12/12/2022] Open
Abstract
Neutrophil extracellular traps (NETs) consist of a decondensed DNA scaffold decorated with neutrophil-derived proteins. The proteome of NETs, or "NETome," has been largely elucidated in vitro. However, components such as plasma and extracellular matrix proteins may affect the NETome under physiological conditions. Here, using a reductionistic approach, we explored the effects of two proteases active during injury and wounding, human thrombin and plasmin, on the NETome. Using high-resolution mass spectrometry, we identified a total of 164 proteins, including those previously not described in NETs. The serine proteases, particularly thrombin, were also found to interact with DNA and bound to NETs in vitro. Among the most abundant proteins were those identified previously, including histones, neutrophil elastase, and antimicrobial proteins. We observed reduced histone (H2B, H3, and H4) and neutrophil elastase levels upon the addition of the two proteases. Analyses of NET-derived tryptic peptides identified subtle changes upon protease treatments. Our results provide evidence that exogenous proteases, present during wounding and inflammation, influence the NETome. Taken together, regulation of NETs and their proteins under different physiological conditions may affect their roles in infection, inflammation, and the host response.
Collapse
Affiliation(s)
- Chun Hwee Lim
- Interdisciplinary Graduate School, NTU Institute for Health Technologies, Nanyang Technological University, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Sunil S Adav
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yeu Khai Choong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Rathi Saravanan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Artur Schmidtchen
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Opdenakker G, Van Damme J, Vranckx JJ. Immunomodulation as Rescue for Chronic Atonic Skin Wounds. Trends Immunol 2018; 39:341-354. [PMID: 29500031 DOI: 10.1016/j.it.2018.01.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/08/2017] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Chronic skin wounds, caused by arterial or venous insufficiency or by physical pressure, constitute an increasing medical problem as populations age. Whereas typical wounds are characterized by local inflammation that participates in the healing process, atonic wounds lack inflammatory markers, such as neutrophil infiltration, and generally do not heal. Recently, prominent roles in the immunopathology of chronic wounds were attributed to dysregulations in specific cytokines, chemokines, matrix metalloproteinases (MMPs), and their substrates. Together with the complement system, these molecular players provide necessary defense against infections, initiate angiogenesis, and prepare tissue reconstitution. Here, we review the current state of the field and include the concept that, aside from surgery and stem cell therapy, healing may be enhanced by immunomodulating agents.
Collapse
Affiliation(s)
- Ghislain Opdenakker
- Laboratory of Immunobiology and Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium; The Glycobiology Institute, University of Oxford, Oxford, UK.
| | - Jo Van Damme
- Laboratory of Immunobiology and Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Jan Jeroen Vranckx
- Department of Development & Regeneration & Department of Plastic & Reconstructive Surgery, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| |
Collapse
|
29
|
Manes NP, Nita-Lazar A. Application of targeted mass spectrometry in bottom-up proteomics for systems biology research. J Proteomics 2018; 189:75-90. [PMID: 29452276 DOI: 10.1016/j.jprot.2018.02.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/25/2018] [Accepted: 02/07/2018] [Indexed: 02/08/2023]
Abstract
The enormous diversity of proteoforms produces tremendous complexity within cellular proteomes, facilitates intricate networks of molecular interactions, and constitutes a formidable analytical challenge for biomedical researchers. Currently, quantitative whole-proteome profiling often relies on non-targeted liquid chromatography-mass spectrometry (LC-MS), which samples proteoforms broadly, but can suffer from lower accuracy, sensitivity, and reproducibility compared with targeted LC-MS. Recent advances in bottom-up proteomics using targeted LC-MS have enabled previously unachievable identification and quantification of target proteins and posttranslational modifications within complex samples. Consequently, targeted LC-MS is rapidly advancing biomedical research, especially systems biology research in diverse areas that include proteogenomics, interactomics, kinomics, and biological pathway modeling. With the recent development of targeted LC-MS assays for nearly the entire human proteome, targeted LC-MS is positioned to enable quantitative proteomic profiling of unprecedented quality and accessibility to support fundamental and clinical research. Here we review recent applications of bottom-up proteomics using targeted LC-MS for systems biology research. SIGNIFICANCE: Advances in targeted proteomics are rapidly advancing systems biology research. Recent applications include systems-level investigations focused on posttranslational modifications (such as phosphoproteomics), protein conformation, protein-protein interaction, kinomics, proteogenomics, and metabolic and signaling pathways. Notably, absolute quantification of metabolic and signaling pathway proteins has enabled accurate pathway modeling and engineering. Integration of targeted proteomics with other technologies, such as RNA-seq, has facilitated diverse research such as the identification of hundreds of "missing" human proteins (genes and transcripts that appear to encode proteins but direct experimental evidence was lacking).
Collapse
Affiliation(s)
- Nathan P Manes
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aleksandra Nita-Lazar
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Sabino F, Egli FE, Savickas S, Holstein J, Kaspar D, Rollmann M, Kizhakkedathu JN, Pohlemann T, Smola H, Auf dem Keller U. Comparative Degradomics of Porcine and Human Wound Exudates Unravels Biomarker Candidates for Assessment of Wound Healing Progression in Trauma Patients. J Invest Dermatol 2018; 138:413-422. [PMID: 28899681 DOI: 10.1016/j.jid.2017.08.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/15/2017] [Accepted: 08/17/2017] [Indexed: 12/20/2022]
Abstract
Impaired cutaneous wound healing is a major complication in elderly people and patients suffering from diabetes, the rate of which is rising in industrialized countries. Heterogeneity of clinical manifestations hampers effective molecular diagnostics and decisions for appropriate therapeutic regimens. Using a customized positional quantitative proteomics workflow, we have established a time-resolved proteome and N-terminome resource from wound exudates in a clinically relevant pig wound model that we exploited as a robust template to interpret a heterogeneous dataset from patients undergoing the same wound treatment. With zyxin, IQGA1, and HtrA1, this analysis and validation by targeted proteomics identified differential abundances and proteolytic processing of proteins of epidermal and dermal origin as prospective biomarker candidates for assessment of critical turning points in wound progression. Thus, we show the possibility of using a fine-tuned animal wound model to bridge the translational gap as a prerequisite for future extended clinical studies with large cohorts of individuals affected by healing impairments. Data are available via ProteomeXchange with identifier PXD006674.
Collapse
Affiliation(s)
- Fabio Sabino
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Zurich, Switzerland; Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Fabian E Egli
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Simonas Savickas
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Jörg Holstein
- Department of Trauma, Hand, and Reconstructive Surgery, Saarland University Hospital, Homburg, Germany
| | | | - Mika Rollmann
- Department of Trauma, Hand, and Reconstructive Surgery, Saarland University Hospital, Homburg, Germany
| | - Jayachandran N Kizhakkedathu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Chemistry, Centre for Blood Research, 4.401 Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Tim Pohlemann
- Department of Trauma, Hand, and Reconstructive Surgery, Saarland University Hospital, Homburg, Germany
| | | | - Ulrich Auf dem Keller
- ETH Zurich, Department of Biology, Institute of Molecular Health Sciences, Zurich, Switzerland; Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
31
|
Savickas S, Auf dem Keller U. Targeted degradomics in protein terminomics and protease substrate discovery. Biol Chem 2017; 399:47-54. [PMID: 28850541 DOI: 10.1515/hsz-2017-0187] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
Abstract
Targeted degradomics integrates positional information into mass spectrometry (MS)-based targeted proteomics workflows and thereby enables analysis of proteolytic cleavage events with unprecedented specificity and sensitivity. Rapid progress in the establishment of protease-substrate relations provides extensive degradomics target lists that now can be tested with help of selected and parallel reaction monitoring (S/PRM) in complex biological systems, where proteases act in physiological environments. In this minireview, we describe the general principles of targeted degradomics, outline the generic experimental workflow of the methodology and highlight recent and future applications in protease research.
Collapse
Affiliation(s)
- Simonas Savickas
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, CH-8093 Zurich, Switzerland
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Anker Engelunds Vej, Building 301, DK-2800 Kgs. Lyngby, Denmark
| | - Ulrich Auf dem Keller
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, CH-8093 Zurich, Switzerland
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Anker Engelunds Vej, Building 301, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
32
|
Proteolytic signatures define unique thrombin-derived peptides present in human wound fluid in vivo. Sci Rep 2017; 7:13136. [PMID: 29030565 PMCID: PMC5640616 DOI: 10.1038/s41598-017-13197-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/19/2017] [Indexed: 01/13/2023] Open
Abstract
The disease burden of failing skin repair and non-healing ulcers is extensive. There is an unmet need for new diagnostic approaches to better predict healing activity and wound infection. Uncontrolled and excessive protease activity, of endogenous or bacterial origin, has been described as a major contributor to wound healing impairments. Proteolytic peptide patterns could therefore correlate and “report” healing activity and infection. This work describes a proof of principle delineating a strategy by which peptides from a selected protein, human thrombin, are detected and attributed to proteolytic actions. With a particular focus on thrombin-derived C-terminal peptides (TCP), we show that distinct peptide patterns are generated in vitro by the human S1 peptidases human neutrophil elastase and cathepsin G, and the bacterial M4 peptidases Pseudomonas aeruginosa elastase and Staphylococcus aureus aureolysin, respectively. Corresponding peptide sequences were identified in wound fluids from acute and non-healing ulcers, and notably, one peptide, FYT21 (FYTHVFRLKKWIQKVIDQFGE), was only present in wound fluid from non-healing ulcers colonized by P. aeruginosa and S. aureus. Our result is a proof of principle pointing at the possibility of defining peptide biomarkers reporting distinct proteolytic activities, of potential implication for improved diagnosis of wound healing and infection.
Collapse
|
33
|
Stolzenburg-Veeser L, Golubnitschaja O. Mini-encyclopaedia of the wound healing - Opportunities for integrating multi-omic approaches into medical practice. J Proteomics 2017; 188:71-84. [PMID: 28757465 DOI: 10.1016/j.jprot.2017.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 12/12/2022]
Abstract
Wound healing is a highly complex life-important repair process triggered by plenty of local and/or systemic organ and tissue damaging events, such as an acute surgical invasion, accidental organ and tissue damages, acute and chronic diseases, aggressive local and systemic therapeutic approaches (e.g. irradiation and systemic chemotherapy). Individual health condition determines over the quality of wound healing. Impaired wound healing, in turn, may lead, for example, to post-surgical complications frequently observed in elderly, chronic ulcers in diabetic patients, hindered and ineffective pain management, etc. However, these well-acknowledged examples are just the tip of the iceberg. The entire spectrum of potential consequences is much broader. Therefore, all the aspects of wound healing need to receive a dedicated attention of many specialised medical fields and healthcare as a whole. In contrast, there is still strongly limited knowledge collected regarding the molecular and cellular mechanisms underlying the physiological versus impaired wound healing. The contents of this article might be of great importance for multi-professional considerations as well as for the experts working in specific fields such as clinical proteomics, general practice, laboratory medicine, surgery including plastic surgery and aesthetic medicine, gerontology, psychology, diabetology, endocrinology, oncology, cardiovascular disease, radiology, and healthcare economy. SIGNIFICANCE The contents of this article are strongly motivated by the particular value of wound healing quality for medical care and might be of great importance for multi-professional considerations and experts working in specialised fields: predictive and preventive medicine, general practitioners, laboratory medicine, surgery including plastic surgery and aesthetic medicine, gerontology, psychology, diabetology, endocrinology, oncology, cardiovascular disease, radiology, and healthcare economy. The article is aiming at both educational and scientific purposes: on one side it summarises comprehensive information available regarding wound healing mechanisms and molecular pathways involved. On the other side the article provides highly innovative hypotheses for multi-professional considerations relevant for several research fields which may potentially advance medical services in the close future such as clinical proteomics and multi-omics.
Collapse
Affiliation(s)
| | - Olga Golubnitschaja
- Radiological Clinic, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany; Breast Cancer Research Centre, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany; Centre for Integrated Oncology, Cologne-Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.
| |
Collapse
|
34
|
|
35
|
Bingeman TS, Perlman DH, Storey DG, Lewis IA. Digestomics: an emerging strategy for comprehensive analysis of protein catabolism. Curr Opin Biotechnol 2016; 43:134-140. [PMID: 28025112 DOI: 10.1016/j.copbio.2016.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/01/2016] [Accepted: 11/04/2016] [Indexed: 11/29/2022]
Abstract
When cells mobilize nutrients from protein, they generate a fingerprint of peptide fragments that reflects the net action of proteases and the identities of the affected proteins. Analyzing these mixtures falls into a grey area between proteomics and metabolomics that is poorly served by existing technology. Herein, we describe an emerging digestomics strategy that bridges this gap and allows mixtures of proteolytic fragments to be quantitatively mapped with an amino acid level of resolution. We describe recent successes using this technique, including a case where digestomics provided the link between hemoglobin digestion by the malaria parasite and the world-wide distribution of chloroquine resistance. We highlight other areas of microbiology and cancer research that are well-suited to this emerging technology.
Collapse
Affiliation(s)
- Travis S Bingeman
- Department of Biological Science, University of Calgary, 2500 University Dr NW, Calgary, AB, Canada T2N 1N4
| | - David H Perlman
- Department of Biological Science, University of Calgary, 2500 University Dr NW, Calgary, AB, Canada T2N 1N4
| | - Douglas G Storey
- Department of Biological Science, University of Calgary, 2500 University Dr NW, Calgary, AB, Canada T2N 1N4
| | - Ian A Lewis
- Department of Biological Science, University of Calgary, 2500 University Dr NW, Calgary, AB, Canada T2N 1N4.
| |
Collapse
|
36
|
Müller SA, Scilabra SD, Lichtenthaler SF. Proteomic Substrate Identification for Membrane Proteases in the Brain. Front Mol Neurosci 2016; 9:96. [PMID: 27790089 PMCID: PMC5062031 DOI: 10.3389/fnmol.2016.00096] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/21/2016] [Indexed: 12/26/2022] Open
Abstract
Cell-cell communication in the brain is controlled by multiple mechanisms, including proteolysis. Membrane-bound proteases generate signaling molecules from membrane-bound precursor proteins and control the length and function of cell surface membrane proteins. These proteases belong to different families, including members of the “a disintegrin and metalloprotease” (ADAM), the beta-site amyloid precursor protein cleaving enzymes (BACE), membrane-type matrix metalloproteases (MT-MMP) and rhomboids. Some of these proteases, in particular ADAM10 and BACE1 have been shown to be essential not only for the correct development of the mammalian brain, but also for myelination and maintaining neuronal connections in the adult nervous system. Additionally, these proteases are considered as drug targets for brain diseases, including Alzheimer’s disease (AD), schizophrenia and cancer. Despite their biomedical relevance, the molecular functions of these proteases in the brain have not been explored in much detail, as little was known about their substrates. This has changed with the recent development of novel proteomic methods which allow to identify substrates of membrane-bound proteases from cultured cells, primary neurons and other primary brain cells and even in vivo from minute amounts of mouse cerebrospinal fluid (CSF). This review summarizes the recent advances and highlights the strengths of the individual proteomic methods. Finally, using the example of the Alzheimer-related proteases BACE1, ADAM10 and γ-secretase, as well as ADAM17 and signal peptide peptidase like 3 (SPPL3), we illustrate how substrate identification with novel methods is instrumental in elucidating broad physiological functions of these proteases in the brain and other organs.
Collapse
Affiliation(s)
- Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany
| | - Simone D Scilabra
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE)Munich, Germany; Neuroproteomics, Klinikum rechts der Isar, Technische Universität MünchenMunich, Germany; Institute for Advanced Study, Technische Universität MunichGarching, Germany; Munich Cluster for Systems Neurology (SyNergy)Munich, Germany
| |
Collapse
|
37
|
Rognoni E, Ruppert R, Fässler R. The kindlin family: functions, signaling properties and implications for human disease. J Cell Sci 2016; 129:17-27. [PMID: 26729028 DOI: 10.1242/jcs.161190] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The kindlin (or fermitin) family of proteins comprises three members (kindlin-1,-2 and -3) of evolutionarily conserved focal adhesion (FA) proteins, whose best-known task is to increase integrin affinity for a ligand (also referred as integrin activation) through binding of β-integrin tails. The consequence of kindlin-mediated integrin activation and integrin-ligand binding is cell adhesion, spreading and migration, assembly of the extracellular matrix (ECM), cell survival, proliferation and differentiation. Another hallmark of kindlins is their involvement in disease. Mutations in the KINDLIN-1 (also known as FERMT1) gene cause Kindler syndrome (KS)--in which mainly skin and intestine are affected, whereas mutations in the KINDLIN-3 (also known as FERMT3) gene cause leukocyte adhesion deficiency type III (LAD III), which is characterized by impaired extravasation of blood effector cells and severe, spontaneous bleedings. Also, aberrant expression of kindlins in various forms of cancer and in tissue fibrosis has been reported. Although the malfunctioning of integrins represent a major cause leading to kindlin-associated diseases, increasing evidence also point to integrin-independent functions of kindlins that play an important role in the pathogenesis of certain disease aspects. Furthermore, isoform-specific kindlin functions have been discovered, explaining, for example, why loss of kindlins differentially affects tissue stem cell homeostasis or tumor development. This Commentary focuses on new and isoform-specific kindlin functions in different tissues and discusses their potential role in disease development and progression.
Collapse
Affiliation(s)
- Emanuel Rognoni
- Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Raphael Ruppert
- Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Reinhard Fässler
- Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| |
Collapse
|
38
|
Growth factor pathways in hypertrophic scars: Molecular pathogenesis and therapeutic implications. Biomed Pharmacother 2016; 84:42-50. [PMID: 27636511 DOI: 10.1016/j.biopha.2016.09.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 08/26/2016] [Accepted: 09/05/2016] [Indexed: 01/07/2023] Open
Abstract
Hypertrophic scars represent the most common complication of skin injury and are caused by excessive cutaneous wound healing characterized by hypervascularity and pathological deposition of extracellular matrix (ECM) components. To date, the optimal and specific treatment methods for hypertrophic scars have not been available in the clinic. Current paradigm has established fibroblasts and myofibroblasts as pivotal effector cells in the pathophysiology of wound healing. Their biological properties including origin, proliferation, migration, contraction and ECM regulation have profound impacts on the progression and regression of hypertrophic scars. These complex processes are executed and modulated by a signaling network involving a number of growth factors and cytokines. Of particular importance is transforming growth factor-β, platelet-derived growth factor, connective tissue growth factor, epidermal growth factor, and vascular endothelial growth factor. This review article briefly describes the biological functions of fibroblasts and myofibroblasts during hypertrophic scars, and thereafter examines the up-to-date molecular knowledge on the roles of key growth factor pathways in the pathophysiology of hypertrophic scars. Importantly, the therapeutic implications and future challenges of these molecular discoveries are critically discussed in the hope of advancing therapeutic approaches to limit pathological scar formation.
Collapse
|
39
|
Wilson CH, Zhang HE, Gorrell MD, Abbott CA. Dipeptidyl peptidase 9 substrates and their discovery: current progress and the application of mass spectrometry-based approaches. Biol Chem 2016; 397:837-856. [PMID: 27410463 DOI: 10.1515/hsz-2016-0174] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/04/2016] [Indexed: 02/11/2025]
Abstract
The enzyme members of the dipeptidyl peptidase 4 (DPP4) gene family have the very unusual capacity to cleave the post-proline bond to release dipeptides from the N-terminus of peptide/protein substrates. DPP4 and related enzymes are current and potential therapeutic targets in the treatment of type II diabetes, inflammatory conditions and cancer. Despite this, the precise biological function of individual dipeptidyl peptidases (DPPs), other than DPP4, and knowledge of their in vivo substrates remains largely unknown. For many years, identification of physiological DPP substrates has been difficult due to limitations in the available tools. Now, with advances in mass spectrometry based approaches, we can discover DPP substrates on a system wide-scale. Application of these approaches has helped reveal some of the in vivo natural substrates of DPP8 and DPP9 and their unique biological roles. In this review, we provide a general overview of some tools and approaches available for protease substrate discovery and their applicability to the DPPs with a specific focus on DPP9 substrates. This review provides comment upon potential approaches for future substrate elucidation.
Collapse
|
40
|
Kurinna S, Muzumdar S, Köhler UA, Kockmann T, Auf dem Keller U, Schäfer M, Werner S. Autocrine and Paracrine Regulation of Keratinocyte Proliferation through a Novel Nrf2-IL-36γ Pathway. THE JOURNAL OF IMMUNOLOGY 2016; 196:4663-70. [PMID: 27183581 DOI: 10.4049/jimmunol.1501447] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 03/24/2016] [Indexed: 01/17/2023]
Abstract
The Nrf2 transcription factor is well known for its cytoprotective functions through regulation of genes involved in the detoxification of reactive oxygen species or toxic compounds. Therefore, activation of Nrf2 is a promising strategy for the protection of tissues from various types of insults and for cancer prevention. However, recent studies revealed a proinflammatory activity of activated Nrf2 and a stimulating effect on epithelial cell proliferation, but the underlying mechanisms of action and the responsible target genes are largely unknown. Using a combination of gene expression profiling, chromatin immunoprecipitation, and targeted proteomics via selected reaction monitoring, we show that the gene encoding the proinflammatory cytokine IL-36γ is a novel direct target of Nrf2 in keratinocytes and hepatocytes in vitro and in vivo. As a consequence, upregulation of IL-36γ expression occurred upon genetic or pharmacological activation of Nrf2 in the epidermis and in the normal and regenerating liver. Functional in vitro studies demonstrate that IL-36γ directly stimulates proliferation of keratinocytes. In particular, it induces expression of keratinocyte mitogens in fibroblasts, suggesting that the Nrf2-IL-36γ axis promotes keratinocyte proliferation through a double paracrine loop. These results provide mechanistic insight into Nrf2 action in the control of inflammation and cell proliferation through regulation of a proinflammatory cytokine with a key function in various inflammatory diseases.
Collapse
Affiliation(s)
- Svitlana Kurinna
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland; and
| | - Sukalp Muzumdar
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland; and
| | - Ulrike Anne Köhler
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland; and
| | - Tobias Kockmann
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland; and Functional Genomics Center Zurich, Swiss Federal Institute of Technology (ETH) Zurich/University of Zurich, 8057 Zurich, Switzerland
| | - Ulrich Auf dem Keller
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland; and
| | - Matthias Schäfer
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland; and
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland; and
| |
Collapse
|
41
|
Weng Y, Sui Z, Shan Y, Jiang H, Zhou Y, Zhu X, Liang Z, Zhang L, Zhang Y. In-Depth Proteomic Quantification of Cell Secretome in Serum-Containing Conditioned Medium. Anal Chem 2016; 88:4971-8. [PMID: 27042867 DOI: 10.1021/acs.analchem.6b00910] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yejing Weng
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhigang Sui
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yichu Shan
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Hao Jiang
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuan Zhou
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xudong Zhu
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Liang
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Lihua Zhang
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yukui Zhang
- Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| |
Collapse
|
42
|
Berry IJ, Steele JR, Padula MP, Djordjevic SP. The application of terminomics for the identification of protein start sites and proteoforms in bacteria. Proteomics 2015; 16:257-72. [DOI: 10.1002/pmic.201500319] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/21/2015] [Accepted: 09/30/2015] [Indexed: 01/11/2023]
Affiliation(s)
- Iain J. Berry
- The ithree Institute; University of Technology Sydney; Broadway NSW Australia
- Proteomics Core Facility; University of Technology Sydney; Broadway NSW Australia
| | - Joel R. Steele
- Proteomics Core Facility; University of Technology Sydney; Broadway NSW Australia
| | - Matthew P. Padula
- The ithree Institute; University of Technology Sydney; Broadway NSW Australia
- Proteomics Core Facility; University of Technology Sydney; Broadway NSW Australia
| | - Steven P. Djordjevic
- The ithree Institute; University of Technology Sydney; Broadway NSW Australia
- Proteomics Core Facility; University of Technology Sydney; Broadway NSW Australia
| |
Collapse
|
43
|
Schlage P, Kockmann T, Sabino F, Kizhakkedathu JN, Auf dem Keller U. Matrix Metalloproteinase 10 Degradomics in Keratinocytes and Epidermal Tissue Identifies Bioactive Substrates With Pleiotropic Functions. Mol Cell Proteomics 2015; 14:3234-46. [PMID: 26475864 DOI: 10.1074/mcp.m115.053520] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Indexed: 01/29/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are important players in skin homeostasis, wound repair, and in the pathogenesis of skin cancer. It is now well established that most of their functions are related to processing of bioactive proteins rather than components of the extracellular matrix (ECM). MMP10 is highly expressed in keratinocytes at the wound edge and at the invasive front of tumors, but hardly any non-ECM substrates have been identified and its function in tissue repair and carcinogenesis is unclear. To better understand the role of MMP10 in the epidermis, we employed multiplexed iTRAQ-based Terminal Amine Isotopic Labeling of Substrates (TAILS) and monitored MMP10-dependent proteolysis over time in secretomes from keratinocytes. Time-resolved abundance clustering of neo-N termini classified MMP10-dependent cleavage events by efficiency and refined the MMP10 cleavage site specificity by revealing a so far unknown preference for glutamate in the P1 position. Moreover, we identified and validated the integrin alpha 6 subunit, cysteine-rich angiogenic inducer 61 and dermokine as novel direct MMP10 substrates and provide evidence for MMP10-dependent but indirect processing of phosphatidylethanolamine-binding protein 1. Finally, we sampled the epidermal proteome and degradome in unprecedented depth and confirmed MMP10-dependent processing of dermokine in vivo by TAILS analysis of epidermis from transgenic mice that overexpress a constitutively active mutant of MMP10 in basal keratinocytes. The newly identified substrates are involved in cell adhesion, migration, proliferation, and/or differentiation, indicating a contribution of MMP10 to local modulation of these processes during wound healing and cancer development. Data are available via ProteomeXchange with identifier PXD002474.
Collapse
Affiliation(s)
- Pascal Schlage
- From the ‡ETH Zurich, Department of Biology,Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Tobias Kockmann
- From the ‡ETH Zurich, Department of Biology,Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Fabio Sabino
- From the ‡ETH Zurich, Department of Biology,Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Jayachandran N Kizhakkedathu
- §University of British Columbia, Department of Pathology and Laboratory Medicine and Department of Chemistry, Centre for Blood Research, 4.401Life Sciences Institute, 2350 Health Sciences Mall, Vancouver, British Columbia, Canada V6T 1Z3
| | - Ulrich Auf dem Keller
- From the ‡ETH Zurich, Department of Biology,Institute of Molecular Health Sciences, Otto-Stern-Weg 7, 8093 Zurich, Switzerland;
| |
Collapse
|
44
|
Pagel O, Loroch S, Sickmann A, Zahedi RP. Current strategies and findings in clinically relevant post-translational modification-specific proteomics. Expert Rev Proteomics 2015; 12:235-53. [PMID: 25955281 PMCID: PMC4487610 DOI: 10.1586/14789450.2015.1042867] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mass spectrometry-based proteomics has considerably extended our knowledge about the occurrence and dynamics of protein post-translational modifications (PTMs). So far, quantitative proteomics has been mainly used to study PTM regulation in cell culture models, providing new insights into the role of aberrant PTM patterns in human disease. However, continuous technological and methodical developments have paved the way for an increasing number of PTM-specific proteomic studies using clinical samples, often limited in sample amount. Thus, quantitative proteomics holds a great potential to discover, validate and accurately quantify biomarkers in body fluids and primary tissues. A major effort will be to improve the complete integration of robust but sensitive proteomics technology to clinical environments. Here, we discuss PTMs that are relevant for clinical research, with a focus on phosphorylation, glycosylation and proteolytic cleavage; furthermore, we give an overview on the current developments and novel findings in mass spectrometry-based PTM research.
Collapse
Affiliation(s)
- Oliver Pagel
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Otto-Hahn-Straße 6b, 44227 Dortmund, Germany
| | - Stefan Loroch
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Otto-Hahn-Straße 6b, 44227 Dortmund, Germany
| | | | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Otto-Hahn-Straße 6b, 44227 Dortmund, Germany
| |
Collapse
|
45
|
Schlage P, Kockmann T, Kizhakkedathu JN, auf dem Keller U. Monitoring matrix metalloproteinase activity at the epidermal-dermal interface by SILAC-iTRAQ-TAILS. Proteomics 2015; 15:2491-502. [PMID: 25871442 DOI: 10.1002/pmic.201400627] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 02/18/2015] [Accepted: 04/09/2015] [Indexed: 11/07/2022]
Abstract
Secreted proteases act on interstitial tissue secretomes released from multiple cell types. Thus, substrate proteins might be part of higher molecular complexes constituted by many proteins with diverse and potentially unknown cellular origin. In cell culture, these may be reconstituted by mixing native secretomes from different cell types prior to incubation with a test protease. Although current degradomics techniques could identify novel substrate proteins in these complexes, all information on the cellular origin is lost. To address this limitation, we combined iTRAQ-based terminal amine isotopic labeling of substrates (iTRAQ-TAILS) with SILAC to assign proteins to a specific cell type by MS1- and their cleavage by MS2-based quantification in the same experiment. We demonstrate the power of our newly established workflow by monitoring matrix metalloproteinase (MMP) 10 dependent cleavages in mixtures from light-labeled keratinocyte and heavy-labeled fibroblast secretomes. This analysis correctly assigned extracellular matrix components, such as laminins and collagens, to their respective cellular origins and revealed their processing in an MMP10-dependent manner. Hence, our newly devised degradomics workflow facilitates deeper insight into protease activity in complex intercellular compartments such as the epidermal-dermal interface by integrating multiple modes of quantification with positional proteomics. All MS data have been deposited in the ProteomeXchange with identifier PXD001643 (http://proteomecentral.proteomexchange.org/dataset/PXD001643).
Collapse
Affiliation(s)
- Pascal Schlage
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Tobias Kockmann
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research and Department of Pathology and Laboratory Medicine, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ulrich auf dem Keller
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Proteomic approaches to uncover MMP function. Matrix Biol 2015; 44-46:232-8. [PMID: 25603365 DOI: 10.1016/j.matbio.2015.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/09/2015] [Accepted: 01/09/2015] [Indexed: 12/30/2022]
Abstract
Proteomics has revolutionized protease research and particularly contributed to the identification of novel substrates and their sites of cleavage as key determinants of protease function. New technologies and rapid advancements in development of powerful mass spectrometers allowed unprecedented insights into activities of matrix metalloproteinases (MMPs) within their complex extracellular environments. Mass spectrometry-based proteomics extended our knowledge on MMP cleavage specificities and will help to develop more specific inhibitors as new therapeutics. Quantitative proteomics and N-terminal enrichment strategies have revealed numerous novel MMP substrates and shed light on their modes of action in vitro and in vivo. In this review, we provide an overview of current proteomic technologies in protease research and their application to the functional characterization of MMPs.
Collapse
|