1
|
Iwakura Y, Kobayashi Y, Namba H, Nawa H, Takei N. Epidermal Growth Factor Suppresses the Development of GABAergic Neurons Via the Modulation of Perineuronal Net Formation in the Neocortex of Developing Rodent Brains. Neurochem Res 2024; 49:1347-1358. [PMID: 38353896 DOI: 10.1007/s11064-024-04122-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 04/04/2024]
Abstract
Previously, we reported that epidermal growth factor (EGF) suppresses GABAergic neuronal development in the rodent cortex. Parvalbumin-positive GABAergic neurons (PV neurons) have a unique extracellular structure, perineuronal nets (PNNs). PNNs are formed during the development of PV neurons and are mainly formed from chondroitin sulfate (CS) proteoglycans (CSPGs). We examined the effect of EGF on CSPG production and PNN formation as a potential molecular mechanism for the inhibition of inhibiting GABAergic neuronal development by EGF. In EGF-overexpressing transgenic (EGF-Tg) mice, the number of PNN-positive PV neurons was decreased in the cortex compared with that in wild-type mice, as in our previous report. The amount of CS and neurocan was also lower in the cortex of EGF-Tg mice, with a similar decrease observed in EGF-treated cultured cortical neurons. PD153035, an EGF receptor (ErbB1) kinase inhibitor, prevented those mentioned above excess EGF-induced reduction in PNN. We explored the molecular mechanism underlying the effect of EGF on PNNs using fluorescent substrates for matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases (ADAMs). EGF increased the enzyme activity of MMPs and ADAMs in cultured neurons. These enzyme activities were also increased in the EGF-Tg mice cortex. GM6001, a broad inhibitor of MMPs and ADAMs, also blocked EGF-induced PNN reductions. Therefore, EGF/EGF receptor signals may regulate PNN formation in the developing cortex.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan.
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan.
| | - Yutaro Kobayashi
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Biochemistry, Graduate School of Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hisaaki Namba
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, 640-8156, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, 640-8156, Japan
| | - Nobuyuki Takei
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
| |
Collapse
|
2
|
Franke AK, Wessolowski C, Thaden V, Müller I, Cornils K. Glyco-binding domain chimeric antigen receptors as a new option for cancer immunotherapy. Gene Ther 2023; 30:603-611. [PMID: 36529796 PMCID: PMC10457195 DOI: 10.1038/s41434-022-00374-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 10/11/2022] [Accepted: 11/08/2022] [Indexed: 12/23/2022]
Abstract
In the last decade, treatment using Chimeric Antigen Receptor (CAR) are largely studied and demonstrate the potential of immunotherapeutic strategies, as seen mainly for blood related cancers. Still, efficient CAR-T cell approaches especially for the treatment of solid tumors are needed. Tn- and Sialyl-Tn antigens are tumor associated carbohydrate antigens correlating with poor prognosis and tumor metastasis on a variety of tumor entities. These glycans can be recognized by CD301 (CLEC10A, MGL), which is a surface receptor found primarily on immune cells. In the present study, we hypothesized, that it is possible to use newly generated CD301-bearing CARs, enabling cytotoxic effector cells to recognize and eliminate breast cancer cells. Thus, we genetically modified human NK92 cells with different chimeric receptors based on the carbohydrate recognition domain (CRD) of human CD301. We assessed their cytotoxic activity in vitro demonstrating the specific recognition of CD301 ligand positive cell lines. These results were confirmed by degranulation assays and in cytokine release assays. Overall, this study demonstrates CD301-CARs represent a cost-effective and fast alternative to conventional scFv CARs for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Vanessa Thaden
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Ingo Müller
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Cornils
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Ishii T, Ruiz‐Torruella M, Kim JY, Kanzaki H, Albassam A, Wisitrasameewong W, Shindo S, Pierrelus R, Heidari A, Kandalam U, Nakamura S, Movila A, Minond D, Kawai T. Soluble Sema4D cleaved from osteoclast precursors by TACE suppresses osteoblastogenesis. J Cell Mol Med 2023; 27:1750-1756. [PMID: 37170687 PMCID: PMC10273054 DOI: 10.1111/jcmm.17416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/27/2022] [Accepted: 05/03/2022] [Indexed: 05/13/2023] Open
Abstract
Bone remodelling is mediated by orchestrated communication between osteoclasts and osteoblasts which, in part, is regulated by coupling and anti-coupling factors. Amongst formally known anti-coupling factors, Semaphorin 4D (Sema4D), produced by osteoclasts, plays a key role in downmodulating osteoblastogenesis. Sema4D is produced in both membrane-bound and soluble forms; however, the mechanism responsible for producing sSema4D from osteoclasts is unknown. Sema4D, TACE and MT1-MMP are all expressed on the surface of RANKL-primed osteoclast precursors. However, only Sema4D and TACE were colocalized, not Sema4D and MT1-MMP. When TACE and MT1-MMP were either chemically inhibited or suppressed by siRNA, TACE was found to be more engaged in shedding Sema4D. Anti-TACE-mAb inhibited sSema4D release from osteoclast precursors by ~90%. Supernatant collected from osteoclast precursors (OC-sup) suppressed osteoblastogenesis from MC3T3-E1 cells, as measured by alkaline phosphatase activity, but OC-sup harvested from the osteoclast precursors treated with anti-TACE-mAb restored osteoblastogenesis activity in a manner that compensates for diminished sSema4D. Finally, systemic administration of anti-TACE-mAb downregulated the generation of sSema4D in the mouse model of critical-sized bone defect, whereas local injection of recombinant sSema4D to anti-TACE-mAb-treated defect upregulated local osteoblastogenesis. Therefore, a novel pathway is proposed whereby TACE-mediated shedding of Sema4D expressed on the osteoclast precursors generates functionally active sSema4D to suppress osteoblastogenesis.
Collapse
Affiliation(s)
- Takenobu Ishii
- Department of OrthodonticsTokyo Dental CollegeChibaJapan
| | | | - Jae Young Kim
- Department of ProsthodonticsYonsei University Dental HospitalSeoulKorea
| | - Hiroyuki Kanzaki
- Department of orthodontics, School of Dental MedicineTsurumi UniversityYokohamaJapan
| | - Abdullah Albassam
- Department of Endodontics, Faculty of DentistryKing Abdulaziz UniversityJeddahSaudi Arabia
| | | | - Satoru Shindo
- Department of Oral Science and Translational Research, College of Dental MedicineNova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Roodelyne Pierrelus
- Department of Oral Science and Translational Research, College of Dental MedicineNova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Alireza Heidari
- Department of Oral Science and Translational Research, College of Dental MedicineNova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Umadevi Kandalam
- Department of Oral Science and Translational Research, College of Dental MedicineNova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Shin Nakamura
- Department of Oral Science and Translational Research, College of Dental MedicineNova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Alexandru Movila
- Department of Oral Science and Translational Research, College of Dental MedicineNova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Dmitriy Minond
- Department of Pharmaceutical Sciences, College of PharmacyNova Southeastern UniversityFort LauderdaleFloridaUSA
| | - Toshihisa Kawai
- Department of Oral Science and Translational Research, College of Dental MedicineNova Southeastern UniversityFort LauderdaleFloridaUSA
- Center for Collaborative Research, Cell Therapy InstituteNova Southeastern UniversityFort LauderdaleFloridaUSA
| |
Collapse
|
4
|
Chang M. Matrix metalloproteinase profiling and their roles in disease. RSC Adv 2023; 13:6304-6316. [PMID: 36825288 PMCID: PMC9942564 DOI: 10.1039/d2ra07005g] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
Matrix metalloproteinases (MMPs) play roles in remodelling of the extracellular matrix that occurs during morphogenesis, repair, and angiogenesis. Dysregulation of extracellular matrix remodelling can lead to cell proliferation, invasion, and tissue fibrosis. Identification of a specific MMP(s) in a disease has been challenging due to the presence of 24 closely-related human MMPs, each existing in three forms, of which only one is active and capable of catalysis. This review focuses on methods for MMP profiling, with particular emphasis on the batimastat affinity resin that binds only to the active forms of MMPs and related ADAMs (a disintegrin and metalloproteinases), which are then identified by mass spectrometry. Use of the batimastat affinity resin has identified targets for intervention in several human diseases.
Collapse
Affiliation(s)
- Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame Notre Dame IN 46556 USA
| |
Collapse
|
5
|
Kwak M, Southard KM, Kim WR, Lin A, Kim NH, Gopalappa R, Lee HJ, An M, Choi SH, Jung Y, Noh K, Farlow J, Georgakopoulos A, Robakis NK, Kang MK, Kutys ML, Seo D, Kim HH, Kim YH, Cheon J, Gartner ZJ, Jun YW. Adherens junctions organize size-selective proteolytic hotspots critical for Notch signalling. Nat Cell Biol 2022; 24:1739-1753. [PMID: 36456828 PMCID: PMC10665132 DOI: 10.1038/s41556-022-01031-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/19/2022] [Indexed: 12/02/2022]
Abstract
Adherens junctions (AJs) create spatially, chemically and mechanically discrete microdomains at cellular interfaces. Here, using a mechanogenetic platform that generates artificial AJs with controlled protein localization, clustering and mechanical loading, we find that AJs also organize proteolytic hotspots for γ-secretase with a spatially regulated substrate selectivity that is critical in the processing of Notch and other transmembrane proteins. Membrane microdomains outside of AJs exclusively organize Notch ligand-receptor engagement (LRE microdomains) to initiate receptor activation. Conversely, membrane microdomains within AJs exclusively serve to coordinate regulated intramembrane proteolysis (RIP microdomains). They do so by concentrating γ-secretase and primed receptors while excluding full-length Notch. AJs induce these functionally distinct microdomains by means of lipid-dependent γ-secretase recruitment and size-dependent protein segregation. By excluding full-length Notch from RIP microdomains, AJs prevent inappropriate enzyme-substrate interactions and suppress spurious Notch activation. Ligand-induced ectodomain shedding eliminates size-dependent segregation, releasing Notch to translocate into AJs for processing by γ-secretase. This mechanism directs radial differentiation of ventricular zone-neural progenitor cells in vivo and more broadly regulates the proteolysis of other large cell-surface receptors such as amyloid precursor protein. These findings suggest an unprecedented role of AJs in creating size-selective spatial switches that choreograph γ-secretase processing of multiple transmembrane proteins regulating development, homeostasis and disease.
Collapse
Affiliation(s)
- Minsuk Kwak
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
| | - Kaden M Southard
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Woon Ryoung Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Annie Lin
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Nam Hyeong Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Ramu Gopalappa
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Minji An
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Seo Hyun Choi
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Yunmin Jung
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Kunwoo Noh
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Justin Farlow
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Anastasios Georgakopoulos
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min K Kang
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew L Kutys
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Daeha Seo
- Department of Physics and Chemistry, DGIST, Daegu, Republic of Korea
| | - Hyongbum Henry Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Ho Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Young-Wook Jun
- Department of Otolaryngology, University of California, San Francisco, CA, USA.
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Qu H, Khalil RA. Role of ADAM and ADAMTS Disintegrin and Metalloproteinases in Normal Pregnancy and Preeclampsia. Biochem Pharmacol 2022; 206:115266. [PMID: 36191626 DOI: 10.1016/j.bcp.2022.115266] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022]
Abstract
Normal pregnancy (NP) involves intricate processes starting with egg fertilization, proceeding to embryo implantation, placentation and gestation, and culminating in parturition. These pregnancy-related processes require marked uteroplacental and vascular remodeling by proteolytic enzymes and metalloproteinases. A disintegrin and metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) are members of the zinc-dependent family of proteinases with highly conserved protein structure and sequence homology, which include a pro-domain, and a metalloproteinase, disintegrin and cysteine-rich domain. In NP, ADAMs and ADAMTS regulate sperm-egg fusion, embryo implantation, trophoblast invasion, placental angiogenesis and spiral arteries remodeling through their ectodomain proteolysis of cell surface cytokines, cadherins and growth factors as well as their adhesion with integrins and cell-cell junction proteins. Preeclampsia (PE) is a serious complication of pregnancy characterized by new-onset hypertension (HTN) in pregnancy (HTN-Preg) at or after 20 weeks of gestation, with or without proteinuria. Insufficient trophoblast invasion of the uterine wall, inadequate expansive remodeling of the spiral arteries, reduced uteroplacental perfusion pressure, and placental ischemia/hypoxia are major initiating events in the pathogenesis of PE. Placental ischemia/hypoxia increase the release of reactive oxygen species (ROS), which lead to aberrant expression/activity of certain ADAMs and ADAMTS. In PE, abnormal expression/activity of specific ADAMs and ADAMTS that function as proteolytic sheddases could alter proangiogenic and growth factors, and promote the release of antiangiogenic factors and inflammatory cytokines into the placenta and maternal circulation leading to generalized inflammation, endothelial cell injury and HTN-Preg, renal injury and proteinuria, and further decreases in uteroplacental blood flow, exaggeration of placental ischemia, and consequently fetal growth restriction. Identifying the role of ADAMs and ADAMTS in NP and PE has led to a better understanding of the underlying molecular and vascular pathways, and advanced the potential for novel biomarkers for prediction and early detection, and new approaches for the management of PE.
Collapse
Affiliation(s)
- Hongmei Qu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA.
| |
Collapse
|
7
|
Conrad C, Yildiz D, Cleary SJ, Margraf A, Cook L, Schlomann U, Panaretou B, Bowser JL, Karmouty-Quintana H, Li J, Berg NK, Martin SC, Aljohmani A, Moussavi-Harami SF, Wang KM, Tian JJ, Magnen M, Valet C, Qiu L, Singer JP, Eltzschig HK, Bertrams W, Herold S, Suttorp N, Schmeck B, Ball ZT, Zarbock A, Looney MR, Bartsch JW. ADAM8 signaling drives neutrophil migration and ARDS severity. JCI Insight 2022; 7:e149870. [PMID: 35132956 PMCID: PMC8855804 DOI: 10.1172/jci.insight.149870] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 12/21/2021] [Indexed: 01/27/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) results in catastrophic lung failure and has an urgent, unmet need for improved early recognition and therapeutic development. Neutrophil influx is a hallmark of ARDS and is associated with the release of tissue-destructive immune effectors, such as matrix metalloproteinases (MMPs) and membrane-anchored metalloproteinase disintegrins (ADAMs). Here, we observed using intravital microscopy that Adam8-/- mice had impaired neutrophil transmigration. In mouse pneumonia models, both genetic deletion and pharmacologic inhibition of ADAM8 attenuated neutrophil infiltration and lung injury while improving bacterial containment. Unexpectedly, the alterations of neutrophil function were not attributable to impaired proteolysis but resulted from reduced intracellular interactions of ADAM8 with the actin-based motor molecule Myosin1f that suppressed neutrophil motility. In 2 ARDS cohorts, we analyzed lung fluid proteolytic signatures and identified that ADAM8 activity was positively correlated with disease severity. We propose that in acute inflammatory lung diseases such as pneumonia and ARDS, ADAM8 inhibition might allow fine-tuning of neutrophil responses for therapeutic gain.
Collapse
Affiliation(s)
- Catharina Conrad
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Daniela Yildiz
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - Simon J. Cleary
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Lena Cook
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Uwe Schlomann
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| | - Barry Panaretou
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jessica L. Bowser
- Department of Pathology & Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Jiwen Li
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nathaniel K. Berg
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Ahmad Aljohmani
- Institute of Experimental and Clinical Pharmacology and Toxicology, PZMS, ZHMB, Saarland University, Homburg, Germany
| | - S. Farshid Moussavi-Harami
- Department of Pediatrics, Division of Pediatric Critical Care, University of California, San Francisco, San Francisco, California, USA
| | - Kristin M. Wang
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jennifer J. Tian
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Mélia Magnen
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Colin Valet
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Longhui Qiu
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jonathan P. Singer
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | | | - Wilhelm Bertrams
- Institute for Lung Research (iLung), Philipps-University, Marburg, Germany
| | - Susanne Herold
- Department of Internal Medicine II, University Medical Center Giessen and Marburg, Giessen, Germany
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Schmeck
- Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
- Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Marburg, Germany
- German Center for Infectious Disease Research (DZIF), Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Zachary T. Ball
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Mark R. Looney
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jörg W. Bartsch
- Department of Neurosurgery/Lab, Faculty of Medicine, Philipps-University, Marburg, Germany
| |
Collapse
|
8
|
ADAM and ADAMTS disintegrin and metalloproteinases as major factors and molecular targets in vascular malfunction and disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:255-363. [PMID: 35659374 PMCID: PMC9231755 DOI: 10.1016/bs.apha.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A Disintegrin and Metalloproteinase (ADAM) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) are two closely related families of proteolytic enzymes. ADAMs are largely membrane-bound enzymes that act as molecular scissors or sheddases of membrane-bound proteins, growth factors, cytokines, receptors and ligands, whereas ADAMTS are mainly secreted enzymes. ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and transmembrane domain. Similarly, ADAMTS family members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but instead of a transmembrane domain they have thrombospondin motifs. Most ADAMs and ADAMTS are activated by pro-protein convertases, and can be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C. Activated ADAMs and ADAMTS participate in numerous vascular processes including angiogenesis, vascular smooth muscle cell proliferation and migration, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs and ADAMTS also play a role in vascular malfunction and cardiovascular diseases such as hypertension, atherosclerosis, coronary artery disease, myocardial infarction, heart failure, peripheral artery disease, and vascular aneurysm. Decreased ADAMTS13 is involved in thrombotic thrombocytopenic purpura and microangiopathies. The activity of ADAMs and ADAMTS can be regulated by endogenous tissue inhibitors of metalloproteinases and other synthetic small molecule inhibitors. ADAMs and ADAMTS can be used as diagnostic biomarkers and molecular targets in cardiovascular disease, and modulators of ADAMs and ADAMTS activity may provide potential new approaches for the management of cardiovascular disorders.
Collapse
|
9
|
Clinical Evaluation of Pathognomonic Salivary Protease Fingerprinting for Oral Disease Diagnosis. J Pers Med 2021; 11:jpm11090866. [PMID: 34575643 PMCID: PMC8472161 DOI: 10.3390/jpm11090866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022] Open
Abstract
Dental decay (Caries) and periodontal disease are globally prevalent diseases with significant clinical need for improved diagnosis. As mediators of dental disease-specific extracellular matrix degradation, proteases are promising analytes. We hypothesized that dysregulation of active proteases can be functionally linked to oral disease status and may be used for diagnosis. To address this, we examined a total of 52 patients with varying oral disease states, including healthy controls. Whole mouth saliva samples and caries biopsies were collected and subjected to analysis. Overall proteolytic and substrate specific activities were assessed using five multiplexed, fluorogenic peptides. Peptide cleavage was further described by inhibitors targeting matrix metalloproteases (MMPs) and cysteine, serine, calpain proteases (CSC). Proteolytic fingerprints, supported by supervised machine-learning analysis, were delineated by total proteolytic activity (PepE) and substrate preference combined with inhibition profiles. Caries and peridontitis showed increased enzymatic activities of MMPs with common (PepA) and divergent substrate cleavage patterns (PepE), suggesting different MMP contribution in particular disease states. Overall, sensitivity and specificity values of 84.6% and 90.0%, respectively, were attained. Thus, a combined analysis of protease derived individual and arrayed substrate cleavage rates in conjunction with inhibitor profiles may represent a sensitive and specific tool for oral disease detection.
Collapse
|
10
|
Stoyell-Conti FF, Itty S, Abraham C, Rigatto K, West CA, Speth RC. 125I-Angiotensin 1-7 binds to a different site than angiotensin 1-7 in tissue membrane preparations. Endocrine 2021; 72:529-538. [PMID: 33415576 DOI: 10.1007/s12020-020-02572-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE To study the receptor for Angiotensin (Ang) 1-7 using a radioligand (125I-Ang 1-7)-binding assay. For more than a decade, Mas has been viewed as the receptor for Ang 1-7; however, Ang 1-7 binding has not been pharmacologically characterized in tissue membrane preparations. METHODS Radioligand-binding assays were carried out using tissue membrane preparations using radioiodinated Angiotensin 1-7 (125I-Ang 1-7) to characterize its binding site. Non-radioactive 127I-Ang 1-7 was used to test if the addition of an iodine to the tyrosine4 moiety of Ang 1-7 changes the ability of Ang 1-7 to competitively inhibit 125I-Ang 1-7 binding. RESULTS 125I-Ang 1-7 binds saturably, with moderately high affinity (10-20 nM) to a binding site in rat liver membranes that is displaceable by 127I-Ang 1-7 at nanomolar concentrations (IC50 = 62 nM) while Ang 1-7 displaces at micromolar concentrations (IC50 = 80 µM) at ~22 °C. This binding was also displaceable by inhibitors of metalloproteases at room temperature. This suggests that 125I-Ang 1-7 binds to MMPs and/or ADAMs as well as other liver membrane elements at ~ 22 °C. However, when 125I-Ang 1-7-binding assays were run at 0-4 °C, the same MMP inhibitors did not effectively compete for 125I-Ang 1-7. CONCLUSIONS The addition of an iodine molecule to the tyrosine in position 4 of Ang 1-7 drastically changes the binding characteristics of this peptide making it unsuitable for characterization of Ang 1-7 receptors.
Collapse
Affiliation(s)
- Filipe F Stoyell-Conti
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Surgery, University of Miami, Miami, FL, USA
| | - Sarin Itty
- Halmos College of Natural Science & Oceanography, Nova Southeastern University, Fort Lauderdale, FL, USA
- Kiran P. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Christy Abraham
- Halmos College of Natural Science & Oceanography, Nova Southeastern University, Fort Lauderdale, FL, USA
- Kiran P. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Katya Rigatto
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Laboratório de Fisiologia Translacional, Universidade Federal de Ciências da Saúde de Porto, Alegre, RS, Brazil
| | - Crystal A West
- Department of Biology, Appalachian State University, North Carolina Research Campus, Kannapolis, NC, USA
| | - Robert C Speth
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA.
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, USA.
| |
Collapse
|
11
|
Chander Y, Kumar R, Khandelwal N, Singh N, Shringi BN, Barua S, Kumar N. Role of p38 mitogen-activated protein kinase signalling in virus replication and potential for developing broad spectrum antiviral drugs. Rev Med Virol 2021; 31:1-16. [PMID: 33450133 DOI: 10.1002/rmv.2217] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) play a key role in complex cellular processes such as proliferation, development, differentiation, transformation and apoptosis. Mammals express at least four distinctly regulated groups of MAPKs which include extracellular signal-related kinases (ERK)-1/2, p38 proteins, Jun amino-terminal kinases (JNK1/2/3) and ERK5. p38 MAPK is activated by a wide range of cellular stresses and modulates activity of several downstream kinases and transcription factors which are involved in regulating cytoskeleton remodeling, cell cycle modulation, inflammation, antiviral response and apoptosis. In viral infections, activation of cell signalling pathways is part of the cellular defense mechanism with the basic aim of inducing an antiviral state. However, viruses can exploit enhanced cell signalling activities to support various stages of their replication cycles. Kinase activity can be inhibited by small molecule chemical inhibitors, so one strategy to develop antiviral drugs is to target these cellular signalling pathways. In this review, we provide an overview on the current understanding of various cellular and viral events regulated by the p38 signalling pathway, with a special emphasis on targeting these events for antiviral drug development which might identify candidates with broad spectrum activity.
Collapse
Affiliation(s)
- Yogesh Chander
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Ram Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Nitin Khandelwal
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India.,Department of Biotechnology, GLA University, Mathura, India
| | - Namita Singh
- Department of Bio and Nano Technology, Guru Jambeshwar University of Science and Technology, Hisar, Haryana, India
| | - Brij Nandan Shringi
- Department of Veterinary Microbiology and Biotechnology, Rajasthan University of Veterinary and Animal Sciences, Bikaner, India
| | - Sanjay Barua
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| | - Naveen Kumar
- National Centre for Veterinary Type Cultures, ICAR-National Research Centre on Equines, Hisar, Haryana, India
| |
Collapse
|
12
|
Pinci F, Gaidt MM, Jung C, Kuut G, Jackson MA, Bauernfried S, Hornung V. C-tag TNF: a reporter system to study TNF shedding. J Biol Chem 2020; 295:18065-18075. [PMID: 33082141 PMCID: PMC7939438 DOI: 10.1074/jbc.ra120.015248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/14/2020] [Indexed: 12/26/2022] Open
Abstract
TNF is a highly pro-inflammatory cytokine that contributes not only to the regulation of immune responses but also to the development of severe inflammatory diseases. TNF is synthesized as a transmembrane protein, which is further matured via proteolytic cleavage by metalloproteases such as ADAM17, a process known as shedding. At present, TNF is mainly detected by measuring the precursor or the mature cytokine of bulk cell populations by techniques such as ELISA or immunoblotting. However, these methods do not provide information on the exact timing and extent of TNF cleavage at single-cell resolution and they do not allow the live visualization of shedding events. Here, we generated C-tag TNF as a genetically encoded reporter to study TNF shedding at the single-cell level. The functionality of the C-tag TNF reporter is based on the exposure of a cryptic epitope on the C terminus of the transmembrane portion of pro-TNF on cleavage. In both denatured and nondenatured samples, this epitope can be detected by a nanobody in a highly sensitive and specific manner only upon TNF shedding. As such, C-tag TNF can successfully be used for the detection of TNF cleavage in flow cytometry and live-cell imaging applications. We furthermore demonstrate its applicability in a forward genetic screen geared toward the identification of genetic regulators of TNF maturation. In summary, the C-tag TNF reporter can be employed to gain novel insights into the complex regulation of ADAM-dependent TNF shedding.
Collapse
Affiliation(s)
- Francesca Pinci
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Moritz M Gaidt
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christophe Jung
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gunnar Kuut
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Margaret A Jackson
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan Bauernfried
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
13
|
Zhang Y, Chen X, Yuan S, Wang L, Guan X. Joint Entropy-Assisted Graphene Oxide-Based Multiplexing Biosensing Platform for Simultaneous Detection of Multiple Proteases. Anal Chem 2020; 92:15042-15049. [PMID: 33118812 DOI: 10.1021/acs.analchem.0c03007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Due to the limited clinical utility of individual biomarkers, there is growing recognition of the need for combining multiple biomarkers as a panel to improve the accuracy and efficacy of disease diagnosis and prognosis. The conventional method to detect multiple analyte species is to construct a sensor array, which consists of an array of individual selective probes for different species. In this work, by using cancer biomarker matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases (ADAMs) as model analytes and functionalized nanographene oxide (nGO) as a sensing element, we developed a multiplexing fluorescence sensor in a nonarray format for simultaneous measurement of the activities of multiple proteases. The constructed nGO-based biosensor was rapid, sensitive, and selective and was also utilized for the successful profiling of ADAMs/MMPs in simulated serum samples. Furthermore, we showed that joint entropy and programming could be utilized to guide experiment design, especially in terms of the selection of a subset of proteases from the entire MMPs/ADAMs family as an appropriate biomarker panel. Our developed nGO-based multiplex sensing platform should find useful application in early cancer detection and diagnosis.
Collapse
Affiliation(s)
- Youwen Zhang
- Department of Chemistry, Illinois Institute of Technology, 3101 South Dearborn Street, Chicago, Illinois 60616, United States
| | - Xiaohan Chen
- Department of Chemistry, Illinois Institute of Technology, 3101 South Dearborn Street, Chicago, Illinois 60616, United States
| | - Shaoqing Yuan
- Amazon, 2121 Seventh Avenue, Seattle, Washington 98121, United States
| | - Liang Wang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China.,Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Xiyun Guan
- Department of Chemistry, Illinois Institute of Technology, 3101 South Dearborn Street, Chicago, Illinois 60616, United States
| |
Collapse
|
14
|
Maurer DM, Adamik J, Santos PM, Shi J, Shurin MR, Kirkwood JM, Storkus WJ, Butterfield LH. Dysregulated NF-κB-Dependent ICOSL Expression in Human Dendritic Cell Vaccines Impairs T-cell Responses in Patients with Melanoma. Cancer Immunol Res 2020; 8:1554-1567. [PMID: 33051240 PMCID: PMC8018573 DOI: 10.1158/2326-6066.cir-20-0274] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/02/2020] [Accepted: 09/18/2020] [Indexed: 12/25/2022]
Abstract
Therapeutic cancer vaccines targeting melanoma-associated antigens are commonly immunogenic but are rarely effective in promoting objective clinical responses. To identify critical molecules for activation of effective antitumor immunity, we have profiled autologous dendritic cell (DC) vaccines used to treat 35 patients with melanoma. We showed that checkpoint molecules induced by ex vivo maturation correlated with in vivo DC vaccine activity. Melanoma patient DCs had reduced expression of cell surface inducible T-cell costimulator ligand (ICOSL) and had defective intrinsic NF-κB signaling. Chromatin immunoprecipitation assays revealed NF-κB-dependent transcriptional regulation of ICOSL expression by DCs. Blockade of ICOSL on DCs reduced priming of antigen-specific CD8+ and CD4+ T cells from naïve donors in vitro Concentration of extracellular/soluble ICOSL released from vaccine DCs positively correlated with patient clinical outcomes, which we showed to be partially regulated by ADAM10/17 sheddase activity. These data point to the critical role of canonical NF-κB signaling, the regulation of matrix metalloproteinases, and DC-derived ICOSL in the specific priming of cognate T-cell responses in the cancer setting. This study supports the implementation of targeted strategies to augment these pathways for improved immunotherapeutic outcomes in patients with cancer.
Collapse
Affiliation(s)
- Deena M Maurer
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Juraj Adamik
- Parker Institute for Cancer Immunotherapy, and University of California San Francisco, Microbiology and Immunology, San Francisco, California
| | - Patricia M Santos
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jian Shi
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael R Shurin
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John M Kirkwood
- UPMC Hillman Cancer Center, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Walter J Storkus
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lisa H Butterfield
- Parker Institute for Cancer Immunotherapy, and University of California San Francisco, Microbiology and Immunology, San Francisco, California.
| |
Collapse
|
15
|
Adem S, Jain S, Sveiven M, Zhou X, O'Donoghue AJ, Hall DA. Giant magnetoresistive biosensors for real-time quantitative detection of protease activity. Sci Rep 2020; 10:7941. [PMID: 32409675 PMCID: PMC7224196 DOI: 10.1038/s41598-020-62910-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/18/2020] [Indexed: 12/19/2022] Open
Abstract
Proteases are enzymes that cleave proteins and are crucial to physiological processes such as digestion, blood clotting, and wound healing. Unregulated protease activity is a biomarker of several human diseases. Synthetic peptides that are selectively hydrolyzed by a protease of interest can be used as reporter substrates of unregulated protease activity. We developed an activity-based protease sensor by immobilizing magnetic nanoparticles (MNPs) to the surface of a giant magnetoresistive spin-valve (GMR SV) sensor using peptides. Cleavage of these peptides by a protease releases the magnetic nanoparticles resulting in a time-dependent change in the local magnetic field. Using this approach, we detected a significant release of MNPs after 3.5 minutes incubation using just 4 nM of the cysteine protease, papain. In addition, we show that proteases in healthy human urine do not release the MNPs, however addition of 20 nM of papain to the urine samples resulted in a time-dependent change in magnetoresistance. This study lays the foundation for using GMR SV sensors as a platform for real-time, quantitative detection of protease activity in biological fluids.
Collapse
Affiliation(s)
- Sandeep Adem
- University of California - San Diego, Department of Bioengineering, La Jolla, CA, 92093, USA
| | - Sonal Jain
- University of California - San Diego, Department of Bioengineering, La Jolla, CA, 92093, USA
| | - Michael Sveiven
- University of California - San Diego, Department of Bioengineering, La Jolla, CA, 92093, USA
| | - Xiahan Zhou
- University of California - San Diego, Department of Electrical and Computer Engineering, La Jolla, CA, 92093, USA
| | - Anthony J O'Donoghue
- University of California - San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, 92093, USA.
| | - Drew A Hall
- University of California - San Diego, Department of Bioengineering, La Jolla, CA, 92093, USA.
- University of California - San Diego, Department of Electrical and Computer Engineering, La Jolla, CA, 92093, USA.
| |
Collapse
|
16
|
Swindell WR, Bojanowski K, Chaudhuri RK. A Zingerone Analog, Acetyl Zingerone, Bolsters Matrisome Synthesis, Inhibits Matrix Metallopeptidases, and Represses IL-17A Target Gene Expression. J Invest Dermatol 2020; 140:602-614.e15. [DOI: 10.1016/j.jid.2019.07.715] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/02/2019] [Accepted: 07/30/2019] [Indexed: 01/27/2023]
|
17
|
Proteolytic processing of PD-L1 by ADAM proteases in breast cancer cells. Cancer Immunol Immunother 2019; 69:43-55. [PMID: 31796994 DOI: 10.1007/s00262-019-02437-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/27/2019] [Indexed: 12/31/2022]
Abstract
Expression of programmed death ligand 1 (PD-L1) on the surface of tumor cells and its interaction with programmed cell death protein 1 (PD-1) on tumor-infiltrating lymphocytes suppress anti-tumor immunity. In breast tumors, PD-L1 expression levels are the highest in estrogen receptor-negative, progesterone receptor-negative, and human epidermal growth factor receptor 2-negative (triple-negative) cancers. In this study, we show that a portion of PD-L1 exogenously expressed in several triple-negative breast cancer cell lines, as well as endogenous PD-L1, is proteolytically cleaved by cell surface metalloproteases. The cleavage generates an ~ 37-kDa N-terminal PD-L1 fragment that is released to the media and a C-terminal PD-L1 fragment that remains associated with cells but is efficiently eliminated by lysosomal degradation. We identify ADAM10 and ADAM17, two closely related members of the ADAM family of cell surface metalloproteases, as enzymes mediating PD-L1 cleavage. Notably, treatment of cells with ionomycin, a calcium ionophore and a known activator of ADAM10, or with phorbol 12-myristate 13-acetate, an activator of ADAM17, dramatically increases the release of soluble PD-L1 to the media. We postulate that ADAM10 and/or ADAM17 may contribute to the regulation of the PD-L1/PD-1 pathway and, ultimately, to anti-tumor immunity in triple-negative breast cancer.
Collapse
|
18
|
Single-cell proteolytic activity measurement using microfluidics for rare cell populations. Methods Enzymol 2019. [PMID: 31668226 DOI: 10.1016/bs.mie.2019.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Proteolytic degradation of the extracellular matrix represents a key step in cancer dissemination and metastasis. To probe cellular proteolytic activity, fluorescent sensing substrate was developed, yet prior studies focused on average activity of thousands of cells. Considerable evidence suggests a specialized subset of cancer cells are driving metastasis, highlighting the value of single-cell approach to reveal cancer cellular heterogeneity. In addition, when only a small number of cells are available, single-cell analysis is required to draw a statistical conclusion. Here, we present a microfluidic platform that provides high-efficiency cell loading and simple valveless isolation, so the proteolytic activity of a small number (10-100) of cells can be individually characterized. Furthermore, the platform allows monitoring single cells at multiple time points for the investigation of dynamics in proteolytic activity. The presented platform represents a simple and reliable tool for single-cell proteolytic analysis, illuminating the heterogeneous and dynamic nature of cancer cells.
Collapse
|
19
|
Albrechtsen R, Wewer Albrechtsen NJ, Gnosa S, Schwarz J, Dyrskjøt L, Kveiborg M. Identification of ADAM12 as a Novel Basigin Sheddase. Int J Mol Sci 2019; 20:ijms20081957. [PMID: 31013576 PMCID: PMC6514901 DOI: 10.3390/ijms20081957] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 02/08/2023] Open
Abstract
The transmembrane glycoprotein basigin, a member of the immunoglobulin superfamily, stimulates matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) degradation and thereby drives cancer cell invasion. Basigin is proteolytically shed from the cell surface and high concentrations of soluble basigin in the blood dictates poor prognosis in cancer patients. A positive correlation between basigin and a disintegrin and metalloproteinase (ADAM)-12 in serum from prostate cancer patients has been reported. Yet, the functional relevance of this correlation is unknown. Here, we show that ADAM12 interacts with basigin and cleaves it in the juxtamembrane region. Specifically, overexpression of ADAM12 increases ectodomain shedding of an alkaline phosphatase-tagged basigin reporter protein from the cell surface. Moreover, CRISPR/Cas9-mediated knockout of ADAM12 in human HeLa carcinoma cells results in reduced shedding of the basigin reporter, which can be rescued by ADAM12 re-expression. We detected endogenous basigin fragments, corresponding to the expected size of the ADAM12-generated ectodomain, in conditioned media from ADAM12 expressing cancer cell-lines, as well as serum samples from a healthy pregnant donor and five bladder cancer patients, known to contain high ADAM12 levels. Supporting the cancer relevance of our findings, we identified several cancer-associated mutations in the basigin membrane proximal region. Subsequent in vitro expression showed that some of these mutants are more prone to ADAM12-mediated shedding and that the shed ectodomain can enhance gelatin degradation by cancer cells. In conclusion, we identified ADAM12 as a novel basigin sheddase with a potential implication in cancer.
Collapse
Affiliation(s)
- Reidar Albrechtsen
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences and Department of Clinical Biochemistry, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Sebastian Gnosa
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Jeanette Schwarz
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Lars Dyrskjøt
- Department of Molecular Medicine (MOMA), Aarhus University Hospital, 8200 Aarhus, Denmark.
| | - Marie Kveiborg
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
20
|
Zhong S, Khalil RA. A Disintegrin and Metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) family in vascular biology and disease. Biochem Pharmacol 2019; 164:188-204. [PMID: 30905657 DOI: 10.1016/j.bcp.2019.03.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022]
Abstract
A Disintegrin and Metalloproteinase (ADAM) is a family of proteolytic enzymes that possess sheddase function and regulate shedding of membrane-bound proteins, growth factors, cytokines, ligands and receptors. Typically, ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and a characteristic transmembrane domain. Most ADAMs are activated by proprotein convertases, but can also be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C activators. A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) is a family of secreted enzymes closely related to ADAMs. Like ADAMs, ADAMTS members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but they lack a transmembrane domain and instead have characteristic thrombospondin motifs. Activated ADAMs perform several functions and participate in multiple cardiovascular processes including vascular smooth muscle cell proliferation and migration, angiogenesis, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs may also be involved in pathological conditions and cardiovascular diseases such as atherosclerosis, hypertension, aneurysm, coronary artery disease, myocardial infarction and heart failure. Like ADAMs, ADAMTS have a wide-spectrum role in vascular biology and cardiovascular pathophysiology. ADAMs and ADAMTS activity is naturally controlled by endogenous inhibitors such as tissue inhibitors of metalloproteinases (TIMPs), and their activity can also be suppressed by synthetic small molecule inhibitors. ADAMs and ADAMTS can serve as important diagnostic biomarkers and potential therapeutic targets for cardiovascular disorders. Natural and synthetic inhibitors of ADAMs and ADAMTS could be potential therapeutic tools for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Sheng Zhong
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Ravindra KC, Ahrens CC, Wang Y, Ramseier JY, Wishnok JS, Griffith LG, Grodzinsky AJ, Tannenbaum SR. Chemoproteomics of matrix metalloproteases in a model of cartilage degeneration suggests functional biomarkers associated with posttraumatic osteoarthritis. J Biol Chem 2018; 293:11459-11469. [PMID: 29794029 DOI: 10.1074/jbc.m117.818542] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 05/21/2018] [Indexed: 12/14/2022] Open
Abstract
Active matrix metalloproteases (MMPs) play a significant role in the pathogenesis of many diseases including osteoarthritis (OA), which involves progressive proteolytic degradation of cartilage. Clinical success of OA interventions that target MMPs has been limited by a lack of information about the presence and activity of specific disease-related proteases. We therefore developed a chemoproteomics approach based on MS to characterize the release and activity of MMPs in an in vitro model of the early inflammatory phase of posttraumatic OA (PTOA). We designed and synthesized chemical activity-based probes (ABPs) to identify active MMPs in bovine cartilage explants cultured for 30 days with the proinflammatory cytokine, interleukin-1α. Using these probes in an activity-based protein profiling-multidimensional identification technology (ABPP-MudPIT) approach, we identified active MMP-1, -2, -3, -7, -9, -12, and -13 in the medium after 10 days of culture, the time at which irreversible proteolysis of the collagen network in the explant was detected using proteolytic activation of FRET-quenched MMP substrates. Total MMP levels were quantified by shotgun proteomics, which, taken with ABPP-MudPIT data, indicated the presence of predominantly inactive MMPs in the culture medium. The selectivity of the ABPP-MudPIT approach was further validated by detection of specific endogenous MMPs activated de novo with 4-aminophenylmurcuric acetate. The utility of the new ABPP-MudPIT approach for detecting molecular biomarkers of PTOA disease initiation and potential targets for therapeutics motivates possible application in other diseases involving MMP activity.
Collapse
Affiliation(s)
- Kodihalli C Ravindra
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Caroline C Ahrens
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Yang Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Julie Y Ramseier
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - John S Wishnok
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Steven R Tannenbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139.
| |
Collapse
|
22
|
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: 19] [Impact Index Per Article: 3.2] [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
|
23
|
Determining the Substrate Specificity of Matrix Metalloproteases using Fluorogenic Peptide Substrates. Methods Mol Biol 2018. [PMID: 28299736 DOI: 10.1007/978-1-4939-6863-3_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
A continuous assay method, such as the one that utilizes an increase in fluorescence upon hydrolysis, allows for rapid and convenient kinetic evaluation of proteases. To better understand MMP behaviors toward native substrates, a variety of fluorescence resonance energy transfer (FRET)/intramolecular fluorescence energy transfer (IFET) triple-helical substrates have been constructed to examine the collagenolytic activity of MMP family members. Results of these studies have been valuable for providing insights into (a) the relative triple-helical peptidase activities of the various collagenolytic MMPs, (b) the collagen preferences of these MMPs, and (c) the relative roles of MMP domains and specific residues in efficient collagenolysis. The present chapter provides an overview of MMP FRET triple-helical substrates and describes how to construct and utilize these substrates.
Collapse
|
24
|
Yoneyama T, Gorry M, Miller MA, Gaither-Davis A, Lin Y, Moss ML, Griffith LG, Lauffenburger DA, Stabile LP, Herman JG, Vujanovic NL. Modification of proteolytic activity matrix analysis (PrAMA) to measure ADAM10 and ADAM17 sheddase activities in cell and tissue lysates. J Cancer 2017; 8:3916-3932. [PMID: 29187866 PMCID: PMC5705993 DOI: 10.7150/jca.20779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/06/2017] [Indexed: 01/29/2023] Open
Abstract
Increases in expression of ADAM10 and ADAM17 genes and proteins have been evaluated, but not validated as cancer biomarkers. Specific enzyme activities better reflect enzyme cellular functions, and might be better biomarkers than enzyme genes or proteins. However, no high throughput assay is available to test this possibility. Recent studies have developed the high throughput real-time proteolytic activity matrix analysis (PrAMA) that integrates the enzymatic processing of multiple enzyme substrates with mathematical-modeling computation. The original PrAMA measures with significant accuracy the activities of individual metalloproteinases expressed on live cells. To make the biomarker assay usable in clinical practice, we modified PrAMA by testing enzymatic activities in cell and tissue lysates supplemented with broad-spectrum non-MP enzyme inhibitors, and by maximizing the assay specificity using systematic mathematical-modeling analyses. The modified PrAMA accurately measured the absence and decreases of ADAM10 sheddase activity (ADAM10sa) and ADAM17sa in ADAM10-/- and ADAM17-/- mouse embryonic fibroblasts (MEFs), and ADAM10- and ADAM17-siRNA transfected human cancer cells, respectively. It also measured the restoration and inhibition of ADAM10sa in ADAM10-cDNA-transfected ADAM10-/- MEFs and GI254023X-treated human cancer cell and tissue lysates, respectively. Additionally, the modified PrAMA simultaneously quantified with significant accuracy ADAM10sa and ADAM17sa in multiple human tumor specimens, and showed the essential characteristics of a robust high throughput multiplex assay that could be broadly used in biomarker studies. Selectively measuring specific enzyme activities, this new clinically applicable assay is potentially superior to the standard protein- and gene-expression assays that do not distinguish active and inactive enzyme forms.
Collapse
Affiliation(s)
- Toshie Yoneyama
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, PA.,VAPHS, Pittsburgh, PA
| | - Michael Gorry
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, PA.,VAPHS, Pittsburgh, PA
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Autumn Gaither-Davis
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Yan Lin
- Department of Biostatistics, University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | | | - Linda G Griffith
- Department of Biologic Engineering, Massachusetts Institute of Technology
| | | | - Laura P Stabile
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - James G Herman
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Nikola L Vujanovic
- Department of Pathology, University of Pittsburgh Cancer Institute, Pittsburgh, PA.,Department of Immunology, University of Pittsburgh Cancer Institute, Pittsburgh, PA.,VAPHS, Pittsburgh, PA
| |
Collapse
|
25
|
Chen R, Jin G, McIntyre TM. The soluble protease ADAMDEC1 released from activated platelets hydrolyzes platelet membrane pro-epidermal growth factor (EGF) to active high-molecular-weight EGF. J Biol Chem 2017; 292:10112-10122. [PMID: 28455445 DOI: 10.1074/jbc.m116.771642] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/20/2017] [Indexed: 12/11/2022] Open
Abstract
Platelets are the sole source of EGF in circulation, yet how EGF is stored or released from stimulated cells is undefined. In fact, we found platelets did not store EGF, synthesized as a single 6-kDa domain in pro-EGF, but rather expressed intact pro-EGF precursor on granular and plasma membranes. Activated platelets released high-molecular-weight (HMW)-EGF, produced by a single cleavage between the EGF and the transmembrane domains of pro-EGF. We synthesized a fluorogenic peptide encompassing residues surrounding the putative sessile arginyl residue and found stimulated platelets released soluble activity that cleaved this pro-EGF1020-1027 peptide. High throughput screening identified chymostatins, bacterial peptides with a central cyclic arginyl structure, as inhibitors of this activity. In contrast, the matrix metalloproteinase/TACE (tumor necrosis factor-α-converting enzyme) inhibitor GM6001 was ineffective. Stimulated platelets released the soluble protease ADAMDEC1, recombinant ADAMDEC1 hydrolyzed pro-EGF1020-1027, and this activity was inhibited by chymostatin and not GM6001. Biotinylating platelet surface proteins showed ADAMDEC1 hydrolyzed surface pro-EGF to HMW-EGF that stimulated HeLa EGF receptor (EGFR) reporter cells and EGFR-dependent tumor cell migration. This proteolysis was inhibited by chymostatin and not GM6001. Metabolizing pro-EGF Arg1023 to citrulline with recombinant polypeptide arginine deiminase 4 (PAD4) abolished ADAMDEC1-catalyzed pro-EGF1020-1027 peptidolysis, while pretreating intact platelets with PAD4 suppressed ADAMDEC1-, thrombin-, or collagen-induced release of HMW-EGF. We conclude that activated platelets release ADAMDEC1, which hydrolyzes pro-EGF to soluble HMW-EGF, that HMW-EGF is active, that proteolytic cleavage of pro-EGF first occurs at the C-terminal arginyl residue of the EGF domain, and that proteolysis is the regulated and rate-limiting step in generating soluble EGF bioactivity from activated platelets.
Collapse
Affiliation(s)
- Rui Chen
- From the Departments of Cellular and Molecular Medicine and
| | - Ge Jin
- the Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, Ohio 44106
| | - Thomas M McIntyre
- From the Departments of Cellular and Molecular Medicine and .,Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland 44195 Ohio and
| |
Collapse
|
26
|
Characterization of the catalytic properties of the membrane-anchored metalloproteinase ADAM9 in cell-based assays. Biochem J 2017; 474:1467-1479. [PMID: 28264989 DOI: 10.1042/bcj20170075] [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: 01/19/2017] [Revised: 03/01/2017] [Accepted: 03/06/2017] [Indexed: 11/17/2022]
Abstract
ADAM9 (A Disintegrin And Metalloprotease 9) is a membrane-anchored metalloproteinase that has been implicated in pathological retinal neovascularization and in tumor progression. ADAM9 has constitutive catalytic activity in both biochemical and cell-based assays and can cleave several membrane proteins, including epidermal growth factor and Ephrin receptor B4; yet little is currently known about the catalytic properties of ADAM9 and its post-translational regulation and inhibitor profile in cell-based assays. To address this question, we monitored processing of the membrane-anchored Ephrin receptor B4 (EphB4) by co-expressing ADAM9, with the catalytically inactive ADAM9 E > A mutant serving as a negative control. We found that ADAM9-dependent shedding of EphB4 was not stimulated by three commonly employed activators of ADAM-dependent ectodomain shedding: phorbol esters, pervanadate or calcium ionophores. With respect to the inhibitor profile, we found that ADAM9 was inhibited by the hydroxamate-based metalloprotease inhibitors marimastat, TAPI-2, BB94, GM6001 and GW280264X, and by 10 nM of the tissue inhibitor of metalloproteinases (TIMP)-3, but not by up to 20 nM of TIMP-1 or -2. Additionally, we screened a non-hydroxamate small-molecule library for novel ADAM9 inhibitors and identified four compounds that selectively inhibited ADAM9-dependent proteolysis over ADAM10- or ADAM17-dependent processing. Taken together, the present study provides new information about the molecular fingerprint of ADAM9 in cell-based assays by showing that it is not stimulated by strong activators of ectodomain shedding and by defining a characteristic inhibitor profile. The identification of novel non-hydroxamate inhibitors of ADAM9 could provide the basis for designing more selective compounds that block the contribution of ADAM9 to pathological neovascularization and cancer.
Collapse
|
27
|
Iwakura Y, Wang R, Inamura N, Araki K, Higashiyama S, Takei N, Nawa H. Glutamate-dependent ectodomain shedding of neuregulin-1 type II precursors in rat forebrain neurons. PLoS One 2017; 12:e0174780. [PMID: 28350885 PMCID: PMC5370147 DOI: 10.1371/journal.pone.0174780] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 03/15/2017] [Indexed: 01/30/2023] Open
Abstract
The neurotrophic factor neuregulin 1 (NRG1) regulates neuronal development, glial differentiation, and excitatory synapse maturation. NRG1 is synthesized as a membrane-anchored precursor and is then liberated by proteolytic processing or exocytosis. Mature NRG1 then binds to its receptors expressed by neighboring neurons or glial cells. However, the molecular mechanisms that govern this process in the nervous system are not defined in detail. Here we prepared neuron-enriched and glia-enriched cultures from embryonic rat neocortex to investigate the role of neurotransmitters that regulate the liberation/release of NRG1 from the membrane of neurons or glial cells. Using a two-site enzyme immunoassay to detect soluble NRG1, we show that, of various neurotransmitters, glutamate was the most potent inducer of NRG1 release in neuron-enriched cultures. NRG1 release in glia-enriched cultures was relatively limited. Furthermore, among glutamate receptor agonists, N-Methyl-D-Aspartate (NMDA) and kainate (KA), but not AMPA or tACPD, mimicked the effects of glutamate. Similar findings were acquired from analysis of the hippocampus of rats with KA-induced seizures. To evaluate the contribution of members of a disintegrin and metalloproteinase (ADAM) families to NRG1 release, we transfected primary cultures of neurons with cDNA vectors encoding NRG1 types I, II, or III precursors, each tagged with the alkaline phosphatase reporter. Analysis of alkaline phosphatase activity revealed that the NRG1 type II precursor was subjected to tumor necrosis factor-α-converting enzyme (TACE) / a Disintegrin And Metalloproteinase 17 (ADAM17) -dependent ectodomain shedding in a protein kinase C-dependent manner. These results suggest that glutamatergic neurotransmission positively regulates the ectodomain shedding of NRG1 type II precursors and liberates the active NRG1 domain in an activity-dependent manner.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- * E-mail:
| | - Ran Wang
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Naoko Inamura
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kazuaki Araki
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shigeki Higashiyama
- Department of Biochemistry and Molecular Genetics, Ehime University, Graduate School of Medicine, Ehime, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
28
|
Taggart C, Mall MA, Lalmanach G, Cataldo D, Ludwig A, Janciauskiene S, Heath N, Meiners S, Overall CM, Schultz C, Turk B, Borensztajn KS. Protean proteases: at the cutting edge of lung diseases. Eur Respir J 2017; 49:49/2/1501200. [PMID: 28179435 DOI: 10.1183/13993003.01200-2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/27/2016] [Indexed: 12/14/2022]
Abstract
Proteases were traditionally viewed as mere protein-degrading enzymes with a very restricted spectrum of substrates. A major expansion in protease research has uncovered a variety of novel substrates, and it is now evident that proteases are critical pleiotropic actors orchestrating pathophysiological processes. Recent findings evidenced that the net proteolytic activity also relies upon interconnections between different protease and protease inhibitor families in the protease web.In this review, we provide an overview of these novel concepts with a particular focus on pulmonary pathophysiology. We describe the emerging roles of several protease families including cysteine and serine proteases.The complexity of the protease web is exemplified in the light of multidimensional regulation of serine protease activity by matrix metalloproteases through cognate serine protease inhibitor processing. Finally, we will highlight how deregulated protease activity during pulmonary pathogenesis may be exploited for diagnosis/prognosis purposes, and utilised as a therapeutic tool using nanotechnologies.Considering proteases as part of an integrative biology perspective may pave the way for the development of new therapeutic targets to treat pulmonary diseases related to intrinsic protease deregulation.
Collapse
Affiliation(s)
- Clifford Taggart
- Airway Innate Immunity Research group (AiiR), Centre for Experimental Medicine, Queen's University Belfast, UK
| | - Marcus A Mall
- Dept of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany.,Division of Pediatric Pulmonology & Allergy and Cystic Fibrosis Center, Dept of Pediatrics, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Gilles Lalmanach
- INSERM UMR1100 Centre d'Etude des Pathologies Respiratoires (CEPR), Equipe: Mécanismes Protéolytiques dans l'Inflammation, Université François Rabelais, Tours, France
| | - Didier Cataldo
- Laboratory of Tumors and Development and Dept of Respiratory Diseases, University of Liege, Liege, Belgium
| | - Andreas Ludwig
- Inflammation Pharmacology Research Group, Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Sabina Janciauskiene
- Dept of Respiratory Medicine, a member of The German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Nicole Heath
- Division of Pediatric Pulmonology & Allergy and Cystic Fibrosis Center, Dept of Pediatrics, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christopher M Overall
- Centre for Blood Research, Dept of Oral Biological and Medical Research University of British Columbia, Vancouver, BC, Canada
| | - Carsten Schultz
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Boris Turk
- Dept of Biochemistry & Molecular & Structural Biology, J. Stefan Institute, Ljubljana, Slovenia
| | - Keren S Borensztajn
- INSERM UMR _S933, Université Pierre et Marie Curie, Paris, France .,INSERM UMR1152 Université Paris Diderot, Faculté de Médecine - site Bichat, Paris, France
| |
Collapse
|
29
|
Conrad C, Miller MA, Bartsch JW, Schlomann U, Lauffenburger DA. Simultaneous Detection of Metalloprotease Activities in Complex Biological Samples Using the PrAMA (Proteolytic Activity Matrix Assay) Method. Methods Mol Biol 2017; 1574:243-253. [PMID: 28315256 DOI: 10.1007/978-1-4939-6850-3_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Proteolytic Activity Matrix Analysis (PrAMA) is a method for simultaneously determining the activities of specific Matrix Metalloproteinases (MMPs) and A Disintegrin and Metalloproteinases (ADAMs) in complex biological samples. In mixtures of unknown proteases, PrAMA infers selective metalloproteinase activities by using a panel of moderately specific FRET-based polypeptide protease substrates in parallel, typically monitored by a plate-reader in a 96-well format. Fluorescence measurements are then quantitatively compared to a standard table of catalytic efficiencies measured from purified mixtures of individual metalloproteinases and FRET substrates. Computational inference of specific activities is performed with an easily used Matlab program, which is provided herein. Thus, we describe PrAMA as a combined experimental and mathematical approach to determine real-time metalloproteinase activities, which has previously been applied to live-cell cultures, cellular lysates, cell culture supernatants, and body fluids from patients.
Collapse
Affiliation(s)
- Catharina Conrad
- Department of Neurosurgery, Marburg University, 35033, Marburg, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital, Albert-Schweitzer Campus 1, 48149, Münster, Germany
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Jörg W Bartsch
- Department of Neurosurgery, Marburg University, 35033, Marburg, Germany.
| | - Uwe Schlomann
- Department of Neurosurgery, Marburg University, 35033, Marburg, Germany
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
30
|
Nishi K, Sato Y, Ohno M, Hiraoka Y, Saijo S, Sakamoto J, Chen PM, Morita Y, Matsuda S, Iwasaki K, Sugizaki K, Harada N, Mukumoto Y, Kiyonari H, Furuyama K, Kawaguchi Y, Uemoto S, Kita T, Inagaki N, Kimura T, Nishi E. Nardilysin Is Required for Maintaining Pancreatic β-Cell Function. Diabetes 2016; 65:3015-27. [PMID: 27385158 DOI: 10.2337/db16-0178] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/15/2016] [Indexed: 11/13/2022]
Abstract
Type 2 diabetes (T2D) is associated with pancreatic β-cell dysfunction, manifested by reduced glucose-stimulated insulin secretion (GSIS). Several transcription factors enriched in β-cells, such as MafA, control β-cell function by organizing genes involved in GSIS. Here we demonstrate that nardilysin (N-arginine dibasic convertase; Nrd1 and NRDc) critically regulates β-cell function through MafA. Nrd1(-/-) mice showed glucose intolerance and severely decreased GSIS. Islets isolated from Nrd1(-/-) mice exhibited reduced insulin content and impaired GSIS in vitro. Moreover, β-cell-specific NRDc-deficient (Nrd1(delβ)) mice showed a diabetic phenotype with markedly reduced GSIS. MafA was specifically downregulated in islets from Nrd1(delβ) mice, whereas overexpression of NRDc upregulated MafA and insulin expression in INS832/13 cells. Chromatin immunoprecipitation assay revealed that NRDc is associated with Islet-1 in the enhancer region of MafA, where NRDc controls the recruitment of Islet-1 and MafA transcription. Our findings demonstrate that NRDc controls β-cell function via regulation of the Islet-1-MafA pathway.
Collapse
Affiliation(s)
- Kiyoto Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yuichi Sato
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Mikiko Ohno
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yoshinori Hiraoka
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan Division of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, Chuo-ku, Kobe, Japan
| | - Sayaka Saijo
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Jiro Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Po-Min Chen
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yusuke Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Shintaro Matsuda
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Kanako Iwasaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Kazu Sugizaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Norio Harada
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yoshiko Mukumoto
- Genetic Engineering Team, RIKEN Center for Life Science Technologies, Chuo-ku, Kobe, Japan
| | - Hiroshi Kiyonari
- Genetic Engineering Team, RIKEN Center for Life Science Technologies, Chuo-ku, Kobe, Japan Animal Resource Development Unit, RIKEN Center for Life Science Technologies, Chuo-ku, Kobe, Japan
| | - Kenichiro Furuyama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yoshiya Kawaguchi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Shinji Uemoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Toru Kita
- Kobe City Medical Center General Hospital, Chuo-ku, Kobe, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Takeshi Kimura
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Eiichiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
31
|
Moss ML, Miller MA, Vujanovic N, Yoneyama T, Rasmussen FH. Fluorescent substrates for ADAM15 useful for assaying and high throughput screening. Anal Biochem 2016; 514:42-47. [PMID: 27637923 DOI: 10.1016/j.ab.2016.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 08/29/2016] [Accepted: 09/12/2016] [Indexed: 11/17/2022]
Abstract
A disintegrin and metalloproteinase 15 (ADAM15), also known as metargidin, plays important roles in regulating inflammation, wound healing, neovascularization, and is an attractive drug target. Fluorescence resonance energy transfer (FRET)-based peptide substrates were tested to identify candidate reagents for high throughput screening and detection of ADAM15 in biological samples. ADAM15 exhibits a unique and diverse activity profile compared to other metalloproteinases. Two FRET substrates, Dabcyl-Gly-Pro-Leu-Gly-Met-Arg-Gly-Lys(FAM)-NH2 (PEPDAB011) and Dabcyl-Ala-Pro-Arg-Trp-Ile-Gln-Asp-Lys(FAM)-NH2 (PEPDAB017), which also detect activities of several matrix metalloproteinases (MMPs -2, -9, and -13), were efficiently cleaved by ADAM15 with specificity constants of 5800 M-1 s-1 and 4300 M-1 s-1, respectively. Additionally, ADAM15 efficiently processed Dabcyl-Leu-Arg-Glu-Gln-Gln-Arg-Leu-Lys-Ser-Lys(FAM)-NH2 (PEPDAB022), which is based on a physiological CD23 cleavage site, with a specificity constant (kcat/Km) of 5200 M-1 s-1. PEPDAB022 was used to screen the ability of known metalloproteinase inhibitors including TAPI-2, marimastat, GI-254023, and the Tissue Inhibitor of Metalloproteinases(TIMPs) 1 and 3 to block ADAM15 activity. Even though ADAM15 exhibits similar substrate preferences to other metalloproteinases, many broad spectrum inhibitors failed to block ADAM15 activity at concentrations as high as 50 μM. Thus, a clear need exists to develop potent and selective ADAM15 inhibitors, and the FRET substrates described herein should aid future research efforts towards this aim.
Collapse
Affiliation(s)
- Marcia L Moss
- BioZyme Inc, 1513 Old White Oak Church Rd., Apex, NC 27523, USA.
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nikola Vujanovic
- University of Pittsburgh Cancer Institute, Departments of Pathology, VA Healthcare System, Pittsburgh, PA, USA
| | - Toshie Yoneyama
- University of Pittsburgh Cancer Institute, Departments of Pathology, VA Healthcare System, Pittsburgh, PA, USA
| | | |
Collapse
|
32
|
Moss ML, Minond D, Yoneyama T, Hansen HP, Vujanovic N, Rasmussen FH. An improved fluorescent substrate for assaying soluble and membrane-associated ADAM family member activities. Anal Biochem 2016; 507:13-7. [PMID: 27177841 DOI: 10.1016/j.ab.2016.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 04/21/2016] [Accepted: 05/01/2016] [Indexed: 10/21/2022]
Abstract
A fluorescent resonance energy transfer substrate with improved sensitivity for ADAM17, -10, and -9 (where ADAM represents a disintegrin and metalloproteinase) has been designed. The new substrate, Dabcyl-Pro-Arg-Ala-Ala-Ala-Homophe-Thr-Ser-Pro-Lys(FAM)-NH2, has specificity constants of 6.3 (±0.3) × 10(4) M(-1) s(-1) and 2.4 (±0.3) × 10(3) M(-1) s(-1) for ADAM17 and ADAM10, respectively. The substrate is more sensitive than widely used peptides based on the precursor tumor necrosis factor-alpha (TNF-alpha) cleavage site, PEPDAB010 or Dabcyl-Ser-Pro-Leu-Ala-Gln-Ala-Val-Arg-Ser-Ser-Lys(FAM)-NH2 and Mca-Pro-Leu-Ala-Gln-Ala-Val-Dpa-Arg-Ser-Ser-Arg-NH2. ADAM9 also processes the new peptide more than 18-fold better than the TNF-alpha-based substrates. The new substrate has a unique selectivity profile because it is processed less efficiently by ADAM8 and MMP1, -2, -3, -8, -9, -12, and -14. This substrate provides a unique tool in which to assess ADAM17, -10, and -9 activities.
Collapse
Affiliation(s)
| | - Dmitriy Minond
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Toshie Yoneyama
- Department of Pathology, University of Pittsburgh Cancer Institute, VA Healthcare System, Pittsburgh, PA 15232, USA
| | - Hinrich P Hansen
- Department of Internal Medicine I, University Clinic Cologne, Cologne 50937, Germany
| | - Nikola Vujanovic
- Department of Pathology, University of Pittsburgh Cancer Institute, VA Healthcare System, Pittsburgh, PA 15232, USA
| | | |
Collapse
|
33
|
Dreymueller D, Ludwig A. Considerations on inhibition approaches for proinflammatory functions of ADAM proteases. Platelets 2016; 28:354-361. [PMID: 27460023 DOI: 10.1080/09537104.2016.1203396] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proteases of the disintegrin and metalloproteinase (ADAM) family mediate the proteolytic shedding of various surface molecules including cytokine precursors, adhesion molecules, growth factors, and receptors. Within the vasculature ADAM10 and ADAM17 regulate endothelial permeability, transendothelial leukocyte migration, and the adhesion of leukocytes and platelets. In vivo studies show that both proteases are implicated in several inflammatory pathologies, for example, edema formation, leukocyte infiltration, and thrombosis. However, both proteases also contribute to developmental and regenerative processes. Thus, although ADAMs can be regarded as valuable drug targets in many aspects, the danger of severe side effects is clearly visible. To circumvent these side effects, traditional inhibition approaches have to be improved to target ADAMs at the right time in the right place. Moreover, the inhibitors need to be more selective for the target protease and if possible also for the substrate. Antibodies recognizing the active conformation of ADAMs or small molecules blocking exosites of ADAM proteases may represent inhibitors with the desired selectivities.
Collapse
Affiliation(s)
- Daniela Dreymueller
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| | - Andreas Ludwig
- a Institute of Pharmacology and Toxicology , RWTH Aachen University , Aachen , Germany
| |
Collapse
|
34
|
Matus CE, Ehrenfeld P, Pavicic F, González CB, Concha M, Bhoola KD, Burgos RA, Figueroa CD. Activation of the human keratinocyte B1 bradykinin receptor induces expression and secretion of metalloproteases 2 and 9 by transactivation of epidermal growth factor receptor. Exp Dermatol 2016; 25:694-700. [PMID: 27093919 DOI: 10.1111/exd.13038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2016] [Indexed: 12/16/2022]
Abstract
The B1 bradykinin receptor (BDKRB1) is a component of the kinin cascade localized in the human skin. Some of the effects produced by stimulation of BDKRB1 depend on transactivation of epidermal growth factor receptor (EGFR), but the mechanisms involved in this process have not been clarified yet. The primary purpose of this study was to determine the effect of a BDKRB1 agonist on wound healing in a mouse model and the migration and secretion of metalloproteases 2 and 9 from human HaCaT keratinocytes and delineate the signalling pathways that triggered their secretion. Although stimulation of BDKRB1 induces weak chemotactic migration of keratinocytes and wound closure in an in vitro scratch-wound assay, the BDKRB1 agonist improved wound closure in a mouse model. BDKRB1 stimulation triggers synthesis and secretion of both metalloproteases, effects that depend on the activity of EGFR and subsequent phosphorylation of ERK1/2 and p38 mitogen-activated protein kinases and PI3K/Akt. In the mouse model, immunoreactivity for both gelatinases was concentrated around wound borders. EGFR transactivation by BDKRB1 agonist involves Src kinases family and ADAM17. In addition to extracellular matrix degradation, metalloproteases 2 and 9 regulate cell migration and differentiation, cell functions that are associated with the role of BDKRB1 in keratinocyte differentiation. Considering that BDKRB1 is up-regulated by inflammation and/or by cytokines that are abundant in the inflammatory milieu, more stable BDKRB1 agonists may be of therapeutic value to modulate wound healing.
Collapse
Affiliation(s)
- Carola E Matus
- Instituto de Morfofisiología y Farmacología, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratorio de Patología Celular, Instituto de Anatomía, Histología & Patología, Universidad Austral de Chile, Valdivia, Chile
| | - Francisca Pavicic
- Laboratorio de Patología Celular, Instituto de Anatomía, Histología & Patología, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos B González
- Instituto de Fisiología, Universidad Austral de Chile, Valdivia, Chile
| | - Miguel Concha
- Laboratorio de Patología Celular, Instituto de Anatomía, Histología & Patología, Universidad Austral de Chile, Valdivia, Chile
| | - Kanti D Bhoola
- Laboratorio de Patología Celular, Instituto de Anatomía, Histología & Patología, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael A Burgos
- Instituto de Morfofisiología y Farmacología, Universidad Austral de Chile, Valdivia, Chile
| | - Carlos D Figueroa
- Laboratorio de Patología Celular, Instituto de Anatomía, Histología & Patología, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
35
|
Chen YC, Cheng YH, Ingram P, Yoon E. Single Cell Proteolytic Assays to Investigate Cancer Clonal Heterogeneity and Cell Dynamics Using an Efficient Cell Loading Scheme. Sci Rep 2016; 6:27154. [PMID: 27283981 PMCID: PMC4901291 DOI: 10.1038/srep27154] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/03/2016] [Indexed: 02/07/2023] Open
Abstract
Proteolytic degradation of the extracellular matrix (ECM) is critical in cancer invasion, and recent work suggests that heterogeneous cancer populations cooperate in this process. Despite the importance of cell heterogeneity, conventional proteolytic assays measure average activity, requiring thousands of cells and providing limited information about heterogeneity and dynamics. Here, we developed a microfluidic platform that provides high-efficiency cell loading and simple valveless isolation, so the proteolytic activity of a small sample (10-100 cells) can be easily characterized. Combined with a single cell derived (clonal) sphere formation platform, we have successfully demonstrated the importance of microenvironmental cues for proteolytic activity and also investigated the difference between clones. Furthermore, the platform allows monitoring single cells at multiple time points, unveiling different cancer cell line dynamics in proteolytic activity. The presented tool facilitates single cell proteolytic analysis using small samples, and our findings illuminate the heterogeneous and dynamic nature of proteolytic activity.
Collapse
Affiliation(s)
- Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122, USA
- University of Michigan Comprehensive Cancer Center, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Yu-Heng Cheng
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122, USA
| | - Patrick Ingram
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd. Ann Arbor, MI 48109-2099, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122, USA
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd. Ann Arbor, MI 48109-2099, USA
| |
Collapse
|
36
|
Lorenzatti Hiles G, Bucheit A, Rubin JR, Hayward A, Cates AL, Day KC, El-Sawy L, Kunju LP, Daignault S, Lee CT, Liebert M, Hussain M, Day ML. ADAM15 Is Functionally Associated with the Metastatic Progression of Human Bladder Cancer. PLoS One 2016; 11:e0150138. [PMID: 26930657 PMCID: PMC4773041 DOI: 10.1371/journal.pone.0150138] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 02/09/2016] [Indexed: 12/21/2022] Open
Abstract
ADAM15 is a member of a family of catalytically active disintegrin membrane metalloproteinases that function as molecular signaling switches, shed membrane bound growth factors and/or cleave and inactivate cell adhesion molecules. Aberrant metalloproteinase function of ADAM15 may contribute to tumor progression through the release of growth factors or disruption of cell adhesion. In this study, we utilized human bladder cancer tissues and cell lines to evaluate the expression and function of ADAM15 in the progression of human bladder cancer. Examination of genome and transcriptome databases revealed that ADAM15 ranked in the top 5% of amplified genes and its mRNA was significantly overexpressed in invasive and metastatic bladder cancer compared to noninvasive disease. Immunostaining of a bladder tumor tissue array designed to evaluate disease progression revealed increased ADAM15 immunoreactivity associated with increasing cancer stage and exhibited significantly stronger staining in metastatic samples. About half of the invasive tumors and the majority of the metastatic cases exhibited high ADAM15 staining index, while all low grade and noninvasive cases exhibited negative or low staining. The knockdown of ADAM15 mRNA expression significantly inhibited bladder tumor cell migration and reduced the invasive capacity of bladder tumor cells through MatrigelTM and monolayers of vascular endothelium. The knockdown of ADAM15 in a human xenograft model of bladder cancer inhibited tumor growth by 45% compared to controls. Structural modeling of the catalytic domain led to the design of a novel ADAM15-specific sulfonamide inhibitor that demonstrated bioactivity and significantly reduced the viability of bladder cancer cells in vitro and in human bladder cancer xenografts. Taken together, the results revealed an undescribed role of ADAM15 in the invasion of human bladder cancer and suggested that the ADAM15 catalytic domain may represent a viable therapeutic target in patients with advanced disease.
Collapse
Affiliation(s)
- Guadalupe Lorenzatti Hiles
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Amanda Bucheit
- Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John R. Rubin
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Alexandra Hayward
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
- School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Angelica L. Cates
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kathleen C. Day
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Layla El-Sawy
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
- European Egyptian Pharmaceuticals, Alexandria, Egypt
| | - L. Priya Kunju
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Stephanie Daignault
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Cheryl T. Lee
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Monica Liebert
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Maha Hussain
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mark L. Day
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
37
|
Facey A, Pinar I, Arthur JF, Qiao J, Jing J, Mado B, Carberry J, Andrews RK, Gardiner EE. A-Disintegrin-And-Metalloproteinase (ADAM) 10 Activity on Resting and Activated Platelets. Biochemistry 2016; 55:1187-94. [DOI: 10.1021/acs.biochem.5b01102] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Adam Facey
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Isaac Pinar
- Department
of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia 3168
| | - Jane F. Arthur
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Jianlin Qiao
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Jing Jing
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Belden Mado
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Josie Carberry
- Department
of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia 3168
| | - Robert K. Andrews
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| | - Elizabeth E. Gardiner
- Australian
Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia 3004
| |
Collapse
|
38
|
Post S, Rozeveld D, Jonker MR, Bischoff R, van Oosterhout AJ, Heijink IH. ADAM10 mediates the house dust mite-induced release of chemokine ligand CCL20 by airway epithelium. Allergy 2015; 70:1545-52. [PMID: 26296735 DOI: 10.1111/all.12730] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND House dust mite (HDM) acts on the airway epithelium to induce airway inflammation in asthma. We previously showed that the ability of HDM to induce allergic sensitization in mice is related to airway epithelial CCL20 secretion. OBJECTIVE As a disintegrin and metalloprotease (ADAM)s have been implicated in chemokine shedding, we sought to determine their involvement in HDM-induced release of chemokines, including CCL20, by airway epithelial cells. METHODS We studied the effects of pharmacological ADAM inhibitors as well as ADAM10 and ADAM17 siRNA downregulation on chemokine release using (multiplex) ELISA in supernatants from HDM-exposed human bronchial epithelial 16HBE cells and primary normal human bronchial epithelial cells (NHBE) at 4-24 h. RESULTS House dust) mite markedly increased CCL20 levels in both 16HBE and NHBE cells (16-24 h). In 16HBE cells, the HDM-induced increase was observed as early as 4 h upon exposure and the use of specific inhibitors indicated the involvement of ADAM10/17-mediated shedding. siRNA knockdown of ADAM10, but not of ADAM17, significantly reduced the HDM-induced release of CCL20 in both 16HBE and NHBE cells. A similar effect was observed for HDM-induced CCL2, CCL5, and CXCL8 release in NHBE cells. The HDM-induced increase in CCL20 levels was not affected by protein synthesis inhibitor cycloheximide nor protein transport inhibitor monensin, indicating that HDM induces surface shedding of chemokines. CONCLUSION Our data show for the first time that ADAM10 activity contributes to HDM-induced shedding of chemokines, including CCL20. The ADAM10/CCL20 axis may be a target for novel therapeutic strategies in asthma.
Collapse
Affiliation(s)
- S. Post
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
- GRIAC Research Institute; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - D. Rozeveld
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
- GRIAC Research Institute; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - M. R Jonker
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
- GRIAC Research Institute; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - R. Bischoff
- Department of Pharmacy, Analytical Biochemistry; University of Groningen; Groningen The Netherlands
| | - A. J. van Oosterhout
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
- GRIAC Research Institute; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| | - I. H. Heijink
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
- GRIAC Research Institute; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
- Department of Pulmonology; University of Groningen; University Medical Center Groningen; Groningen The Netherlands
| |
Collapse
|
39
|
Arenaccio C, Anticoli S, Manfredi F, Chiozzini C, Olivetta E, Federico M. Latent HIV-1 is activated by exosomes from cells infected with either replication-competent or defective HIV-1. Retrovirology 2015; 12:87. [PMID: 26502902 PMCID: PMC4623921 DOI: 10.1186/s12977-015-0216-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 10/17/2015] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Completion of HIV life cycle in CD4(+) T lymphocytes needs cell activation. We recently reported that treatment of resting CD4(+) T lymphocytes with exosomes produced by HIV-1 infected cells induces cell activation and susceptibility to HIV replication. Here, we present data regarding the effects of these exosomes on cells latently infected with HIV-1. RESULTS HIV-1 latently infecting U937-derived U1 cells was activated upon challenge with exosomes purified from the supernatant of U937 cells chronically infected with HIV-1. This effect was no more detectable when exosomes from cells infected with HIV-1 strains either nef-deleted or expressing a functionally defective Nef were used, indicating that Nef is the viral determinant of exosome-induced HIV-1 activation. Treatment with either TAPI-2, i.e., a specific inhibitor of the pro-TNFα-processing ADAM17 enzyme, or anti-TNFα Abs abolished HIV-1 activation. Hence, similar to what previously demonstrated for the exosome-mediated activation of uninfected CD4(+) T lymphocytes, the Nef-ADAM17-TNFα axis is part of the mechanism of latent HIV-1 activation. It is noteworthy that these observations have been reproduced using: (1) primary CD4(+) T lymphocytes latently infected with HIV-1; (2) exosomes from both primary CD4(+) T lymphocytes and macrophages acutely infected with HIV-1; (3) co-cultures of HIV-1 acutely infected CD4(+) T lymphocytes and autologous lymphocytes latently infected with HIV-1, and (4) exosomes from cells expressing a defective HIV-1. CONCLUSIONS Our results strongly suggest that latent HIV-1 can be activated by TNFα released by cells upon ingestion of exosomes released by infected cells, and that this effect depends on the activity of exosome-associated ADAM17. These pieces of evidence shed new light on the mechanism of HIV reactivation in latent reservoirs, and might also be relevant to design new therapeutic interventions focused on HIV eradication.
Collapse
Affiliation(s)
- Claudia Arenaccio
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy. .,Department of Sciences, University Roma Tre, Rome, Italy.
| | - Simona Anticoli
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Francesco Manfredi
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Chiara Chiozzini
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Eleonora Olivetta
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Maurizio Federico
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| |
Collapse
|
40
|
Targeting autocrine HB-EGF signaling with specific ADAM12 inhibition using recombinant ADAM12 prodomain. Sci Rep 2015; 5:15150. [PMID: 26477568 PMCID: PMC4609913 DOI: 10.1038/srep15150] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 09/07/2015] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of ErbB-family signaling underlies numerous pathologies and has been therapeutically targeted through inhibiting ErbB-receptors themselves or their cognate ligands. For the latter, “decoy” antibodies have been developed to sequester ligands including heparin-binding epidermal growth factor (HB-EGF); however, demonstrating sufficient efficacy has been difficult. Here, we hypothesized that this strategy depends on properties such as ligand-receptor binding affinity, which varies widely across the known ErbB-family ligands. Guided by computational modeling, we found that high-affinity ligands such as HB-EGF are more difficult to target with decoy antibodies compared to low-affinity ligands such as amphiregulin (AREG). To address this issue, we developed an alternative method for inhibiting HB-EGF activity by targeting its cleavage from the cell surface. In a model of the invasive disease endometriosis, we identified A Disintegrin and Metalloproteinase 12 (ADAM12) as a protease implicated in HB-EGF shedding. We designed a specific inhibitor of ADAM12 based on its recombinant prodomain (PA12), which selectively inhibits ADAM12 but not ADAM10 or ADAM17. In endometriotic cells, PA12 significantly reduced HB-EGF shedding and resultant cellular migration. Overall, specific inhibition of ligand shedding represents a possible alternative to decoy antibodies, especially for ligands such as HB-EGF that exhibit high binding affinity and localized signaling.
Collapse
|
41
|
Moss ML, Koller G, Bartsch JW, Rakow S, Schlomann U, Rasmussen FH. A colorimetric-based amplification system for proteinases including MMP2 and ADAM8. Anal Biochem 2015; 484:75-81. [DOI: 10.1016/j.ab.2015.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/01/2015] [Accepted: 05/18/2015] [Indexed: 11/25/2022]
|
42
|
Li R, Uttarwar L, Gao B, Charbonneau M, Shi Y, Chan JSD, Dubois CM, Krepinsky JC. High Glucose Up-regulates ADAM17 through HIF-1α in Mesangial Cells. J Biol Chem 2015; 290:21603-14. [PMID: 26175156 DOI: 10.1074/jbc.m115.651604] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Indexed: 12/26/2022] Open
Abstract
We previously showed that ADAM17 mediates high glucose-induced matrix production by kidney mesangial cells. ADAM17 expression is increased in diabetic kidneys, suggesting that its up-regulation may augment high glucose profibrotic responses. We thus studied the effects of high glucose on ADAM17 gene regulation. Primary rat mesangial cells were treated with high glucose (30 mm) or mannitol as osmotic control. High glucose dose-dependently increased ADAM17 promoter activity, transcript, and protein levels. This correlated with augmented ADAM17 activity after 24 h versus 1 h of high glucose. We tested involvement of transcription factors shown in other settings to regulate ADAM17 transcription. Promoter activation was not affected by NF-κB or Sp1 inhibitors, but was blocked by hypoxia-inducible factor-1α (HIF-1α) inhibition or down-regulation. This also prevented ADAM17 transcript and protein increases. HIF-1α activation by high glucose was shown by its increased nuclear translocation and activation of the HIF-responsive hypoxia-response element (HRE)-luciferase reporter construct. Assessment of ADAM17 promoter deletion constructs coupled with mutation analysis and ChIP studies identified HIF-1α binding to its consensus element at -607 as critical for the high glucose response. Finally, inhibitors of epidermal growth factor receptor (EGFR) and downstream PI3K/Akt, or ADAM17 itself, prevented high glucose-induced HIF-1α activation and ADAM17 up-regulation. Thus, high glucose induces ADAM17 transcriptional up-regulation in mesangial cells, which is associated with augmentation of its activity. This is mediated by HIF-1α and requires EGFR/ADAM17 signaling, demonstrating the potentiation by ADAM17 of its own up-regulation. ADAM17 inhibition thus provides a potential novel therapeutic strategy for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Renzhong Li
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6
| | - Lalita Uttarwar
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6
| | - Bo Gao
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6
| | - Martine Charbonneau
- the Division of Immunology, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, and
| | - Yixuan Shi
- the Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - John S D Chan
- the Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Claire M Dubois
- the Division of Immunology, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, and
| | - Joan C Krepinsky
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6,
| |
Collapse
|
43
|
van Duijnhoven SMJ, Robillard MS, Langereis S, Grüll H. Bioresponsive probes for molecular imaging: concepts and in vivo applications. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:282-308. [PMID: 25873263 DOI: 10.1002/cmmi.1636] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 01/24/2015] [Accepted: 02/03/2015] [Indexed: 12/30/2022]
Abstract
Molecular imaging is a powerful tool to visualize and characterize biological processes at the cellular and molecular level in vivo. In most molecular imaging approaches, probes are used to bind to disease-specific biomarkers highlighting disease target sites. In recent years, a new subset of molecular imaging probes, known as bioresponsive molecular probes, has been developed. These probes generally benefit from signal enhancement at the site of interaction with its target. There are mainly two classes of bioresponsive imaging probes. The first class consists of probes that show direct activation of the imaging label (from "off" to "on" state) and have been applied in optical imaging and magnetic resonance imaging (MRI). The other class consists of probes that show specific retention of the imaging label at the site of target interaction and these probes have found application in all different imaging modalities, including photoacoustic imaging and nuclear imaging. In this review, we present a comprehensive overview of bioresponsive imaging probes in order to discuss the various molecular imaging strategies. The focus of the present article is the rationale behind the design of bioresponsive molecular imaging probes and their potential in vivo application for the detection of endogenous molecular targets in pathologies such as cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Sander M J van Duijnhoven
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Minimally Invasive Healthcare, Philips Research, Eindhoven, The Netherlands
| | - Marc S Robillard
- Department of Minimally Invasive Healthcare, Philips Research, Eindhoven, The Netherlands
| | - Sander Langereis
- Department of Minimally Invasive Healthcare, Philips Research, Eindhoven, The Netherlands
| | - Holger Grüll
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Minimally Invasive Healthcare, Philips Research, Eindhoven, The Netherlands
| |
Collapse
|
44
|
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a grim prognosis with less than 5% survivors after 5 years. High expression levels of ADAM8, a metalloprotease-disintegrin, are correlated with poor clinical outcome. We show that ADAM8 expression is associated with increased migration and invasiveness of PDAC cells caused by activation of ERK 1/2 and higher MMP activities. For biological function, ADAM8 requires multimerisation and associates with β1-integrin on the cell surface. A peptidomimetic ADAM8 inhibitor, BK-1361, designed by structural modelling of the disintegrin domain, prevents ADAM8 multimerisation. In PDAC cells, BK-1361 affects ADAM8 function leading to reduced invasiveness, and less ERK 1/2 and MMP activation. BK-1361 application in mice decreased tumour burden and metastasis of implanted pancreatic tumour cells and provides improved metrics of clinical symptoms and survival in a KrasG12D-driven mouse model of PDAC. Thus, our data integrate ADAM8 in pancreatic cancer signalling and validate ADAM8 as a target for PDAC therapy.
Collapse
|
45
|
Exosomes from human immunodeficiency virus type 1 (HIV-1)-infected cells license quiescent CD4+ T lymphocytes to replicate HIV-1 through a Nef- and ADAM17-dependent mechanism. J Virol 2014; 88:11529-39. [PMID: 25056899 DOI: 10.1128/jvi.01712-14] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Resting CD4+ T lymphocytes resist human immunodeficiency virus (HIV) infection. Here, we provide evidence that exosomes from HIV-1-infected cells render resting human primary CD4+ T lymphocytes permissive to HIV-1 replication. These results were obtained with transwell cocultures of HIV-1-infected cells with quiescent CD4+ T lymphocytes in the presence of inhibitors of exosome release and were confirmed using exosomes purified from supernatants of HIV-1-infected primary CD4+ T lymphocytes. We found that the expression of HIV-1 Nef in exosome-producing cells is both necessary and sufficient for cell activation as well as HIV-1 replication in target CD4+ T lymphocytes. We also identified a Nef domain important for the effects we observed, i.e., the 62EEEE65 acidic cluster domain. In addition, we observed that ADAM17, i.e., a disintegrin and metalloprotease converting pro-tumor necrosis factor alpha (TNF-α) in its mature form, associates with exosomes from HIV-1-infected cells, and plays a key role in the HIV-1 replication in quiescent CD4+ T lymphocytes. Treatment with an inhibitor of ADAM17 abolished both activation and HIV-1 replication in resting CD4+ T lymphocytes. TNF-α is the downstream effector of ADAM17 since the treatment of resting lymphocytes with anti-TNF-α antibodies blocked the HIV-1 replication. The data presented here are consistent with a model where Nef induces intercellular communication through exosomes to activate bystander quiescent CD4+ T lymphocytes, thus stimulating viral spread. IMPORTANCE Overall, our findings support the idea that HIV evolved to usurp the exosome-based intercellular communication network to favor its spread in infected hosts.
Collapse
|
46
|
Arenaccio C, Chiozzini C, Columba-Cabezas S, Manfredi F, Federico M. Cell activation and HIV-1 replication in unstimulated CD4+ T lymphocytes ingesting exosomes from cells expressing defective HIV-1. Retrovirology 2014; 11:46. [PMID: 24924541 PMCID: PMC4229896 DOI: 10.1186/1742-4690-11-46] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 05/14/2014] [Indexed: 01/01/2023] Open
Abstract
Background A relevant burden of defective HIV-1 genomes populates PBMCs from HIV-1 infected patients, especially during HAART treatment. These viral genomes, although unable to codify for infectious viral particles, can express viral proteins which may affect functions of host cells as well as bystander ones. Cells expressing defective HIV-1 have a lifespan longer than that of cells producing infectious particles. Hence, their interaction with other cell types, including resting lymphocytes, is expected to occur frequently in tissues where HIV actively replicates. We investigated the effects of the expression of a prototype of functionally defective HIV-1 on bystander, unstimulated CD4+ T lymphocytes. Results We observed that unstimulated human primary CD4+ T lymphocytes were activated and became permissive for HIV-1 replication when co-cultivated with cells expressing a functionally defective HIV-1 (F12/Hut-78 cells). This effect depended on the presence in F12/Hut-78 supernatants of nanovesicles we identified as exosomes. By inspecting the underlying mechanism, we found that ADAM17, i.e., a disintegrin and metalloprotease converting pro-TNF-α in its mature form, associated with exosomes from F12/Hut-78 cells, and played a key role in the HIV-1 replication in unstimulated CD4+ T lymphocytes. In fact, the treatment with an inhibitor of ADAM17 abolished both activation and HIV-1 replication in unstimulated CD4+ T lymphocytes. TNF-α appeared to be the downstream effector of ADAM17 since the treatment of unstimulated lymphocytes with antibodies against TNF-α or its receptors blocked the HIV-1 replication. Finally, we found that the expression of NefF12 in exosome-producing cells was sufficient to induce the susceptibility to HIV-1 infection in unstimulated CD4+ T lymphocytes. Conclusions Exosomes from cells expressing a functionally defective mutant can induce cell activation and HIV-1 susceptibility in unstimulated CD4+ T lymphocytes. This evidence highlights the relevance for AIDS pathogenesis of the expression of viral products from defective HIV-1 genomes.
Collapse
Affiliation(s)
| | | | | | | | - Maurizio Federico
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy.
| |
Collapse
|
47
|
Gandhi R, Yi J, Ha J, Shi H, Ismail O, Nathoo S, Bonventre JV, Zhang X, Gunaratnam L. Accelerated receptor shedding inhibits kidney injury molecule-1 (KIM-1)-mediated efferocytosis. Am J Physiol Renal Physiol 2014; 307:F205-21. [PMID: 24829508 DOI: 10.1152/ajprenal.00638.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Efficient clearance of apoptotic cells (efferocytosis) prevents inflammation and permits repair following tissue injury. Kidney injury molecule-1 (KIM-1) is a receptor for phosphatidylserine, an "eat-me" signal exposed on the surface of apoptotic cells that marks them for phagocytic clearance. KIM-1 is upregulated on proximal tubule epithelial cells (PTECs) during ischemic acute kidney injury (AKI), enabling efferocytosis by surviving PTECs. KIM-1 is spontaneously cleaved at its ectodomain region to generate a soluble fragment that serves a sensitive and specific biomarker for AKI, but the biological relevance of KIM-1 shedding is unknown. Here, we sought to determine how KIM-1 shedding might regulate efferocytosis. Using cells that endogenously and exogenously express KIM-1, we found that hydrogen peroxide-mediated oxidative injury or PMA treatment accelerated KIM-1 shedding in a dose-dependent manner. KIM-1 shedding was also accelerated when apoptotic cells were added. Accelerated shedding or the presence of excess soluble KIM-1 in the extracellular milieu significantly inhibited efferocytosis. We also identified that TNF-α-converting enzyme (TACE or ADAM17) mediates both the spontaneous and PMA-accelerated shedding of KIM-1. While accelerated shedding inhibited efferocytosis, we found that spontaneous KIM-1 cleavage does not affect the phagocytic efficiency of PTECs. Our results suggest that KIM-1 shedding is accelerated by worsening cellular injury, and excess soluble KIM-1 competitively inhibits efferocytosis. These findings may be important in AKI when there is severe cellular injury.
Collapse
Affiliation(s)
- Rushi Gandhi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - James Yi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Jihyen Ha
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Hang Shi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Centre for Human Immunology, Western University, London, Ontario, Canada; and
| | - Ola Ismail
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Sahra Nathoo
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xizhong Zhang
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Lakshman Gunaratnam
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada; Centre for Human Immunology, Western University, London, Ontario, Canada; and
| |
Collapse
|
48
|
Altshuler AE, Lamadrid I, Li D, Ma SR, Kurre L, Schmid-Schönbein GW, Penn AH. Transmural intestinal wall permeability in severe ischemia after enteral protease inhibition. PLoS One 2014; 9:e96655. [PMID: 24805256 PMCID: PMC4013012 DOI: 10.1371/journal.pone.0096655] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 04/10/2014] [Indexed: 01/13/2023] Open
Abstract
In intestinal ischemia, inflammatory mediators in the small intestine's lumen such as food byproducts, bacteria, and digestive enzymes leak into the peritoneal space, lymph, and circulation, but the mechanisms by which the intestinal wall permeability initially increases are not well defined. We hypothesize that wall protease activity (independent of luminal proteases) and apoptosis contribute to the increased transmural permeability of the intestine's wall in an acutely ischemic small intestine. To model intestinal ischemia, the proximal jejunum to the distal ileum in the rat was excised, the lumen was rapidly flushed with saline to remove luminal contents, sectioned into equal length segments, and filled with a tracer (fluorescein) in saline, glucose, or protease inhibitors. The transmural fluorescein transport was determined over 2 hours. Villi structure and epithelial junctional proteins were analyzed. After ischemia, there was increased transmural permeability, loss of villi structure, and destruction of epithelial proteins. Supplementation with luminal glucose preserved the epithelium and significantly attenuated permeability and villi damage. Matrix metalloproteinase (MMP) inhibitors (doxycycline, GM 6001), and serine protease inhibitor (tranexamic acid) in the lumen, significantly reduced the fluorescein transport compared to saline for 90 min of ischemia. Based on these results, we tested in an in-vivo model of hemorrhagic shock (90 min 30 mmHg, 3 hours observation) for intestinal lesion formation. Single enteral interventions (saline, glucose, tranexamic acid) did not prevent intestinal lesions, while the combination of enteral glucose and tranexamic acid prevented lesion formation after hemorrhagic shock. The results suggest that apoptotic and protease mediated breakdown cause increased permeability and damage to the intestinal wall. Metabolic support in the lumen of an ischemic intestine with glucose reduces the transport from the lumen across the wall and enteral proteolytic inhibition attenuates tissue breakdown. These combined interventions ameliorate lesion formation in the small intestine after hemorrhagic shock.
Collapse
Affiliation(s)
- Angelina E. Altshuler
- Department of Bioengineering, The Institute of Engineering in Medicine, University Of California San Diego, La Jolla, California, United States of America
- * E-mail:
| | - Itze Lamadrid
- Department of Bioengineering, The Institute of Engineering in Medicine, University Of California San Diego, La Jolla, California, United States of America
| | - Diana Li
- Department of Bioengineering, The Institute of Engineering in Medicine, University Of California San Diego, La Jolla, California, United States of America
| | - Stephanie R. Ma
- Department of Bioengineering, The Institute of Engineering in Medicine, University Of California San Diego, La Jolla, California, United States of America
| | - Leena Kurre
- Department of Bioengineering, The Institute of Engineering in Medicine, University Of California San Diego, La Jolla, California, United States of America
| | - Geert W. Schmid-Schönbein
- Department of Bioengineering, The Institute of Engineering in Medicine, University Of California San Diego, La Jolla, California, United States of America
| | - Alexander H. Penn
- Department of Bioengineering, The Institute of Engineering in Medicine, University Of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
49
|
Kotzsch A, Skovgaard T, Buus U, Andersen S, Devkota K, Berthelsen J. A substrate-optimized electrophoretic mobility shift assay for ADAM12. Anal Biochem 2014; 452:34-42. [PMID: 24534253 DOI: 10.1016/j.ab.2014.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/31/2014] [Accepted: 02/02/2014] [Indexed: 11/17/2022]
Abstract
ADAM12 belongs to the A disintegrin and metalloprotease (ADAM) family of secreted sheddases activating extracellular growth factors such as epidermal growth factor receptor (EGFR) ligands and tumor necrosis factor-alpha (TNF-α). ADAM proteases, most notably ADAM17 (TNF-α-converting enzyme), have long been investigated as pharmaceutical drug targets; however, due to lack of potency and in vivo side effects, none of the small-molecule inhibitors discovered so far has made it beyond clinical testing. Ongoing research on novel selective inhibitors of ADAMs requires reliable biochemical assays to validate molecular probes from large-scale screening efforts. Here we describe an electrophoretic mobility shift assay for ADAM12 based on the identification of an optimized peptide substrate that is characterized by excellent performance and reproducibility.
Collapse
Affiliation(s)
- Alexander Kotzsch
- Facility for Protein Purification and Function at the Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark.
| | - Tine Skovgaard
- Facility for Protein Purification and Function at the Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Uwe Buus
- Facility for Protein Purification and Function at the Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Simon Andersen
- Facility for Protein Purification and Function at the Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Kanchan Devkota
- Facility for Protein Purification and Function at the Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Jens Berthelsen
- Facility for Protein Purification and Function at the Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
50
|
Scaling AL, Prossnitz ER, Hathaway HJ. GPER mediates estrogen-induced signaling and proliferation in human breast epithelial cells and normal and malignant breast. Discov Oncol 2014; 5:146-160. [PMID: 24718936 DOI: 10.1007/s12672-014-0174-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/14/2014] [Indexed: 02/06/2023] Open
Abstract
17β-Estradiol (estrogen), through receptor binding and activation, is required for mammary gland development. Estrogen stimulates epithelial proliferation in the mammary gland, promoting ductal elongation and morphogenesis. In addition to a developmental role, estrogen promotes proliferation in tumorigenic settings, particularly breast cancer. The proliferative effects of estrogen in the normal breast and breast tumors are attributed to estrogen receptor α. Although in vitro studies have demonstrated that the G protein-coupled estrogen receptor (GPER, previously called GPR30) can modulate proliferation in breast cancer cells both positively and negatively depending on cellular context, its role in proliferation in the intact normal or malignant breast remains unclear. Estrogen-induced GPER-dependent proliferation was assessed in the immortalized nontumorigenic human breast epithelial cell line, MCF10A, and an ex vivo organ culture model employing human breast tissue from reduction mammoplasty or tumor resections. Stimulation by estrogen and the GPER-selective agonist G-1 increased the mitotic index in MCF10A cells and proportion of cells in the cell cycle in human breast and breast cancer explants, suggesting increased proliferation. Inhibition of candidate signaling pathways that may link GPER activation to proliferation revealed a dependence on Src, epidermal growth factor receptor transactivation by heparin-bound EGF and subsequent ERK phosphorylation. Proliferation was not dependent on matrix metalloproteinase cleavage of membrane-bound pro-HB-EGF. The contribution of GPER to estrogen-induced proliferation in MCF10A cells and breast tissue was confirmed by the ability of GPER-selective antagonist G36 to abrogate estrogen- and G-1-induced proliferation, and the ability of siRNA knockdown of GPER to reduce estrogen- and G-1-induced proliferation in MCF10A cells. This is the first study to demonstrate GPER-dependent proliferation in primary normal and malignant human tissue, revealing a role for GPER in estrogen-induced breast physiology and pathology.
Collapse
Affiliation(s)
- Allison L Scaling
- Department of Cell Biology & Physiology, and Cancer Research and Treatment Center, University of New Mexico School of Medicine, Albuquerque, NM
| | - Eric R Prossnitz
- Department of Cell Biology & Physiology, and Cancer Research and Treatment Center, University of New Mexico School of Medicine, Albuquerque, NM
| | - Helen J Hathaway
- Department of Cell Biology & Physiology, and Cancer Research and Treatment Center, University of New Mexico School of Medicine, Albuquerque, NM
| |
Collapse
|