151
|
Haxaire C, Hakobyan N, Pannellini T, Carballo C, McIlwain D, Mak TW, Rodeo S, Acharya S, Li D, Szymonifka J, Song X, Monette S, Srivastava A, Salmon JE, Blobel CP. Blood-induced bone loss in murine hemophilic arthropathy is prevented by blocking the iRhom2/ADAM17/TNF-α pathway. Blood 2018; 132:1064-1074. [PMID: 29776906 PMCID: PMC6128089 DOI: 10.1182/blood-2017-12-820571] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/14/2018] [Indexed: 12/14/2022] Open
Abstract
Hemophilic arthropathy (HA) is a debilitating degenerative joint disease that is a major manifestation of the bleeding disorder hemophilia A. HA typically begins with hemophilic synovitis that resembles inflammatory arthritides, such as rheumatoid arthritis, and frequently results in bone loss in patients. A major cause of rheumatoid arthritis is inappropriate release of the proinflammatory cytokine tumor necrosis factor-α (TNF-α) by the TNF-α convertase (TACE; also referred to as ADAM17) and its regulator, iRhom2. Therefore, we hypothesized that iRhom2/ADAM17-dependent shedding of TNF-α also has a pivotal role in mediating HA. Here, we show that addition of blood or its components to macrophages activates iRhom2/ADAM17-dependent TNF-α shedding, providing the premise to study the activation of this pathway by blood in the joint in vivo. For this, we turned to hemophilic FVIII-deficient mice (F8-/- mice), which develop a hemarthrosis following needle puncture injury with synovial inflammation and significant osteopenia adjacent to the affected joint. We found that needle puncture-induced bleeding leads to increased TNF-α levels in the affected joint of F8-/- mice. Moreover, inactivation of TNF-α or iRhom2 in F8-/- mice reduced the osteopenia and synovial inflammation that develops in this mouse model for HA. Taken together, our results suggest that blood entering the joint activates the iRhom2/ADAM17/TNF-α pathway, thereby contributing to osteopenia and synovitis in mice. Therefore, this proinflammatory signaling pathway could emerge as an attractive new target to prevent osteoporosis and joint damage in HA patients.
Collapse
Affiliation(s)
- Coline Haxaire
- Arthritis and Tissue Degeneration Program and
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY
| | - Narine Hakobyan
- Pediatric Hematology/Oncology, Rush University Medical Center, Chicago, IL
| | | | - Camila Carballo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY
| | - David McIlwain
- Baxter Laboratory in Stem Cell Biology, Department of Microbiology and Immunology, Stanford University, Stanford, CA
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
| | - Scott Rodeo
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY
| | - Suchitra Acharya
- Pediatric Hematology/Oncology, Northwell Health, New Hyde Park, NY
| | - Daniel Li
- Arthritis and Tissue Degeneration Program and
| | - Jackie Szymonifka
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY
| | - Xiangqian Song
- Pediatric Hematology/Oncology, Rush University Medical Center, Chicago, IL
| | - Sébastien Monette
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, NY
| | - Alok Srivastava
- Department of Hematology, Christian Medical College, Vellore, India
| | - Jane E Salmon
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, NY
- Department of Medicine and
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program and
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Center, University Health Network, Toronto, ON, Canada
- Department of Medicine and
- Department of Biophysics, Physiology, and Systems Biology, Weill Cornell Medicine, New York, NY; and
- Institute for Advanced Studies, Technical University Munich, Garching, Germany
| |
Collapse
|
152
|
The Rhomboid Superfamily: Structural Mechanisms and Chemical Biology Opportunities. Trends Biochem Sci 2018; 43:726-739. [DOI: 10.1016/j.tibs.2018.06.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 06/08/2018] [Accepted: 06/30/2018] [Indexed: 12/27/2022]
|
153
|
Xu MX, Ge CX, Qin YT, Gu TT, Lou DS, Li Q, Hu LF, Tan J. Multicombination Approach Suppresses Listeria monocytogenes-Induced Septicemia-Associated Acute Hepatic Failure: The Role of iRhom2 Signaling. Adv Healthc Mater 2018; 7:e1800427. [PMID: 29944201 DOI: 10.1002/adhm.201800427] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/26/2018] [Indexed: 12/18/2022]
Abstract
The mortality rate of acute liver failure significantly increases due to fatal septicemia. Inactive rhomboid protein 2 (iRhom2) is an essential regulator of shedding TNF-α by trafficking with TNF-α converting enzyme (TACE). Fisetin, a flavonoid present in various fruits and plants, possesses anti-oxidative stress and anti-inflammatory activities. Here, multi-combination nanoparticles Fe@Au conjugated with fisetin, iRhom2 small interfering RNA (siRNA), and TNF-α inhibitor (FN) are prepared to examine their effects on fatal septicemia-associated hepatic failure induced by Listeria monocytogenes (LM) in mice and to reveal the underlying mechanisms. After LM infection, upregulation of glutamic-oxalacetic transaminease, glutamic-pyruvic transaminase, alkaline phosphatase, TNF-α, malondialdehyde, H2 O2 , and O2- is observedcompared to FN-treated mice. The iRhom2/TACE/TNF-α signals are enhanced in vivo and in vitro, resulting in oxidative stress, which is especially associated with the activation of kupffer cells and other macrophages. Decrease in Nrf2 activation and increase of inflammation-associated regulators are also noted in vivo and in vitro. Furthermore, overexpression of TNF-α derived from macrophages aggravates hepatic failure. Inversely, the processes above are restored by FN nanoparticles through the regulation of the iRhom2/TACE/TNF-α axis and Nrf2 activation. These findings suggest that FN may be a potential approach to protect against bacterial septicemia-related diseases by targeting iRhom2.
Collapse
Affiliation(s)
- Min-Xuan Xu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Chen-Xu Ge
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Yu-Ting Qin
- School of Medicine and Pharmacy; Ocean University of China; Qingdao 266100 P. R. China
| | - Ting-Ting Gu
- College of Engineering and Applied Sciences; Nanjing University; Nanjing 210023 P. R. China
| | - De-Shuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Qiang Li
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Lin-Feng Hu
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region; School of Biological and Chemical Engineering; Chongqing University of Education; Chongqing 400067 P. R. China
- Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years; Chongqing University of Education; Chongqing 400067 P. R. China
| |
Collapse
|
154
|
iRhom2 promotes atherosclerosis through macrophage inflammation and induction of oxidative stress. Biochem Biophys Res Commun 2018; 503:1897-1904. [DOI: 10.1016/j.bbrc.2018.07.133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 07/25/2018] [Indexed: 12/21/2022]
|
155
|
Merilahti JAM, Elenius K. Gamma-secretase-dependent signaling of receptor tyrosine kinases. Oncogene 2018; 38:151-163. [PMID: 30166589 PMCID: PMC6756091 DOI: 10.1038/s41388-018-0465-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/28/2022]
Abstract
Human genome harbors 55 receptor tyrosine kinases (RTK). At least half of the RTKs have been reported to be cleaved by gamma-secretase-mediated regulated intramembrane proteolysis. The two-step process involves releasing the RTK ectodomain to the extracellular space by proteolytic cleavage called shedding, followed by cleavage in the RTK transmembrane domain by the gamma-secretase complex resulting in release of a soluble RTK intracellular domain. This intracellular domain, including the tyrosine kinase domain, can in turn translocate to various cellular compartments, such as the nucleus or proteasome. The soluble intracellular domain may interact with transcriptional regulators and other proteins to induce specific effects on cell survival, proliferation, and differentiation, establishing an additional signaling mode for the cleavable RTKs. On the other hand, the same process can facilitate RTK turnover and proteasomal degradation. In this review we focus on the regulation of RTK shedding and gamma-secretase cleavage, as well as signaling promoted by the soluble RTK ICDs. In addition, therapeutic implications of increased knowledge on RTK cleavage on cancer drug development are discussed.
Collapse
Affiliation(s)
- Johannes A M Merilahti
- Institute of Biomedicine, University of Turku, 20520, Turku, Finland.,Medicity Research Laboratory, University of Turku, 20520, Turku, Finland.,Turku Doctoral Programme of Molecular Medicine, University of Turku, 20520, Turku, Finland
| | - Klaus Elenius
- Institute of Biomedicine, University of Turku, 20520, Turku, Finland. .,Medicity Research Laboratory, University of Turku, 20520, Turku, Finland. .,Department of Oncology, Turku University Hospital, 20520, Turku, Finland.
| |
Collapse
|
156
|
Neutrophil and Macrophage Cell Surface Colony-Stimulating Factor 1 Shed by ADAM17 Drives Mouse Macrophage Proliferation in Acute and Chronic Inflammation. Mol Cell Biol 2018; 38:MCB.00103-18. [PMID: 29891514 DOI: 10.1128/mcb.00103-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/06/2018] [Indexed: 02/04/2023] Open
Abstract
Macrophages are prominent cells in acute and chronic inflammatory diseases. Recent studies highlight a role for macrophage proliferation post-monocyte recruitment under inflammatory conditions. Using an acute peritonitis model, we identify a significant defect in macrophage proliferation in mice lacking the leukocyte transmembrane protease ADAM17. The defect is associated with decreased levels of macrophage colony-stimulating factor 1 (CSF-1) in the peritoneum and is rescued by intraperitoneal injection of CSF-1. Cell surface CSF-1 (csCSF-1) is one of the substrates of ADAM17. We demonstrate that both infiltrated neutrophils and macrophages are major sources of csCSF-1. Furthermore, acute shedding of csCSF-1 following neutrophil extravasation is associated with elevated expression of iRhom2, a member of the rhomboid-like superfamily, which promotes ADAM17 maturation and trafficking to the neutrophil surface. Accordingly, deletion of hematopoietic iRhom2 is sufficient to prevent csCSF-1 release from neutrophils and macrophages and to prevent macrophage proliferation. In acute inflammation, csCSF-1 release and macrophage proliferation are self-limiting due to transient leukocyte recruitment and temporally restricted csCSF-1 expression. In chronic inflammation, such as atherosclerosis, the ADAM17-mediated lesional macrophage proliferative response is prolonged. Our results demonstrate a novel mechanism whereby ADAM17 promotes macrophage proliferation in states of acute and chronic inflammation.
Collapse
|
157
|
Lichtenthaler SF, Lemberg MK, Fluhrer R. Proteolytic ectodomain shedding of membrane proteins in mammals-hardware, concepts, and recent developments. EMBO J 2018; 37:e99456. [PMID: 29976761 PMCID: PMC6068445 DOI: 10.15252/embj.201899456] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/05/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Proteolytic removal of membrane protein ectodomains (ectodomain shedding) is a post-translational modification that controls levels and function of hundreds of membrane proteins. The contributing proteases, referred to as sheddases, act as important molecular switches in processes ranging from signaling to cell adhesion. When deregulated, ectodomain shedding is linked to pathologies such as inflammation and Alzheimer's disease. While proteases of the "a disintegrin and metalloprotease" (ADAM) and "beta-site APP cleaving enzyme" (BACE) families are widely considered as sheddases, in recent years a much broader range of proteases, including intramembrane and soluble proteases, were shown to catalyze similar cleavage reactions. This review demonstrates that shedding is a fundamental process in cell biology and discusses the current understanding of sheddases and their substrates, molecular mechanisms and cellular localizations, as well as physiological functions of protein ectodomain shedding. Moreover, we provide an operational definition of shedding and highlight recent conceptual advances in the field. While new developments in proteomics facilitate substrate discovery, we expect that shedding is not a rare exception, but rather the rule for many membrane proteins, and that many more interesting shedding functions await discovery.
Collapse
Affiliation(s)
- Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, Klinikum rechts der Isar, School of Medicine, and Institute for Advanced Study, Technical University Munich, Munich, Germany
- Munich Center for Systems Neurology (SyNergy), Munich, Germany
| | - Marius K Lemberg
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Regina Fluhrer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Biomedizinisches Centrum (BMC), Ludwig-Maximilians University of Munich, Munich, Germany
| |
Collapse
|
158
|
Hosur V, Farley ML, Low BE, Burzenski LM, Shultz LD, Wiles MV. RHBDF2-Regulated Growth Factor Signaling in a Rare Human Disease, Tylosis With Esophageal Cancer: What Can We Learn From Murine Models? Front Genet 2018; 9:233. [PMID: 30022999 PMCID: PMC6039722 DOI: 10.3389/fgene.2018.00233] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022] Open
Abstract
Tylosis with esophageal cancer syndrome (TOC) is a rare autosomal dominant proliferative skin disease caused by missense mutations in the rhomboid 5 homolog 2 (RHBDF2) gene. TOC is characterized by thickening of the skin in the palms and feet and is strongly linked with the development of esophageal squamous cell carcinoma. Murine models of human diseases have been valuable tools for investigating the underlying genetic and molecular mechanisms of a broad range of diseases. Although current mouse models do not fully recapitulate all aspects of human TOC, and the molecular mechanisms underlying TOC are still emerging, the available mouse models exhibit several key aspects of the disease, including a proliferative skin phenotype, a rapid wound healing phenotype, susceptibility to epithelial cancer, and aberrant epidermal growth factor receptor (EGFR) signaling. Furthermore, we and other investigators have used these models to generate new insights into the causes and progression of TOC, including findings suggesting a tissue-specific role of the RHBDF2-EGFR pathway, rather than a role of the immune system, in mediating TOC; and indicating that amphiregulin, an EGFR ligand, is a functional driver of the disease. This review highlights the mouse models of TOC available to researchers for use in investigating the disease mechanisms and possible therapies, and the significance of genetic modifiers of the disease identified in these models in delineating the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Vishnu Hosur
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | | | | | | | | |
Collapse
|
159
|
Künzel U, Grieve AG, Meng Y, Sieber B, Cowley SA, Freeman M. FRMD8 promotes inflammatory and growth factor signalling by stabilising the iRhom/ADAM17 sheddase complex. eLife 2018; 7:e35012. [PMID: 29897336 PMCID: PMC6042961 DOI: 10.7554/elife.35012] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/26/2018] [Indexed: 01/04/2023] Open
Abstract
Many intercellular signals are synthesised as transmembrane precursors that are released by proteolytic cleavage ('shedding') from the cell surface. ADAM17, a membrane-tethered metalloprotease, is the primary shedding enzyme responsible for the release of the inflammatory cytokine TNFα and several EGF receptor ligands. ADAM17 exists in complex with the rhomboid-like iRhom proteins, which act as cofactors that regulate ADAM17 substrate shedding. Here we report that the poorly characterised FERM domain-containing protein FRMD8 is a new component of the iRhom2/ADAM17 sheddase complex. FRMD8 binds to the cytoplasmic N-terminus of iRhoms and is necessary to stabilise iRhoms and ADAM17 at the cell surface. In the absence of FRMD8, iRhom2 and ADAM17 are degraded via the endolysosomal pathway, resulting in the reduction of ADAM17-mediated shedding. We have confirmed the pathophysiological significance of FRMD8 in iPSC-derived human macrophages and mouse tissues, thus demonstrating its role in the regulated release of multiple cytokine and growth factor signals.
Collapse
Affiliation(s)
- Ulrike Künzel
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUnited Kingdom
| | - Adam Graham Grieve
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUnited Kingdom
| | - Yao Meng
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUnited Kingdom
| | - Boris Sieber
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUnited Kingdom
| | - Sally A Cowley
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUnited Kingdom
| | - Matthew Freeman
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUnited Kingdom
| |
Collapse
|
160
|
Phosphorylation of iRhom2 Controls Stimulated Proteolytic Shedding by the Metalloprotease ADAM17/TACE. Cell Rep 2018; 21:745-757. [PMID: 29045841 PMCID: PMC5656746 DOI: 10.1016/j.celrep.2017.09.074] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/12/2017] [Accepted: 09/22/2017] [Indexed: 12/02/2022] Open
Abstract
Cell surface metalloproteases coordinate signaling during development, tissue homeostasis, and disease. TACE (TNF-α-converting enzyme), is responsible for cleavage (“shedding”) of membrane-tethered signaling molecules, including the cytokine TNF, and activating ligands of the EGFR. The trafficking of TACE within the secretory pathway requires its binding to iRhom2, which mediates the exit of TACE from the endoplasmic reticulum. An important, but mechanistically unclear, feature of TACE biology is its ability to be stimulated rapidly on the cell surface by numerous inflammatory and growth-promoting agents. Here, we report a role for iRhom2 in TACE stimulation on the cell surface. TACE shedding stimuli trigger MAP kinase-dependent phosphorylation of iRhom2 N-terminal cytoplasmic tail. This recruits 14-3-3 proteins, enforcing the dissociation of TACE from complexes with iRhom2, promoting the cleavage of TACE substrates. Our data reveal that iRhom2 controls multiple aspects of TACE biology, including stimulated shedding on the cell surface. iRhom2 is phosphorylated in response to stimuli that activate the sheddase TACE Blocking iRhom phosphorylation represses TACE stimulated shedding Phosphorylated iRhom2 recruits 14-3-3 and dissociates from TACE, enabling shedding iRhom2 is thus a signal integrator and transducer of stimulated TACE shedding
Collapse
|
161
|
Oikonomidi I, Burbridge E, Cavadas M, Sullivan G, Collis B, Naegele H, Clancy D, Brezinova J, Hu T, Bileck A, Gerner C, Bolado A, von Kriegsheim A, Martin SJ, Steinberg F, Strisovsky K, Adrain C. iTAP, a novel iRhom interactor, controls TNF secretion by policing the stability of iRhom/TACE. eLife 2018; 7:35032. [PMID: 29897333 PMCID: PMC6042963 DOI: 10.7554/elife.35032] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/10/2018] [Indexed: 12/11/2022] Open
Abstract
The apical inflammatory cytokine TNF regulates numerous important biological processes including inflammation and cell death, and drives inflammatory diseases. TNF secretion requires TACE (also called ADAM17), which cleaves TNF from its transmembrane tether. The trafficking of TACE to the cell surface, and stimulation of its proteolytic activity, depends on membrane proteins, called iRhoms. To delineate how the TNF/TACE/iRhom axis is regulated, we performed an immunoprecipitation/mass spectrometry screen to identify iRhom-binding proteins. This identified a novel protein, that we name iTAP (iRhom Tail-Associated Protein) that binds to iRhoms, enhancing the cell surface stability of iRhoms and TACE, preventing their degradation in lysosomes. Depleting iTAP in primary human macrophages profoundly impaired TNF production and tissues from iTAP KO mice exhibit a pronounced depletion in active TACE levels. Our work identifies iTAP as a physiological regulator of TNF signalling and a novel target for the control of inflammation. Inflammation forms part of the body's defense system against pathogens, but if the system becomes faulty, it can cause problems linked to inflammatory and autoimmune diseases. Immune cells coordinate their activity using specific signaling molecules called cytokines. For example, the cytokine TNF is an important trigger of inflammation and is produced at the surface of immune cells. A specific enzyme called TACE is needed to release TNF, as well as other signaling molecules, including proteins that trigger healing. Previous work revealed that TACE works with proteins called iRhoms, which regulate its activity and help TACE to reach the surface of the cell to release TNF. To find out how, Oikonomidi et al. screened human cells to see what other proteins interact with iRhoms. The results revealed a new protein named iTAP, which is required to release TNF from the surface of cells. It also protects the TACE-iRhom complex from being destroyed by the cell’s waste disposal system. When iTAP was experimentally removed in human immune cells, the cells were unable to release TNF. Instead, iRhom and TACE travelled to the cell's garbage system, the lysosome, where the proteins were destroyed. Removing the iTAP gene in mice had the same effect, and the TACE-iRhom complex was no longer found on the surface of the cell, but instead degraded in lysosomes. This suggests that in healthy cells, the iTAP protein prevents the cell from destroying this protein complex. TNF controls many beneficial processes, including fighting infection and cancer. However, when the immune system releases too many cytokines, it can lead to inflammatory diseases or even cause cancer. Specific drugs that target TNF are not always effective administered on their own, and sometimes, patients stop responding to the drugs. Since the new protein iTAP works as a switch to turn TNF release on or off, it could provide a target for the development of new treatments.
Collapse
Affiliation(s)
- Ioanna Oikonomidi
- Membrane Traffic Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Emma Burbridge
- Membrane Traffic Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Miguel Cavadas
- Membrane Traffic Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Graeme Sullivan
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College Dublin, Dublin, Ireland
| | - Blanka Collis
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Heike Naegele
- Center for Biological Systems Analysis, Faculty of Biology, Albert Ludwigs Universitaet Freiburg, Freiburg, Germany
| | - Danielle Clancy
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College Dublin, Dublin, Ireland
| | - Jana Brezinova
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tianyi Hu
- Membrane Traffic Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Andrea Bileck
- Institut für Analytische Chemie, Universität Wien, Vienna, Austria
| | | | - Alfonso Bolado
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Alex von Kriegsheim
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Seamus J Martin
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College Dublin, Dublin, Ireland
| | - Florian Steinberg
- Center for Biological Systems Analysis, Faculty of Biology, Albert Ludwigs Universitaet Freiburg, Freiburg, Germany
| | - Kvido Strisovsky
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Colin Adrain
- Membrane Traffic Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| |
Collapse
|
162
|
Herrlich A, Kefaloyianni E. iRhoms: A Potential Path to More Specific Therapeutic Targeting of Lupus Nephritis. Am J Kidney Dis 2018; 72:617-619. [PMID: 29887489 DOI: 10.1053/j.ajkd.2018.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/09/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Andreas Herrlich
- Washington University School of Medicine in St. Louis, St. Louis, MO.
| | | |
Collapse
|
163
|
Lee MY, Kang JS, Go RE, Byun YS, Wi YJ, Hwang KA, Choi JH, Kim HC, Choi KC, Nam KH. Collagen-Induced Arthritis Analysis in Rhbdf2 Knockout Mouse. Biomol Ther (Seoul) 2018; 26:298-305. [PMID: 29223140 PMCID: PMC5933897 DOI: 10.4062/biomolther.2017.103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 12/02/2022] Open
Abstract
Rhomboid family member 2 gene (Rhbdf2) is an inactive homologue lacking essential catalytic residues of rhomboid intramembrane serine proteases. The protein is necessary for maturation of tumor necrosis factor-alpha (TNF-α) converting enzyme, which is the molecule responsible for the release of TNF-α. In this study, Rhbdf2 knockout (KO) mice were produced by CRISPR/CAS9. To see the effects of the failure of TNF-α release induced by Rhbdf2 gene KO, collagen-induced arthritis (CIA), which is the representative TNF-α related disease, was induced in the Rhbdf2 mutant mouse using chicken collagen type II. The severity of the CIA was measured by traditional clinical scores and histopathological analysis of hind limb joints. A rota-rod test and grip strength test were employed to evaluate the severity of CIA based on losses of physical functions. The results indicated that Rhbdf2 mutant mice showed clear alleviation of the clinical severity of CIA as demonstrated by the significantly lower severity indexes. Moreover, a grip strength test was shown to be useful for the evaluation of physical functional losses by CIA. Overall, the results showed that the Rhbdf2 gene has a significant effect on the induction of CIA, which is related to TNF-α.
Collapse
Affiliation(s)
- Min-Young Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon 28116, Republic of Korea.,Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Ju-Seong Kang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon 28116, Republic of Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Yong-Sub Byun
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon 28116, Republic of Korea
| | - Young Jin Wi
- Department of Life Science, College of Natureal Sciences, Research Institute of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natureal Sciences, Research Institute of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyoung-Chin Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon 28116, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Ki-Hoan Nam
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon 28116, Republic of Korea
| |
Collapse
|
164
|
Mendioroz M, Martínez-Merino L, Blanco-Luquin I, Urdánoz A, Roldán M, Jericó I. Liquid biopsy: a new source of candidate biomarkers in amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2018; 5:763-768. [PMID: 29928659 PMCID: PMC5989775 DOI: 10.1002/acn3.565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/12/2018] [Accepted: 03/20/2018] [Indexed: 12/26/2022] Open
Abstract
Noninvasive tests to diagnose and monitor the progression of neurodegenerative disorders have been a challenge for decades. The aim of this study was to explore the feasibility of applying liquid biopsy procedures to patients with a neurodegenerative disease such as amyotrophic lateral sclerosis (ALS). We isolated plasma cell‐free DNA (cfDNA) in 20 ALS patients and 20 controls and used cfDNA to identify a novel differentially methylated mark in RHBDF2 gene in ALS patients compared to controls. Our findings support the notion that liquid biopsy may be applied to living patients as a source of potential epigenetic biomarkers for neurodegenerative disorders.
Collapse
Affiliation(s)
- Maite Mendioroz
- Department of Neurology Complejo Hospitalario de Navarra IdiSNA (Navarra Institute for Health Research) Pamplona Spain.,Neuroepigenetics Laboratory- Navarrabiomed Complejo Hospitalario de Navarra (CHN) Public University of Navarre (UPNA) IdiSNA (Navarra Institute for Health Research Pamplona Spain
| | - Leyre Martínez-Merino
- Department of Neurology Complejo Hospitalario de Navarra IdiSNA (Navarra Institute for Health Research) Pamplona Spain
| | - Idoia Blanco-Luquin
- Neuroepigenetics Laboratory- Navarrabiomed Complejo Hospitalario de Navarra (CHN) Public University of Navarre (UPNA) IdiSNA (Navarra Institute for Health Research Pamplona Spain
| | - Amaya Urdánoz
- Neuroepigenetics Laboratory- Navarrabiomed Complejo Hospitalario de Navarra (CHN) Public University of Navarre (UPNA) IdiSNA (Navarra Institute for Health Research Pamplona Spain
| | - Miren Roldán
- Neuroepigenetics Laboratory- Navarrabiomed Complejo Hospitalario de Navarra (CHN) Public University of Navarre (UPNA) IdiSNA (Navarra Institute for Health Research Pamplona Spain
| | - Ivonne Jericó
- Department of Neurology Complejo Hospitalario de Navarra IdiSNA (Navarra Institute for Health Research) Pamplona Spain
| |
Collapse
|
165
|
Mygind KJ, Störiko T, Freiberg ML, Samsøe-Petersen J, Schwarz J, Andersen OM, Kveiborg M. Sorting nexin 9 (SNX9) regulates levels of the transmembrane ADAM9 at the cell surface. J Biol Chem 2018; 293:8077-8088. [PMID: 29622675 DOI: 10.1074/jbc.ra117.001077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/12/2018] [Indexed: 11/06/2022] Open
Abstract
ADAM9 is an active member of the family of transmembrane ADAMs (a disintegrin and metalloproteases). It plays a role in processes such as bone formation and retinal neovascularization, and importantly, its expression in human cancers correlates with disease stage and poor prognosis. Functionally, ADAM9 can cleave several transmembrane proteins, thereby shedding their ectodomains from the cell surface. Moreover, ADAM9 regulates cell behavior by binding cell-surface receptors such as integrin and membrane-type matrix metalloproteases. Because these functions are mainly restricted to the cell surface, understanding the mechanisms regulating ADAM9 localization and activity at this site is highly important. To this end, we here investigated how intracellular trafficking regulates ADAM9 availability at the cell surface. We found that ADAM9 undergoes constitutive clathrin-dependent internalization and subsequent degradation or recycling to the plasma membrane. We confirmed previous findings of an interaction between ADAM9 and the intracellular sorting protein, sorting nexin 9 (SNX9), as well as its close homolog SNX18. Knockdown of either SNX9 or SNX18 had no apparent effects on ADAM9 internalization or recycling. However, double knockdown of SNX9 and SNX18 decreased ADAM9 internalization significantly, demonstrating a redundant role in this process. Moreover, SNX9 knockdown revealed a nonredundant effect on overall ADAM9 protein levels, resulting in increased ADAM9 levels at the cell surface, and a corresponding increase in the shedding of Ephrin receptor B4, a well-known ADAM9 substrate. Together, our findings demonstrate that intracellular SNX9-mediated trafficking constitutes an important ADAM9 regulatory pathway.
Collapse
Affiliation(s)
- Kasper J Mygind
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Theresa Störiko
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Marie L Freiberg
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Jacob Samsøe-Petersen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Jeanette Schwarz
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Olav M Andersen
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Ole Worms Alle 3, 8000 Aarhus C, Denmark
| | - Marie Kveiborg
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark.
| |
Collapse
|
166
|
Li J, Perfetto M, Neuner R, Bahudhanapati H, Christian L, Mathavan K, Bridges LC, Alfandari D, Wei S. Xenopus ADAM19 regulates Wnt signaling and neural crest specification by stabilizing ADAM13. Development 2018. [PMID: 29540504 DOI: 10.1242/dev.158154] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During vertebrate gastrulation, canonical Wnt signaling induces the formation of neural plate border (NPB). Wnt is also thought to be required for the subsequent specification of neural crest (NC) lineage at the NPB, but the direct evidence is lacking. We found previously that the disintegrin metalloproteinase ADAM13 is required for Wnt activation and NC induction in Xenopus Here, we report that knockdown of ADAM13 or its close paralog ADAM19 severely downregulates Wnt activity at the NPB, inhibiting NC specification without affecting earlier NPB formation. Surprisingly, ADAM19 functions nonproteolytically in NC specification by interacting with ADAM13 and inhibiting its proteasomal degradation. Ectopic expression of stabilized ADAM13 mutants that function independently of ADAM19 can induce the NC marker/specifier snail2 in the future epidermis via Wnt signaling. These results unveil the essential roles of a novel protease-protease interaction in regulating a distinct wave of Wnt signaling, which directly specifies the NC lineage.
Collapse
Affiliation(s)
- Jiejing Li
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA.,Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Mark Perfetto
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA.,Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Russell Neuner
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | | | - Laura Christian
- Department of Biology, West Virginia University, Morgantown, WV 26506, USA
| | - Ketan Mathavan
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Lance C Bridges
- Biochemistry, Molecular and Cell Sciences, Arkansas College of Osteopathic Medicine, Arkansas Colleges of Health Education, Fort Smith, AR 72916, USA
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
167
|
Motani K, Kosako H. Activation of stimulator of interferon genes (STING) induces ADAM17-mediated shedding of the immune semaphorin SEMA4D. J Biol Chem 2018; 293:7717-7726. [PMID: 29618514 DOI: 10.1074/jbc.ra118.002175] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/23/2018] [Indexed: 01/06/2023] Open
Abstract
Stimulator of interferon genes (STING) is an endoplasmic reticulum-resident membrane protein that mediates cytosolic pathogen DNA-induced innate immunity and inflammatory responses in host defenses. STING is activated by cyclic di-nucleotides and is then translocated to the Golgi apparatus, an event that triggers STING assembly with the downstream enzyme TANK-binding kinase 1 (TBK1). This assembly leads to the phosphorylation of the transcription factor interferon regulatory factor 3 (IRF3), which in turn induces expression of type-I interferon (IFN) and chemokine genes. STING also mediates inflammatory responses independently of IRF3, but these molecular pathways are largely unexplored. Here, we analyzed the RAW264.7 macrophage secretome to comprehensively identify proinflammatory factors released into the extracellular medium upon STING activation. In total, we identified 1299 proteins in macrophage culture supernatants, of which 23 were significantly increased after STING activation. These proteins included IRF3-dependent cytokines, as well as previously unknown targets of STING, such as the immune semaphorin SEMA4D/CD100, which possesses proinflammatory cytokine-like activities. Unlike for canonical cytokines, the expression of the SEMA4D gene was not up-regulated. Instead, upon STING activation, membrane-bound SEMA4D was cleaved into a soluble form, suggesting the presence of a post-translational shedding machinery. Importantly, the SEMA4D shedding was blocked by TMI-1, an inhibitor of the sheddase ADAM metallopeptidase domain 17 (ADAM17) but not by the TBK1 inhibitor BX795. These results suggest that STING activates ADAM17 and that this activation produces soluble proinflammatory SEMA4D independently of the TBK1/IRF3-mediated transcriptional pathway.
Collapse
Affiliation(s)
- Kou Motani
- From the Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hidetaka Kosako
- From the Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
168
|
Nakayama H, Sakaue T, Maekawa M, Fujisaki A, Higashiyama S. Cullin 3 regulates ADAMs-mediated ectodomain shedding of amphiregulin. Biochem Biophys Res Commun 2018; 499:17-23. [DOI: 10.1016/j.bbrc.2018.03.097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/13/2018] [Indexed: 01/01/2023]
|
169
|
Zhai M, Guo J, Ma H, Shi W, Jou D, Yan D, Liu T, Tao J, Duan J, Wang Y, Li S, Lv J, Li C, Lin J, Zhang C, Lin L. Ursolic acid prevents angiotensin II-induced abdominal aortic aneurysm in apolipoprotein E-knockout mice. Atherosclerosis 2018; 271:128-135. [PMID: 29499360 DOI: 10.1016/j.atherosclerosis.2018.02.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS Abdominal aortic aneurysms (AAA) is a chronic inflammatory disease in which signal transducer and activator of transcription 3 (STAT3), and disintegrin and metalloproteinase 17 (ADAM17) play important roles. However, it remains unclear whether ursolic acid (UA), a natural pentacyclic triterpenoid carboxylic acid, can have an impact on STAT3 and ADAM17 and hence influence the formation of AAA. The objective of this study was to characterize the potential effect of UA on the pathogenesis of AAA and on STAT3 and ADAM17. METHODS Male ApoE-/- mice were infused with angiotensin II (AngII) (1000 ng/kg/min) for 4 weeks to induce AAAs. Daily intragastric gavage with 100 mg/kg UA or tap water containing Tween 80 as controls was provided. Immunohistochemistry, cell viability assay, colony formation, wound healing assay, and Western blot were used to explore the potential effect of UA on AAA. RESULTS UA decreased the incidence of AngII-induced AAA in mice. UA alleviated the degradation of elastin fibers and inflammation and decreased the expression of MMP2, MMP9, ADAM17 and phospho-STAT3 (pSTAT3) in aorta of mice induced with AngII. UA inhibited the constitutive and stimuli-induced (AngII and tumor necrosis factor-α) expression of MMP2, MMP9, ADAM17 and pSTAT3 in vascular smooth muscle cells (VSMCs). Furthermore, UA decreased cell viability, and suppressed colony formation and wound healing in vitro. CONCLUSIONS We demonstrated that UA ameliorated the severity of AAA and exhibited an inhibitory effect on the expression of pSTAT3 and ADAM17. UA might emerge as a promising agent contributing to the prevention or treatment of AAA.
Collapse
MESH Headings
- ADAM17 Protein/metabolism
- Angiotensin II
- Animals
- Anti-Inflammatory Agents/pharmacology
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Cell Line
- Cell Proliferation/drug effects
- Disease Models, Animal
- Elastin/metabolism
- Male
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phosphorylation
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Triterpenes/pharmacology
- Vascular Remodeling/drug effects
- Wound Healing/drug effects
- Ursolic Acid
Collapse
Affiliation(s)
- Maocai Zhai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyi Guo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiyan Ma
- Division of Cardiology, Department of Internal Medicine, First People's Hospital of Shangqiu, Shangqiu, China
| | - Wei Shi
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - David Jou
- Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, College of Medicine, The Ohio State University, Columbus OH, USA
| | - Dan Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianshu Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwen Tao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jialin Duan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yina Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenglong Li
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville FL, USA
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore MD, USA
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
170
|
Gardiner EE. Proteolytic processing of platelet receptors. Res Pract Thromb Haemost 2018; 2:240-250. [PMID: 30046726 PMCID: PMC6055504 DOI: 10.1002/rth2.12096] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/01/2018] [Indexed: 12/17/2022] Open
Abstract
Platelets have a major role in hemostasis and an emerging role in biological processes including inflammation and immunity. Many of these processes require platelet adhesion and localization at sites of tissue damage or infection and regulated platelet activation, mediated by platelet adheso-signalling receptors, glycoprotein (GP) Ib-IX-V and GPVI. Work from a number of laboratories has demonstrated that levels of these receptors are closely regulated by metalloproteinases of the A Disintegrin And Metalloproteinase (ADAM) family, primarily ADAM17 and ADAM10. It is becoming increasingly evident that platelets have important roles in innate immunity, inflammation, and in combating infection that extends beyond processes of hemostasis. This overview will examine the molecular events that regulate levels of platelet receptors and then assess ramifications for these events in settings where hemostasis, inflammation, and infection processes are triggered.
Collapse
Affiliation(s)
- Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and TherapeuticsJohn Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| |
Collapse
|
171
|
Guerra L, Castori M, Didona B, Castiglia D, Zambruno G. Hereditary palmoplantar keratodermas. Part II: syndromic palmoplantar keratodermas - Diagnostic algorithm and principles of therapy. J Eur Acad Dermatol Venereol 2018; 32:899-925. [DOI: 10.1111/jdv.14834] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/05/2018] [Indexed: 12/19/2022]
Affiliation(s)
- L. Guerra
- Laboratory of Molecular and Cell Biology; Istituto Dermopatico dell'Immacolata-IRCCS; Rome Italy
| | - M. Castori
- Division of Medical Genetics; Casa Sollievo della Sofferenza-IRCCS; San Giovanni Rotondo Italy
| | - B. Didona
- Rare Skin Disease Center; Istituto Dermopatico dell'Immacolata-IRCCS; Rome Italy
| | - D. Castiglia
- Laboratory of Molecular and Cell Biology; Istituto Dermopatico dell'Immacolata-IRCCS; Rome Italy
| | - G. Zambruno
- Genetic and Rare Diseases Research Area and Dermatology Unit; Bambino Gesù Children's Hospital-IRCCS; Rome Italy
| |
Collapse
|
172
|
Hosur V, Farley ML, Burzenski LM, Shultz LD, Wiles MV. ADAM17 is essential for ectodomain shedding of the EGF-receptor ligand amphiregulin. FEBS Open Bio 2018; 8:702-710. [PMID: 29632822 PMCID: PMC5881543 DOI: 10.1002/2211-5463.12407] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/09/2018] [Accepted: 02/13/2018] [Indexed: 12/30/2022] Open
Abstract
The epidermal growth factor (EGF)-receptor ligand amphiregulin (AREG) is a potent growth factor implicated in proliferative skin diseases and in primary and metastatic epithelial cancers. AREG, synthesized as a propeptide, requires conversion to an active peptide by metalloproteases by a process known as ectodomain shedding. Although (ADAM17) a disintegrin and metalloprotease 17 is a key sheddase of AREG, ADAM8-, ADAM15-, and batimastat (broad metalloprotease inhibitor)-sensitive metalloproteases have also been implicated in AREG shedding. In the present study, using a curly bare (Rhbdf2cub ) mouse model that shows loss-of-hair, enlarged sebaceous gland, and rapid cutaneous wound-healing phenotypes mediated by enhanced Areg mRNA and protein levels, we sought to identify the principal ectodomain sheddase of AREG. To this end, we generated Rhbdf2cub mice lacking ADAM17 specifically in the skin and examined the above phenotypes of Rhbdf2cub mice. We find that ADAM17 deficiency in the skin of Rhbdf2cub mice restores a full hair coat, prevents sebaceous gland enlargement, and impairs the rapid wound-healing phenotype observed in Rhbdf2cub mice. Furthermore, in vitro, stimulated shedding of AREG is abolished in Rhbdf2cub mouse embryonic keratinocytes lacking ADAM17. Thus, our data support previous findings demonstrating that ADAM17 is the major ectodomain sheddase of AREG.
Collapse
|
173
|
Arcidiacono P, Webb CM, Brooke MA, Zhou H, Delaney PJ, Ng KE, Blaydon DC, Tinker A, Kelsell DP, Chikh A. p63 is a key regulator of iRHOM2 signalling in the keratinocyte stress response. Nat Commun 2018; 9:1021. [PMID: 29523849 PMCID: PMC5844915 DOI: 10.1038/s41467-018-03470-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 02/14/2018] [Indexed: 02/06/2023] Open
Abstract
Hyperproliferative keratinocytes induced by trauma, hyperkeratosis and/or inflammation display molecular signatures similar to those of palmoplantar epidermis. Inherited gain-of-function mutations in RHBDF2 (encoding iRHOM2) are associated with a hyperproliferative palmoplantar keratoderma and squamous oesophageal cancer syndrome (termed TOC). In contrast, genetic ablation of rhbdf2 in mice leads to a thinning of the mammalian footpad, and reduces keratinocyte hyperproliferation and migration. Here, we report that iRHOM2 is a novel target gene of p63 and that both p63 and iRHOM2 differentially regulate cellular stress-associated signalling pathways in normal and hyperproliferative keratinocytes. We demonstrate that p63-iRHOM2 regulates cell survival and response to oxidative stress via modulation of SURVIVIN and Cytoglobin, respectively. Furthermore, the antioxidant compound Sulforaphane downregulates p63-iRHOM2 expression, leading to reduced proliferation, inflammation, survival and ROS production. These findings elucidate a novel p63-associated pathway that identifies iRHOM2 modulation as a potential therapeutic target to treat hyperproliferative skin disease and neoplasia.
Collapse
Affiliation(s)
- Paola Arcidiacono
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Catherine M Webb
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Matthew A Brooke
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Huiqing Zhou
- Department of Human Genetics, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
- Department of Molecular Developmental Biology, Radboud University, Nijmegen, The Netherlands
| | - Paul J Delaney
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Keat-Eng Ng
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Diana C Blaydon
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - David P Kelsell
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| | - Anissa Chikh
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| |
Collapse
|
174
|
Zhou J, Wu HG, Shi Y. Roles of TNF-α/NF-κB/Snail pathway in regulating epithelial-mesenchymal transition. Shijie Huaren Xiaohua Zazhi 2018; 26:441-448. [DOI: 10.11569/wcjd.v26.i7.441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process of transformation of epithelial cells to mesenchymal cells, and it not only plays an important role in the developmental process, but also participates in tissue healing, organ fibrosis, tumorigenesis, and metastasis. In recent years, it has been found that tumor necrosis factor-α (TNF-α) is a major inflammatory factor that can induce snail expression by binding to nuclear factor-κB (NF-κB), thus mediating EMT. This article briefly introduces the roles of the TNF-α/NF-κB/Snail pathway in mediating EMT, aiming to promote a further understanding of the mechanism of TNF-α in regulating EMT.
Collapse
|
175
|
Qing X, Chinenov Y, Redecha P, Madaio M, Roelofs JJ, Farber G, Issuree PD, Donlin L, Mcllwain DR, Mak TW, Blobel CP, Salmon JE. iRhom2 promotes lupus nephritis through TNF-α and EGFR signaling. J Clin Invest 2018; 128:1397-1412. [PMID: 29369823 DOI: 10.1172/jci97650] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/23/2018] [Indexed: 12/14/2022] Open
Abstract
Lupus nephritis (LN) often results in progressive renal dysfunction. The inactive rhomboid 2 (iRhom2) is a newly identified key regulator of A disintegrin and metalloprotease 17 (ADAM17), whose substrates, such as TNF-α and heparin-binding EGF (HB-EGF), have been implicated in the pathogenesis of chronic kidney diseases. Here, we demonstrate that deficiency of iRhom2 protects the lupus-prone Fcgr2b-/- mice from developing severe kidney damage without altering anti-double-stranded DNA (anti-dsDNA) Ab production by simultaneously blocking HB-EGF/EGFR and TNF-α signaling in the kidney tissues. Unbiased transcriptome profiling of kidneys and kidney macrophages revealed that TNF-α and HB-EGF/EGFR signaling pathways are highly upregulated in Fcgr2b-/- mice, alterations that were diminished in the absence of iRhom2. Pharmacological blockade of either TNF-α or EGFR signaling protected Fcgr2b-/- mice from severe renal damage. Finally, kidneys from LN patients showed increased iRhom2 and HB-EGF expression, with interstitial HB-EGF expression significantly associated with chronicity indices. Our data suggest that activation of iRhom2/ADAM17-dependent TNF-α and EGFR signaling plays a crucial role in mediating irreversible kidney damage in LN, thereby uncovering a target for selective and simultaneous dual inhibition of 2 major pathological pathways in the effector arm of the disease.
Collapse
Affiliation(s)
| | - Yurii Chinenov
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | | | - Michael Madaio
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Joris Jth Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Gregory Farber
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York, USA
| | - Priya D Issuree
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | - Laura Donlin
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA
| | - David R Mcllwain
- Baxter Laboratory in Stem Cell Biology, Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York, USA.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York, USA.,Institute for Advanced Study, Technical University Munich, Munich, Germany.,Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Jane E Salmon
- Program in Inflammation and Autoimmunity, and.,Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
176
|
Role of iRhom2 in intestinal ischemia-reperfusion-mediated acute lung injury. Sci Rep 2018; 8:3797. [PMID: 29491382 PMCID: PMC5830505 DOI: 10.1038/s41598-018-22218-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/19/2018] [Indexed: 11/14/2022] Open
Abstract
Intestinal ischemia-reperfusion (I/R) may cause acute systemic and lung inflammation. However, the detailed mechanism of this inflammatory cascade has not been fully elucidated. Inactive rhomboid protein 2 (iRhom2) is essential for the maturation of TNF-α converting enzyme (TACE), which is required for TNF-α secretion. We evaluated the role of iRhom2 in a mouse model of intestinal I/R using iRhom2 knockout (KO) and wild-type (WT) mice. Lung injury following intestinal I/R was significantly attenuated in iRhom2 KO mice compared with WT mice. After intestinal I/R, lungs from iRhom2 KO mice showed significantly lower myeloperoxidase (MPO) activity and markedly reduced cell apoptosis associated with a decreased level of active caspase 3 and decreased TUNEL staining compared with lungs from WT mice. TNF-α levels were elevated in the serum and lungs of WT mice with intestinal I/R and significantly reduced in iRhom2 KO mice with intestinal I/R. iRhom2 may play a critical role in the pathogenesis of acute lung injury (ALI) after intestinal I/R and thus may be a novel therapeutic target for ALI after intestinal I/R injury.
Collapse
|
177
|
Barnette DN, Cahill TJ, Gunadasa-Rohling M, Carr CA, Freeman M, Riley PR. iRhom2-mediated proinflammatory signalling regulates heart repair following myocardial infarction. JCI Insight 2018; 3:98268. [PMID: 29415889 PMCID: PMC5821194 DOI: 10.1172/jci.insight.98268] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/29/2017] [Indexed: 12/22/2022] Open
Abstract
The role of proinflammation, and specifically TNF-α, on downstream fibrosis and healing after cardiac injury remains unknown. Using iRhom2-deficient mice, which lack myeloid-specific shedding of TNF-α, we reveal increased macrophages (MΦs) that were skewed towards a more proinflammatory (M1) state at day 4, followed by more reparative, antiinflammatory (M2) state at day 7 after myocardial infarction (MI). However, associated functional cytokine expression was significantly reduced in iRhom2-mutant M1 and M2 MΦs, respectively. A dampened proinflammatory signature in iRhom2-deficient mice during the acute phase of injury and subsequent changes in MΦ polarization were associated with reduced phagocytosis and a more sparse distribution within the scar region. This resulted in impaired collagen deposition and fibrosis, and increased left ventricular remodelling and mortality in iRhom2-deficient mice after MI. Our findings reveal a requirement for an iRhom2-mediated proinflammatory response during downstream scarring and fibrosis, which is driven in part by TNF-α signaling. These conclusions challenge the existing model that infarct repair is determined exclusively by antiinflammatory signaling of M2 MΦs, and as such we propose an alternative view of immunomodulation to maintain effective healing after infarction. Optimal scarring and survival after myocardial infarction is dependent upon the initial wave of inflammation after injury.
Collapse
Affiliation(s)
- Damien N Barnette
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom (UK)
| | - Thomas J Cahill
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom (UK).,Department of Cardiovascular Medicine, University of Oxford, Level 6, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom (UK)
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom (UK)
| | - Matthew Freeman
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom (UK)
| |
Collapse
|
178
|
Redundancy of protein disulfide isomerases in the catalysis of the inactivating disulfide switch in A Disintegrin and Metalloprotease 17. Sci Rep 2018; 8:1103. [PMID: 29348576 PMCID: PMC5773583 DOI: 10.1038/s41598-018-19429-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022] Open
Abstract
A Disintegrin and Metalloprotease 17 (ADAM17) can cause the fast release of growth factors and inflammatory mediators from the cell surface. Its activity has to be turned on which occurs by various stimuli. The active form can be inactivated by a structural change in its ectodomain, related to the pattern of the formed disulphide bridges. The switch-off is executed by protein disulfide isomerases (PDIs) that catalyze an isomerization of two disulfide bridges and thereby cause a disulfide switch. We demonstrate that the integrity of the CGHC-motif within the active site of PDIs is indispensable. In particular, no major variation is apparent in the activities of the two catalytic domains of PDIA6. The affinities between PDIA1, PDIA3, PDIA6 and the targeted domain of ADAM17 are all in the nanomolar range and display no significant differences. The redundancy between PDIs and their disulfide switch activity in ectodomains of transmembrane proteins found in vitro appears to be a basic characteristic. However, different PDIs might be required in vivo for disulfide switches in different tissues and under different cellular and physiological situations.
Collapse
|
179
|
Li R, Wang T, Walia K, Gao B, Krepinsky JC. ADAM17 activation and regulation of profibrotic responses by high glucose requires its C-terminus and FAK kinase. J Cell Sci 2018; 131:jcs.208629. [DOI: 10.1242/jcs.208629] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 12/28/2017] [Indexed: 12/23/2022] Open
Abstract
Glomerular matrix accumulation is the hallmark of diabetic nephropathy. The metalloprotease ADAM17 mediates high glucose (HG)-induced matrix production by kidney mesangial cells through release of ligands for the epidermal growth factor receptor. Here we study the mechanism by which HG activates ADAM17. We find that the C-terminus is essential for ADAM17 activation and the profibrotic response to HG. In the C-terminus, Src-mediated Y702 phosphorylation and PI3K/MEK/Erk-mediated T735 phosphorylation are critical to ADAM17 activation, but play divergent roles in ADAM17 trafficking in response to HG. While T735 phosphorylation is required for the HG-induced increase in cell surface mature ADAM17, Y702 phosphorylation is dispensable. Src, however, enables trafficking independently of its phosphorylation of ADAM17. The nonreceptor tyrosine kinase FAK is a central mediator of these processes. These data not only support a critical role for the C-terminus in ADAM17 activation and downstream profibrotic responses to HG, but also highlight FAK as a potential alternate therapeutic target for diabetic nephropathy.
Collapse
Affiliation(s)
- Renzhong Li
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Tony Wang
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Khyati Walia
- Division of Nephrology, McMaster University, Hamilton, Canada
| | - Bo Gao
- Division of Nephrology, McMaster University, Hamilton, Canada
| | | |
Collapse
|
180
|
Embedded in the Membrane: How Lipids Confer Activity and Specificity to Intramembrane Proteases. J Membr Biol 2017; 251:369-378. [PMID: 29260282 DOI: 10.1007/s00232-017-0008-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/12/2017] [Indexed: 10/18/2022]
Abstract
Proteases, sharp yet unforgivable tools of every cell, require tight regulation to ensure specific non-aberrant cleavages. The relatively recent discovered class of intramembrane proteases has gained increasing interest due to their involvement in important signaling pathways linking them to diseases including Alzheimer's disease and cancer. Despite tremendous efforts, their regulatory mechanisms have only started to unravel. There is evidence that the membrane composition itself can regulate intramembrane protease activity and specificity. In this review, we highlight the work on γ-secretase and rhomboid proteases and summarize several studies as to how different lipids impact on enzymatic activity.
Collapse
|
181
|
Liu S, Ye L, Tao J, Ge C, Huang L, Yu J. Total flavones of Abelmoschus manihot improve diabetic nephropathy by inhibiting the iRhom2/TACE signalling pathway activity in rats. PHARMACEUTICAL BIOLOGY 2017; 56:1-11. [PMID: 29221422 PMCID: PMC6130561 DOI: 10.1080/13880209.2017.1412467] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/14/2017] [Accepted: 11/29/2017] [Indexed: 05/21/2023]
Abstract
CONTEXT Total flavones extracted from Abelmoschus manihot L. (Malvaceae) medic (TFA) have been proven clinically effective at improving renal inflammation and glomerular injury in chronic kidney disease (CKD). OBJECTIVE This study evaluated the function of TFA as an inhibitor of iRhom2/TACE (tumour necrosis factor-α converting enzyme) signalling and investigated its anti-DN (diabetic nephropathy) effects in a DN rat model. MATERIALS AND METHODS In vitro, cells were treated with 200 μg/mL advanced glycation end products (AGEs), and then co-cultured with 20 μg/mL TFA for 24 h. Real time PCR, western blotting and co-immunoprecipitation assays were performed. In vivo, DN was induced in 8 week old male Sprague-Dawley rats via unilateral nephrectomy and intraperitoneal injection of streptozotocin, then TFA were administered to rats by gavage for 12 weeks at three different doses (300, 135 and 75 mg/kg/d). 4-Phenylbutanoic acid (2.5 mg/kg/d) was used as a positive control. RESULTS IC50 of TFA is 35.6 μM in HK2 and 39.6 μM in HRMC. TFA treatment (20 μM) inhibited the activation of iRhom2/TACE signalling in cultured cells induced by AGEs. LD50>26 g/kg and ED50=67 mg/kg of TFA in rat by gavage, TFA dose-dependently downregulated the expression of proinflammatory cytokines and exerted anti-inflammatory effects significantly though inhibiting the activation of iRhom2/TACE signalling. DISCUSSION AND CONCLUSIONS Our results show that TFA could dose-dependently ameliorate renal inflammation by inhibiting the activation of iRhom2/TACE signalling and attenuating ER stress. These results suggest that TFA has potential therapeutic value for the treatment of DN in humans.
Collapse
Affiliation(s)
- Su Liu
- Department of Endocrinology, Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lifang Ye
- Department of Endocrinology, Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Tao
- Department of Nephrology, Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chao Ge
- Department of Gastroenterology, Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Liji Huang
- Department of Endocrinology, Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangyi Yu
- Department of Endocrinology, Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- CONTACT Jiangyi YuDepartment of Endocrinology, Jiangsu Province Hosipital of TCM, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
182
|
Chemaly M, McGilligan V, Gibson M, Clauss M, Watterson S, Alexander HD, Bjourson AJ, Peace A. Role of tumour necrosis factor alpha converting enzyme (TACE/ADAM17) and associated proteins in coronary artery disease and cardiac events. Arch Cardiovasc Dis 2017; 110:700-711. [DOI: 10.1016/j.acvd.2017.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/13/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023]
|
183
|
Hosur V, Lyons BL, Burzenski LM, Shultz LD. Tissue-specific role of RHBDF2 in cutaneous wound healing and hyperproliferative skin disease. BMC Res Notes 2017; 10:573. [PMID: 29116018 PMCID: PMC5678570 DOI: 10.1186/s13104-017-2899-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022] Open
Abstract
Objective Gain-of-function (GOF) mutations in RHBDF2 cause tylosis. Patients present with hyperproliferative skin, and keratinocytes from tylosis patients’ skin show an enhanced wound-healing phenotype. The curly bare mouse model of tylosis, carrying a GOF mutation in the Rhbdf2 gene (Rhbdf2cub), presents with epidermal hyperplasia and shows accelerated cutaneous wound-healing phenotype through enhanced secretion of the epidermal growth factor receptor family ligand amphiregulin. Despite these advances in our understanding of tylosis, key questions remain. For instance, it is not known whether the disease is skin-specific, whether the immune system or the surrounding microenvironment plays a role, and whether mouse genetic background influences the hyperproliferative-skin and wound-healing phenotypes observed in Rhbdf2cub mice. Results We performed bone marrow transfers and reciprocal skin transplants and found that bone marrow transfer from C57BL/6 (B6)-Rhbdf2cub/cub donor mice to B6 wildtype recipient mice failed to transfer the hyperproliferative-skin and wound-healing phenotypes in B6 mice. Furthermore, skin grafts from B6 mice to the dorsal skin of B6-Rhbdf2cub/cub mice maintained the phenotype of the donor mice. To test the influence of mouse genetic background, we backcrossed Rhbdf2cub onto the MRL/MpJ strain and found that the hyperproliferative-skin and wound-healing phenotypes caused by the Rhbdf2cub mutation persisted on the MRL/MpJ strain.
Collapse
Affiliation(s)
- Vishnu Hosur
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| | - Bonnie L Lyons
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Lisa M Burzenski
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Leonard D Shultz
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| |
Collapse
|
184
|
Recent Advances in ADAM17 Research: A Promising Target for Cancer and Inflammation. Mediators Inflamm 2017; 2017:9673537. [PMID: 29230082 PMCID: PMC5688260 DOI: 10.1155/2017/9673537] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/15/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023] Open
Abstract
Since its discovery, ADAM17, also known as TNFα converting enzyme or TACE, is now known to process over 80 different substrates. Many of these substrates are mediators of cancer and inflammation. The field of ADAM metalloproteinases is at a crossroad with many of the new potential therapeutic agents for ADAM17 advancing into the clinic. Researchers have now developed potential drugs for ADAM17 that are selective and do not have the side effects which were seen in earlier chemical entities that targeted this enzyme. ADAM17 inhibitors have broad therapeutic potential, with properties ranging from tumor immunosurveillance and overcoming drug and radiation resistance in cancer, as treatments for cardiac hypertrophy and inflammatory conditions such as inflammatory bowel disease and rheumatoid arthritis. This review focuses on substrates and inhibitors identified more recently for ADAM17 and their role in cancer and inflammation.
Collapse
|
185
|
Zunke F, Rose-John S. The shedding protease ADAM17: Physiology and pathophysiology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2059-2070. [DOI: 10.1016/j.bbamcr.2017.07.001] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/08/2017] [Accepted: 07/09/2017] [Indexed: 02/07/2023]
|
186
|
Zhang Y, Li Y, Yang X, Wang J, Wang R, Qian X, Zhang W, Xiao W. Uev1A-Ubc13 catalyzes K63-linked ubiquitination of RHBDF2 to promote TACE maturation. Cell Signal 2017; 42:155-164. [PMID: 29069608 DOI: 10.1016/j.cellsig.2017.10.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/21/2017] [Accepted: 10/21/2017] [Indexed: 01/05/2023]
Abstract
The TNFα-induced NF-κB signaling pathway plays critical roles in multiple biological processes. Extensive studies have explored the mechanisms regulating this signaling cascade, and identified an E2 complex, Uev1A-Ubc13, that mediates K63-linked poly-Ub chain formation and thus recruits NEMO to activate the signaling transduction. In this study, we demonstrate that the Uev1A-Ubc13 complex simultaneously serves as a repressor of the NF-κB pathway. It was found that cells overexpressing UEV1A silence the signal cascade earlier than control cells. Importantly, UEV1A overexpression enhances TACE maturation to shed the TNFα receptor. The Uev1A-Ubc13 complex interacts with RHBDF2, a key factor promoting TACE maturation, and inhibition of the Uev1A-Ubc13 activity interferes with RHBDF2-promoted TACE maturation. Furthermore, upon TNFα stimulation, the Uev1A-Ubc13 complex cooperates with CHIP to promote K63-linked ubiquitination of RHBDF2, enhancing its activity toward TACE maturation and subsequently blocking TNFα-induced NF-κB signaling.
Collapse
Affiliation(s)
- Yiran Zhang
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Yadan Li
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Xiaoran Yang
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Juanjuan Wang
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Ruifeng Wang
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Xianghao Qian
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Weiwei Zhang
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Wei Xiao
- Beijing Key Laboratory of DNA Damage Responses and College of Life Sciences, Capital Normal University, Beijing 100048, China; Department of Microbiology and Immunology, University of Saskatchewan, S7N 5E5, Canada.
| |
Collapse
|
187
|
The role of ADAM17 in the T-cell response against bacterial pathogens. PLoS One 2017; 12:e0184320. [PMID: 28877252 PMCID: PMC5587322 DOI: 10.1371/journal.pone.0184320] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/22/2017] [Indexed: 12/21/2022] Open
Abstract
ADAM17 is a member of the A Disintegrin And Metalloproteinase family of proteases. It is ubiquitously expressed and causes the shedding of a broad spectrum of surface proteins such as adhesion molecules, cytokines and cytokine receptors. By controlled shedding of these proteins from leukocytes, ADAM17 is able to regulate immune responses. Several ADAM17 targets on T cells have been implicated in T-cell migration, differentiation and effector functions. However, the role of ADAM17 in T-cell responses is still unclear. To characterize the function of ADAM17 in T cells, we used Adam17fl/fl×CD4cre+ mice with a T-cell restricted inactivation of the Adam17 gene. Upon stimulation, ADAM17-deficient CD4+ and CD8+ T cells were impaired in shedding of CD62L, IL-6Rα, TNF-α, TNFRI and TNFRII. Surprisingly, we could not detect profound changes in the composition of major T-cell subsets in Adam17fl/fl×CD4cre+ mice. Following infection with Listeria monocytogenes, Adam17fl/fl×CD4cre+ mice mounted regular listeria-specific CD4+ TH1 and CD8+ T-cell responses and were able to control primary and secondary infections. In conclusion, our study indicates that ADAM17 is either not required in T cells under homoeostatic conditions and for control of listeria infection or can be effectively compensated by other mechanisms.
Collapse
|
188
|
Urriola-Muñoz P, Li X, Maretzky T, McIlwain DR, Mak TW, Reyes JG, Blobel CP, Moreno RD. The xenoestrogens biphenol-A and nonylphenol differentially regulate metalloprotease-mediated shedding of EGFR ligands. J Cell Physiol 2017; 233:2247-2256. [PMID: 28703301 DOI: 10.1002/jcp.26097] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 07/11/2017] [Indexed: 12/14/2022]
Abstract
The xenoestrogens bisphenol-A (BPA) and nonylphenol (NP) are endocrine disruptors used in the plastic polymer industry to manufacture different products for human use. Previous studies have suggested a role of these compounds in the shedding of signaling molecules, such as tumor necrosis factor α (TNF-α). The aim of this work was to evaluate the effect of BPA and NP on the sheddase ADAM17 and its newly discovered regulators iRhom1 and iRhom2 in the release of EGFR-ligands. We report that BPA and NP can stimulate the release of the ADAM17-substrates HB-EGF and TGF-α. In cells lacking ADAM17 (Adam17-/- mEFs) BPA-stimulated release of HB-EGF, but not TGF-α, was strongly reduced, whereas NP-stimulated shedding of HB-EGF and TGF-α was completely abolished. Inactivation of both ADAM17 and the related ADAM10 (Adam10/17-/- mEFs) completely prevented the release of these substrates. In the absence of iRhom1, BPA- or NP-stimulated release of HB-EGF or TGF-α was comparable to wild-type control mEFs, conversely the BPA-induced release of HB-EGF was abolished in iRhom2-/- mEFs. The defect in shedding of HB-EGF in iRhom2-/- mEF cells could be rescued by overexpressing iRhom2. Interestingly, the NP-stimulated release of HB-EGF was not affected by the absence of iRhom2, suggesting that NP could potentially activate both ADAM10 and ADAM17. We tested this hypothesis using betacellulin (BTC), an EGFR-ligand that is a substrate for ADAM10. We found that NP, but not BPA stimulated the release of BTC in Adam17-/- , iRhom2-/- , or iRhom1/2-/- , but not in Adam10/17-/- cells. Taken together, our results suggest that BPA and NP stimulate the release of EGFR-ligands by differentially activating ADAM17 or ADAM10. The identification of specific effects of these endocrine disruptors on ADAM10 and ADAM17 will help to provide a better understanding of their roles in cell signaling and proinflammatory processes, and provide new potential targets for treatment of reproductive or inflammatory diseases such as asthma or breast cancer that are promoted by xenoestrogens.
Collapse
Affiliation(s)
- Paulina Urriola-Muñoz
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile.,Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Xue Li
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York.,Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, New York, New York
| | - Thorsten Maretzky
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York
| | - David R McIlwain
- Department of Microbiology and Immunology, Baxter Laboratory in Stem Cell Biology, Stanford University, Stanford, California
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Juan G Reyes
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York.,Institute for Advanced Study, Technische Universität München, Garching, Germany.,Department of Physiology, Biophysics and Systems Biology, Weill Cornell University, New York, New York
| | - Ricardo D Moreno
- Facultad de Ciencias Biológicas, Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
189
|
Luo WW, Shu HB. Emerging roles of rhomboid-like pseudoproteases in inflammatory and innate immune responses. FEBS Lett 2017; 591:3182-3189. [PMID: 28815577 DOI: 10.1002/1873-3468.12796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/28/2017] [Accepted: 08/07/2017] [Indexed: 12/31/2022]
Abstract
Rhomboid-like pseudoproteases are a conserved superfamily of proteins related to the rhomboid intramembrane serine proteases that lack key catalytic residues. iRhom2, a member of the rhomboid-like pseudoprotease superfamily, regulates the maturation and trafficking of ADAM17 and is associated with inflammatory arthritis. Recent studies demonstrate that iRhom2 is also involved in innate immunity by regulating the trafficking and stability of MITA (also called STING), which is a central adaptor in innate antiviral signalling pathways. Here, we summarize recent progress on the roles and mechanisms of iRhom2 and its homologues in innate immunity and also discuss the links between the physiological functions of iRhoms and immunological diseases.
Collapse
Affiliation(s)
- Wei-Wei Luo
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Hong-Bing Shu
- Medical Research Institute, School of Medicine, Wuhan University, China
| |
Collapse
|
190
|
Kappelhoff R, Puente XS, Wilson CH, Seth A, López-Otín C, Overall CM. Overview of transcriptomic analysis of all human proteases, non-proteolytic homologs and inhibitors: Organ, tissue and ovarian cancer cell line expression profiling of the human protease degradome by the CLIP-CHIP™ DNA microarray. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2210-2219. [PMID: 28797648 DOI: 10.1016/j.bbamcr.2017.08.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/01/2017] [Accepted: 08/04/2017] [Indexed: 01/10/2023]
Abstract
The protease degradome is defined as the complete repertoire of proteases and inhibitors, and their nonfunctional homologs present in a cell, tissue or organism at any given time. We review the tissue distribution of virtually the entire degradome in 23 different human tissues and 6 ovarian cancer cell lines. To do so, we developed the CLIP-CHIP™, a custom microarray based on a 70-mer oligonucleotide platform, to specifically profile the transcripts of the entire repertoire of 473 active human proteases, 156 protease inhibitors and 92 non-proteolytically active homologs known at the design date using one specific 70-mer oligonucleotide per transcript. Using the CLIP-CHIP™ we mapped the expression profile of proteases and their inhibitors in 23 different human tissues and 6 ovarian cancer cell lines in 104 sample datasets. Hierarchical cluster analysis showed that expression profiles clustered according to their anatomic locations, cellular composition, physiologic functions, and the germ layer from which they are derived. The human ovarian cancer cell lines cluster according to malignant grade. 110 proteases and 42 inhibitors were tissue specific (1 to 3 tissues). Of these 110 proteases 69% (74) are mainly extracellular, 30% (34) intracellular and 1% intramembrane. Notably, 35% (197/565) of human proteases and 30% (47/156) of inhibitors were ubiquitously expressed in all 23 tissues; 27% (155) of proteases and 21% (32) of inhibitors were broadly expressed in 4-20 tissues. Our datasets provide a valuable resource for the community of baseline protease and inhibitor relative expression in normal human tissues and can be used for comparison with diseased tissue, e.g. ovarian cancer, to decipher pathogenesis, and to aid drug development. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.
Collapse
Affiliation(s)
- Reinhild Kappelhoff
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xose S Puente
- Departamento de Bioquimica y Biologia Molecular, Universidad de Oviedo, Oviedo, Spain
| | - Claire H Wilson
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arun Seth
- Sunnybrook Research Institute, Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Carlos López-Otín
- Departamento de Bioquimica y Biologia Molecular, Universidad de Oviedo, Oviedo, Spain
| | - Christopher M Overall
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
191
|
Murphy JM, Mace PD, Eyers PA. Live and let die: insights into pseudoenzyme mechanisms from structure. Curr Opin Struct Biol 2017; 47:95-104. [PMID: 28787627 DOI: 10.1016/j.sbi.2017.07.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/30/2017] [Accepted: 07/20/2017] [Indexed: 12/22/2022]
Abstract
Pseudoenzymes were first described more than 50 years ago, when it was recognised that a subset of proteins that are structurally homologous to active enzymes lack amino acids necessary for catalytic activity. Recently, interest in pseudoenzymes has surged as it has become apparent that they constitute ∼10% of proteomes and perform essential metabolic and signalling functions that can be experimentally distinguished from catalytic outputs of enzymes. Here, we highlight recent structural studies of pseudoenzymes, which have revealed the molecular basis for roles as allosteric regulators of conventional enzymes, as molecular switches and integrators, as hubs for assembling protein complexes, and as competitors of substrate availability and holoenzyme assembly. As structural studies continue to illuminate pseudoenzyme molecular mechanisms, we anticipate that our knowledge of the breadth of their biological functions will expand in parallel.
Collapse
Affiliation(s)
- James M Murphy
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Peter D Mace
- Biochemistry Department, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.
| |
Collapse
|
192
|
Du X, Jiang C, Lv Y, Dull RO, Zhao YY, Schwartz DE, Hu G. Isoflurane promotes phagocytosis of apoptotic neutrophils through AMPK-mediated ADAM17/Mer signaling. PLoS One 2017; 12:e0180213. [PMID: 28671983 PMCID: PMC5495389 DOI: 10.1371/journal.pone.0180213] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/12/2017] [Indexed: 12/25/2022] Open
Abstract
A patient's recovery from lung inflammatory injury or development of multi-system organ failure is determined by the host's ability to resolve inflammation and repair tissue damage, both of which require the clearance of apoptotic neutrophils by macrophages (efferocytosis). Here, we investigated the effects of isoflurane on macrophage efferocytosis and resolution of lung inflammatory injury. Treatment of murine bone marrow-derived macrophages (BMDMs) or alveolar macrophages with isoflurane dramatically enhanced phagocytosis of apoptotic neutrophils. Isoflurane significantly increased the surface expression of the receptor tyrosine kinase Mer in macrophages, but markedly decreased the levels of a soluble form of Mer protein in the medium. Isoflurane treatment also caused a decrease in a disintegrin and metalloproteinase 17 (ADAM17) on the cell surface and a concomitant increase in its cytoplasmic fraction. These responses induced by isoflurane were completely reversed by a pharmacological inhibitor or genetic deletion of AMP-activated protein kinase (AMPK). In a mouse model of lipopolysaccharide-induced lung injury, isoflurane accelerated the recovery of lung inflammation and injury that was coupled with an increase in the number of alveolar macrophages containing apoptotic bodies. In alveolar macrophage-depleted mice, administration of isoflurane-pretreated BMDMs facilitated resolution of lung inflammation following lipopolysaccharide challenge. Thus, isoflurane promoted resolution of lipopolysaccharide-induced lung inflammatory injury via enhancement of macrophage efferocytosis. Increased macrophage efferocytosis following isoflurane treatment correlates with upregulation of Mer surface expression through AMPK-mediated blockade of ADAM17 trafficking to the cell membrane.
Collapse
Affiliation(s)
- Xueke Du
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chunling Jiang
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Yang Lv
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Randal O. Dull
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - You-Yang Zhao
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - David E. Schwartz
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
193
|
Ishimoto T, Miyake K, Nandi T, Yashiro M, Onishi N, Huang KK, Lin SJ, Kalpana R, Tay ST, Suzuki Y, Cho BC, Kuroda D, Arima K, Izumi D, Iwatsuki M, Baba Y, Oki E, Watanabe M, Saya H, Hirakawa K, Baba H, Tan P. Activation of Transforming Growth Factor Beta 1 Signaling in Gastric Cancer-associated Fibroblasts Increases Their Motility, via Expression of Rhomboid 5 Homolog 2, and Ability to Induce Invasiveness of Gastric Cancer Cells. Gastroenterology 2017; 153:191-204.e16. [PMID: 28390866 DOI: 10.1053/j.gastro.2017.03.046] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 02/16/2017] [Accepted: 03/27/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND & AIMS Fibroblasts that interact with cancer cells are called cancer-associated fibroblasts (CAFs), which promote progression of different tumor types. We investigated the characteristics and functions of CAFs in diffuse-type gastric cancers (DGCs) by analyzing features of their genome and gene expression patterns. METHODS We isolated CAFs and adjacent non-cancer fibroblasts (NFs) from 110 gastric cancer (GC) tissues from patients who underwent gastrectomy in Japan from 2008 through 2016. Cells were identified using specific markers of various cell types by immunoblot and flow cytometry. We selected pairs of CAFs and NFs for whole-exome and RNA sequencing analyses, and compared expression of specific genes using quantitative reverse transcription PCR. Protein levels and phosphorylation were compared by immunoblot and immunofluorescence analyses. Rhomboid 5 homolog 2 (RHBDF2) was overexpressed from a transgene in fibroblasts or knocked down using small interfering RNAs. Motility and invasiveness of isolated fibroblasts and GC cell lines (AGS, KATOIII, MKN45, NUGC3, NUGC4, OCUM-2MD3 and OCUM-12 cell lines) were quantified by real-time imaging analyses. We analyzed 7 independent sets of DNA microarray data from patients with GC and associated expression levels of specific genes with patient survival times. Nude mice were given injections of OCUM-2MD3 in the stomach wall; tumors and metastases were collected and analyzed by immunohistochemistry. RESULTS Many of the genes with increased expression in CAFs compared with NFs were associated with transforming growth factor beta 1 (TGFB1) activity. When CAFs were cultured in extracellular matrix, they became more motile than NFs; DGC cells incubated with CAFs were also more motile and invasive in vitro than DGC cells not incubated with CAFs. When injected into nude mice, CAF-incubated DGC cells invaded a greater number of lymphatic vessels than NF-incubated DGC cells. We identified RHBDF2 as a gene overexpressed in CAFs compared with NFs. Knockdown of RHBDF2 in CAFs reduced their elongation and motility in response to TGFB1, whereas overexpression of RHBDF2 in NFs increased their motility in extracellular matrix. RHBDF2 appeared to regulate oncogenic and non-canonical TGFB1 signaling. Knockdown of RHBDF2 in CAFs reduced cleavage of the TGFB receptor 1 (TGFBR1) by ADAM metallopeptidase domain 17 (ADAM17 or TACE) and reduced expression of genes that regulate motility. Incubation of NFs with in interleukin 1 alpha (IL1A), IL1B or tumor necrosis factor, secreted by DGCs, increased fibroblast expression of RHBDF2. Simultaneous high expression of these cytokines in GC samples was associated with shorter survival times of patients. CONCLUSIONS In CAFs isolated from human DGCs, we observed increased expression of RHBDF2, which regulates TGFB1 signaling. Expression of RHBDF2 in fibroblasts is induced by inflammatory cytokines (such as IL1A, IL1B, and tumor necrosis factor) secreted by DGCs. RHBDF2 promotes cleavage of TGFBR1 by activating TACE and motility of CAFs in response to TGFB1. These highly motile CAFs induce DGCs to invade extracellular matrix and lymphatic vessels in nude mice.
Collapse
Affiliation(s)
- Takatsugu Ishimoto
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore; Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan; International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Keisuke Miyake
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan; International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | | | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Nobuyuki Onishi
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Kie Kyon Huang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | | | | | - Su Ting Tay
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Yuka Suzuki
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Byoung Chul Cho
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Daisuke Kuroda
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Kota Arima
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan; International Research Center of Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Daisuke Izumi
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan.
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore; Genome Institute of Singapore, Singapore.
| |
Collapse
|
194
|
Grötzinger J, Lorenzen I, Düsterhöft S. Molecular insights into the multilayered regulation of ADAM17: The role of the extracellular region. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2088-2095. [PMID: 28571693 DOI: 10.1016/j.bbamcr.2017.05.024] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/25/2017] [Accepted: 05/26/2017] [Indexed: 12/22/2022]
Abstract
In contrast to many other signalling mechanisms shedding of membrane-anchored proteins is an irreversible process. A Disintegrin And Metalloproteinase (ADAM) 17 is one of the major sheddases involved in a variety of physiological and pathophysiological processes including regeneration, differentiation, and cancer progression. Due to its central role in signalling the shedding activity of ADAM17 is tightly regulated, especially on the cell surface, where shedding events take place. The activity of ADAM17 can be subdivided into a catalytic activity and the actual shedding activity. Whereas the catalytic activity is constitutively present, the shedding activity has to be induced and is tightly controlled to prevent pathological situations induced by the release of its substrates. The regulation of the shedding activity of ADAM17 is multilayered and different regions of the protease are involved. Intriguingly, its extracellular domains play crucial roles in different regulatory mechanisms. We will discuss the role of these domains in the control of ADAM17 activity. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.
Collapse
Affiliation(s)
- Joachim Grötzinger
- Institute of Biochemistry, Christian-Albrechts-University, Olshausenstr. 40, 24118 Kiel, Germany.
| | - Inken Lorenzen
- Centre of Biochemistry and Molecular Biology, Structural Biology, Christian-Albrechts-University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Stefan Düsterhöft
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, UK
| |
Collapse
|
195
|
Meprin metalloproteases: Molecular regulation and function in inflammation and fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2096-2104. [PMID: 28502593 DOI: 10.1016/j.bbamcr.2017.05.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 01/03/2023]
Abstract
The zinc-endopeptidases meprin α and meprin β are extracellular proteases involved in connective tissue homeostasis, intestinal barrier function and immunological processes. Meprins are unique among other extracellular proteases with regard to cleavage specificity and structure. Meprin α and meprin β have a strong preference for negatively charged amino acids around the scissile bond, reflected by cleavage sites identified in procollagen I, the amyloid precursor protein (APP) and the interleukin-6 receptor (IL-6R). In this review we report on recent findings that summarize the complex molecular regulation of meprins, particular folding, activation and shedding. Dysregulation of meprin α and meprin β is often associated with pathological conditions such as neurodegeneration, inflammatory bowel disease and fibrosis. Based on mouse models and patient data we suggest meprins as possible key regulators in the onset and progression of fibrotic disorders, leading to severe diseases such as pulmonary hypertension. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.
Collapse
|
196
|
Mishra HK, Ma J, Walcheck B. Ectodomain Shedding by ADAM17: Its Role in Neutrophil Recruitment and the Impairment of This Process during Sepsis. Front Cell Infect Microbiol 2017; 7:138. [PMID: 28487846 PMCID: PMC5403810 DOI: 10.3389/fcimb.2017.00138] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are specialized at killing bacteria and are recruited from the blood in a rapid and robust manner during infection. A cascade of adhesion events direct their attachment to the vascular endothelium and migration into the underlying tissue. A disintegrin and metalloproteinase 17 (ADAM17) functions in the cell membrane of neutrophils and endothelial cells by cleaving its substrates, typically in a cis manner, at an extracellular site proximal to the cell membrane. This process is referred to as ectodomain shedding and it results in the downregulation of various adhesion molecules and receptors, and the release of immune regulating factors. ADAM17 sheddase activity is induced upon cell activation and rapidly modulates intravascular adhesion events in response to diverse environmental stimuli. During sepsis, an excessive systemic inflammatory response against infection, neutrophil migration becomes severely impaired. This involves ADAM17 as indicated by increased levels of its cleaved substrates in the blood of septic patients, and that ADAM17 inactivation improves neutrophil recruitment and bacterial clearance in animal models of sepsis. Excessive ADAM17 sheddase activity during sepsis thus appears to undermine in a direct and indirect manner the necessary balance between intravascular adhesion and de-adhesion events that regulate neutrophil migration into sites of infection. This review provides an overview of ADAM17 function and regulation and its potential contribution to neutrophil dysfunction during sepsis.
Collapse
Affiliation(s)
- Hemant K Mishra
- Department of Veterinary and Biomedical Sciences, University of MinnesotaSt. Paul, MN, USA
| | - Jing Ma
- Department of Veterinary and Biomedical Sciences, University of MinnesotaSt. Paul, MN, USA
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of MinnesotaSt. Paul, MN, USA
| |
Collapse
|
197
|
Grieve AG, Xu H, Künzel U, Bambrough P, Sieber B, Freeman M. Phosphorylation of iRhom2 at the plasma membrane controls mammalian TACE-dependent inflammatory and growth factor signalling. eLife 2017; 6. [PMID: 28432785 PMCID: PMC5436907 DOI: 10.7554/elife.23968] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/20/2017] [Indexed: 12/21/2022] Open
Abstract
Proteolytic cleavage and release from the cell surface of membrane-tethered ligands is an important mechanism of regulating intercellular signalling. TACE is a major shedding protease, responsible for the liberation of the inflammatory cytokine TNFα and ligands of the epidermal growth factor receptor. iRhoms, catalytically inactive members of the rhomboid-like superfamily, have been shown to control the ER-to-Golgi transport and maturation of TACE. Here, we reveal that iRhom2 remains associated with TACE throughout the secretory pathway, and is stabilised at the cell surface by this interaction. At the plasma membrane, ERK1/2-mediated phosphorylation and 14-3-3 protein binding of the cytoplasmic amino-terminus of iRhom2 alter its interaction with mature TACE, thereby licensing its proteolytic activity. We show that this molecular mechanism is responsible for triggering inflammatory responses in primary mouse macrophages. Overall, iRhom2 binds to TACE throughout its lifecycle, implying that iRhom2 is a primary regulator of stimulated cytokine and growth factor signalling. DOI:http://dx.doi.org/10.7554/eLife.23968.001 Injury or infection can cause tissues in the body to become inflamed. The immune system triggers this inflammation to help repair the injury or fight the infection. A signal molecule known as TNF – which is produced by immune cells called macrophages – triggers inflammation. This protein is normally attached to the surface of the macrophage, and it only activates inflammation once it has been cut free. An enzyme called TACE cuts and releases TNF from the surface of macrophages. This enzyme is made inside the cell and is then transported to the surface. On the way, TACE matures from an inactive form to a fully functional enzyme. Previous work revealed that a protein called iRhom2 controls TACE maturation, but it has been unclear whether iRhom2 affects TACE in any additional ways. Grieve et al. studied the relationship between iRhom2 and TACE in more detail. The experiments show two new roles for iRhom2: in protecting TACE from being destroyed at the cell surface, and prompting TACE to release TNF to trigger inflammation. Injury or infection causes small molecules called phosphate groups to be attached to iRhom2 in macrophages, which causes TACE to release TNF. The findings of Grieve et al. provide the first evidence that iRhom2 influences the activity of TACE throughout the enzyme’s lifetime. Excessive inflammation, often triggered by the uncontrolled release of TNF, can lead to rheumatoid arthritis, cancer and many other diseases. Therefore, iRhom2 could be a promising new target for anti-inflammatory drugs that may help to treat these conditions. DOI:http://dx.doi.org/10.7554/eLife.23968.002
Collapse
Affiliation(s)
- Adam Graham Grieve
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Hongmei Xu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Ulrike Künzel
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul Bambrough
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Boris Sieber
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Matthew Freeman
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
198
|
Murphy JM, Farhan H, Eyers PA. Bio-Zombie: the rise of pseudoenzymes in biology. Biochem Soc Trans 2017; 45:537-544. [PMID: 28408493 DOI: 10.1042/bst20160400] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 02/19/2017] [Accepted: 02/21/2017] [Indexed: 11/17/2022]
Abstract
Pseudoenzymes are catalytically dead counterparts of enzymes. Despite their first description some 50 years ago, the importance and functional diversity of these 'fit-for-purpose' polypeptides is only now being appreciated. Pseudoenzymes have been identified throughout all the kingdoms of life and, owing to predicted deficits in enzyme activity due to the absence of catalytic residues, have been variously referred to as pseudoenzymes, non-enzymes, dead enzymes, prozymes or 'zombie' proteins. An important goal of the recent Biochemical Society Pseudoenzymes-focused meeting was to explore the functional and evolutionary diversity of pseudoenzymes and to begin to evaluate their functions in biology, including cell signalling and metabolism. Here, we summarise the impressive breadth of enzyme classes that are known to have pseudoenzyme counterparts and present examples of known cellular functions. We predict that the next decades will represent golden years for the analysis of pseudoenzymes.
Collapse
Affiliation(s)
- James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Hesso Farhan
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, U.K.
| |
Collapse
|
199
|
Miao F, Zhang M, Zhao Y, Li X, Yao R, Wu F, Huang R, Li K, Miao S, Ma C, Ju H, Song W, Wang L. RHBDD1 upregulates EGFR via the AP-1 pathway in colorectal cancer. Oncotarget 2017; 8:25251-25260. [PMID: 28445956 PMCID: PMC5421926 DOI: 10.18632/oncotarget.15694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/24/2017] [Indexed: 01/08/2023] Open
Abstract
Our previous study showed that RHBDD1 can activate the EGFR signaling pathway to promote colorectal cancer growth. In the present study, EGFR was decreased when RHBDD1 was knocked down or inactivated. Further analysis found that c-Jun and EGFR protein expression was decreased in RHBDD1 knockdown and inactivated cells. c-Jun overexpression in RHBDD1-inactivated cells rescued EGFR expression in a dose-dependent manner. RHBDD1 overexpression in RHBDD1-inactivated cells restored EGFR expression, but this effect was counteracted by c-Jun knockdown. Furthermore, EGFR and c-Jun were attenuated in the RHBDD1 knockdown and inactivated groups in animal tumor models. Tissue microarray assays demonstrated a correlation between RHBDD1 and EGFR in colorectal cancer patients. Therefore, our findings indicate that RHBDD1 stimulates EGFR expression by promoting the AP-1 pathway.
Collapse
Affiliation(s)
- Fei Miao
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Mengmeng Zhang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Yuechao Zhao
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Xiaolu Li
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Rongyan Yao
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Fan Wu
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Rong Huang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Kai Li
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Shiying Miao
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Changwu Ma
- Department of Medical Oncology, Chifeng Municipal Hospital, Chifeng 024000, China
| | - Hongge Ju
- Department of Pathology, Baotou Medical College, Baotou 014040, China.,Department of Pathology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China
| | - Wei Song
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| | - Linfang Wang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
200
|
Ostalecki C, Lee JH, Dindorf J, Collenburg L, Schierer S, Simon B, Schliep S, Kremmer E, Schuler G, Baur AS. Multiepitope tissue analysis reveals SPPL3-mediated ADAM10 activation as a key step in the transformation of melanocytes. Sci Signal 2017; 10:10/470/eaai8288. [PMID: 28292959 DOI: 10.1126/scisignal.aai8288] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The evolution of cancer is characterized by the appearance of specific mutations, but these mutations are translated into proteins that must cooperate to induce malignant transformation. Using a systemic approach with the multiepitope ligand cartography (MELC) technology, we analyzed protein expression profiles (PEPs) in nevi and BRAFV600E-positive superficial spreading melanomas (SSMs) from patient tissues to identify key transformation events. The PEPs in nevi and SSMs differed predominantly in the abundance of specific antigens, but the PEPs of nevi- and melanoma-associated keratinocytes gradually changed during the transformation process. A stepwise change in PEP with similar properties occurred in keratinocytes cocultured with melanoma cells. Analysis of the individual steps indicated that activation of the metalloproteinase ADAM10 by signal peptide peptidase-like 3 (SPPL3) triggered by mutant BRAFV600E was a critical transformation event. SPPL3-mediated ADAM10 activation involved the translocation of SPPL3 and ADAM10 into Rab4- or Rab27-positive endosomal compartments. This endosomal translocation, and hence ADAM10 activation, was inhibited by the presence of the tumor suppressor PTEN. Our findings suggest that systematic tissue antigen analysis could complement whole-genome approaches to provide more insight into cancer development.
Collapse
Affiliation(s)
- Christian Ostalecki
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Jung-Hyun Lee
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Jochen Dindorf
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Lena Collenburg
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Stephan Schierer
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Beate Simon
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Stefan Schliep
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, Helmholtz-Zentrum München, Marchioninistraße 25, D-81377 Munich, Germany
| | - Gerold Schuler
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany
| | - Andreas S Baur
- Department of Dermatology, University Hospital Erlangen, Translational Research Center, Schwabachanlage 12, 91054 Erlangen, Germany.
| |
Collapse
|